1
|
Mabry S, Bradshaw JL, Gardner JJ, Wilson EN, Sunuwar J, Yeung H, Shrestha S, Cunningham JT, Cunningham RL. The impact of chronic intermittent hypoxia on enzymatic activity in memory-associated brain regions of male and female rats. Biol Sex Differ 2025; 16:5. [PMID: 39891225 PMCID: PMC11786371 DOI: 10.1186/s13293-025-00688-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 01/24/2025] [Indexed: 02/03/2025] Open
Abstract
BACKGROUND Obstructive sleep apnea (OSA) is an intermittent hypoxia disorder associated with cognitive dysfunction, including learning and memory impairments. There is evidence that alterations in protease activity and neuronal activation are associated with cognitive dysfunction, are dependent on sex, and may be brain region-specific. However, the mechanisms mediating OSA-induced cognitive impairments are unclear. Therefore, we used a rat model of OSA, chronic intermittent hypoxia (CIH) to investigate protease activity (e.g., calpain and caspase-3) on spectrin, a cytoskeletal protein associated with neurotransmitter release, and neuronal activation (early growth response protein 1, EGR-1) in brain regions associated with learning and memory. METHODS Male and female Sprague Dawley rats were exposed to CIH or room air (normoxic) for 14 days. We quantified protease activity and cleaved spectrin products, along with EGR-1 protein expression in hippocampal subregions (CA1, CA3), cortical regions [entorhinal cortex (ETC), retrosplenial cortex (RSC), cerebellar cortex (CC)], and subcortical regions [raphe nucleus (RN), locus coeruleus (LC)] associated with learning and memory. Within each group, Pearson correlations of calpain activity, caspase-3 activity, and EGR-1 expression were performed between brain regions. Sex differences within normoxic and CIH correlations were examined. RESULTS CIH dysregulated calpain activity in male ETC, and female CA1 and RSC. CIH dysregulated caspase-3 activity in male RN, and female CA1 and RSC. CIH decreased calpain and caspase-3 cleavage products in male ETC. CIH decreased calpain-cleaved spectrin in male RSC but increased these products in female RSC. EGR-1 expression was decreased in male and female RN. Correlational analysis revealed CIH increased excitatory connections in males and increased inhibitory connections in females. EGR-1 expression in males shifted from negative to positive correlations. CONCLUSIONS Overall, these data indicate CIH dysregulates protease activity and impairs neuronal function in a brain region- and sex-dependent manner. This indicates that males and females exhibit sex-specific vulnerabilities to mild OSA. These findings concur with our previous behavioral studies that demonstrated memory impairment in CIH-exposed rats.
Collapse
Affiliation(s)
- Steve Mabry
- Department of Pharmaceutical Sciences, System College of Pharmacy, Fort Worth, TX, USA
- North Texas Eye Research Institute, Fort Worth, TX, USA
| | - Jessica L Bradshaw
- Department of Pharmaceutical Sciences, System College of Pharmacy, Fort Worth, TX, USA
| | - Jennifer J Gardner
- Department of Pharmaceutical Sciences, System College of Pharmacy, Fort Worth, TX, USA
| | - E Nicole Wilson
- Department of Pharmaceutical Sciences, System College of Pharmacy, Fort Worth, TX, USA
| | - Janak Sunuwar
- Research Core, Division of Research and Innovation, Fort Worth, TX, USA
| | - Hannah Yeung
- Department of Pharmaceutical Sciences, System College of Pharmacy, Fort Worth, TX, USA
- Texas College of Osteopathic Medicine, Fort Worth, TX, USA
| | - Sharad Shrestha
- Research Core, Division of Research and Innovation, Fort Worth, TX, USA
| | - J Thomas Cunningham
- Department of Physiology and Anatomy, College of Biomedical and Translational Sciences, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Rebecca L Cunningham
- Department of Pharmaceutical Sciences, System College of Pharmacy, Fort Worth, TX, USA.
| |
Collapse
|
2
|
Mabry S, Bradshaw JL, Gardner JJ, Wilson EN, Sunuwar J, Yeung H, Shrestha S, Cunningham JT, Cunningham RL. The impact of chronic intermittent hypoxia on enzymatic activity in memory-associated brain regions of male and female rats. RESEARCH SQUARE 2024:rs.3.rs-5449794. [PMID: 39711575 PMCID: PMC11661378 DOI: 10.21203/rs.3.rs-5449794/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Background Obstructive sleep apnea (OSA) is an intermittent hypoxia disorder associated with cognitive dysfunction, including learning and memory impairments. There is evidence that alterations in protease activity and neuronal activation as associated with cognitive dysfunction, are dependent on sex, and may be brain region-specific. However, the mechanisms mediating OSA-induced cognitive impairments are unclear. Therefore, we used a rat model of OSA, chronic intermittent hypoxia (CIH), to investigate protease activity (e.g., calpain and caspase-3) and neuronal activation (early growth response protein 1, EGR-1) in brain regions associated with learning and memory. We used a rat model of OSA known as chronic intermittent hypoxia (CIH) to investigate protease activity (calpain and caspase-3) and neuronal activation (early growth response protein 1, EGR-1) in brain regions associated with learning and memory. Methods Male and female Sprague Dawley rats were exposed to CIH or room air (normoxic) for 14 days. We quantified protease activity and cleaved spectrin products, along with EGR-1 protein expression in hippocampal subregions (CA1, CA3), cortical regions [entorhinal cortex (ETC), retrosplenial cortex (RSC), cerebellar cortex (CC)], and subcortical regions [raphe nucleus (RN), locus coeruleus (LC)] associated with learning and memory. Within each group, Pearson correlations of calpain activity, caspase-3 activity, and EGR-1 expression were performed between brain regions. Sex differences within normoxic and CIH correlations were examined. Results CIH dysregulated calpain activity in male ETC and female CA1 and RSC. CIH dysregulated caspase-3 activity in male RN and female CA1 and RSC. CIH decreased calpain and caspase-3 cleavage products in male ETC. CIH decreased calpain-cleaved spectrin in male RSC but increased these products in female RSC. EGR-1 expression was decreased in male and female RN. Correlational analysis revealed CIH increased excitatory connections in males and increased inhibitory connections in females. EGR-1 expression in males shifted from negative to positive correlations. Conclusions Overall, these data show that CIH dysregulates protease activity and impairs neuronal function in a brain region- and sex-dependent manner. This indicates that males and females exhibit sex-specific vulnerabilities to mild OSA. These findings concur with our previous behavioral studies that demonstrated memory impairment in CIH-exposed rats.
Collapse
Affiliation(s)
- Steve Mabry
- University of North Texas Health Science Center
| | | | | | | | | | | | | | | | | |
Collapse
|
3
|
Jones SM, Sleiman SJ, McCann KE, Jarmusch AK, Alexander GM, Dudek SM. Prenatal exposure to the mineralocorticoid receptor antagonist spironolactone disrupts hippocampal area CA2 connectivity and alters behavior in mice. Neuropsychopharmacology 2024; 50:378-387. [PMID: 39237618 PMCID: PMC11631951 DOI: 10.1038/s41386-024-01971-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 08/07/2024] [Accepted: 08/14/2024] [Indexed: 09/07/2024]
Abstract
In the brain, the hippocampus is enriched with mineralocorticoid receptors (MR; Nr3c2), a ligand-dependent transcription factor stimulated by the stress hormone corticosterone in rodents. Recently, we discovered that MR is required for the acquisition and maintenance of many features of mouse area CA2 neurons. Notably, we observed that immunofluorescence for the vesicular glutamate transporter 2 (vGluT2), likely representing afferents from the supramammillary nucleus (SuM), was disrupted in the embryonic, but not postnatal, MR knockout mouse CA2. To test whether pharmacological perturbation of MR activity in utero similarly disrupts CA2 connectivity, we implanted slow-release pellets containing the MR antagonist spironolactone in mouse dams during mid-gestation. After confirming that at least one likely active metabolite crossed from the dams' serum into the embryonic brains, we found that spironolactone treatment caused a significant reduction of CA2 axon fluorescence intensity in the CA1 stratum oriens, where CA2 axons preferentially project, and that vGluT2 staining was significantly decreased in both CA2 and dentate gyrus in spironolactone-treated animals. We also found that spironolactone-treated animals exhibited increased reactivity to novel objects, an effect similar to what is seen with embryonic or postnatal CA2-targeted MR knockout. However, we found no difference in preference for social novelty between the treatment groups. We infer these results to suggest that persistent or more severe disruptions in MR function may be required to interfere with this type of social behavior. These findings do indicate, though, that developmental disruption in MR signaling can have persistent effects on hippocampal circuitry and behavior.
Collapse
Affiliation(s)
- Stephanie M Jones
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, Division of Intramural Research, National Institute of Health, Research Triangle Park, NC, 27709, USA
| | - Sarah Jo Sleiman
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, Division of Intramural Research, National Institute of Health, Research Triangle Park, NC, 27709, USA
- Neuroscience Curriculum, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Katharine E McCann
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, Division of Intramural Research, National Institute of Health, Research Triangle Park, NC, 27709, USA
- School of Psychology, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Alan K Jarmusch
- Immunity, Inflammation, and Disease Laboratory, Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, 27709, USA
| | - Georgia M Alexander
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, Division of Intramural Research, National Institute of Health, Research Triangle Park, NC, 27709, USA
| | - Serena M Dudek
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, Division of Intramural Research, National Institute of Health, Research Triangle Park, NC, 27709, USA.
| |
Collapse
|
4
|
Cankar N, Beschorner N, Tsopanidou A, Qvist FL, Colaço AR, Andersen M, Kjaerby C, Delle C, Lambert M, Mundt F, Weikop P, Jucker M, Mann M, Skotte NH, Nedergaard M. Sleep deprivation leads to non-adaptive alterations in sleep microarchitecture and amyloid-β accumulation in a murine Alzheimer model. Cell Rep 2024; 43:114977. [PMID: 39541211 DOI: 10.1016/j.celrep.2024.114977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 09/09/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Impaired sleep is a common aspect of aging and often precedes the onset of Alzheimer's disease. Here, we compare the effects of sleep deprivation in young wild-type mice and their APP/PS1 littermates, a murine model of Alzheimer's disease. After 7 h of sleep deprivation, both genotypes exhibit an increase in EEG slow-wave activity. However, only the wild-type mice demonstrate an increase in the power of infraslow norepinephrine oscillations, which are characteristic of healthy non-rapid eye movement sleep. Notably, the APP/PS1 mice fail to enhance norepinephrine oscillations 24 h after sleep deprivation, coinciding with an accumulation of cerebral amyloid-β protein. Proteome analysis of cerebrospinal fluid and extracellular fluid further supports these findings by showing altered protein clearance in APP/PS1 mice. We propose that the suppression of infraslow norepinephrine oscillations following sleep deprivation contributes to increased vulnerability to sleep loss and heightens the risk of developing amyloid pathology in early stages of Alzheimer's disease.
Collapse
Affiliation(s)
- Neža Cankar
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark; Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Natalie Beschorner
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Anastasia Tsopanidou
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Filippa L Qvist
- NNF Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Ana R Colaço
- Proteomics Research Infrastructure, Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Mie Andersen
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Celia Kjaerby
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Christine Delle
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Marius Lambert
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Filip Mundt
- NNF Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Pia Weikop
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Mathias Jucker
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Matthias Mann
- NNF Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark; Department for Proteomics and Signal Transduction, Max-Planck Institute for Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Niels Henning Skotte
- NNF Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark; Center for Translational Neuromedicine, University of Rochester Medical School, Elmwood Avenue 601, Rochester, NY 14642, USA.
| |
Collapse
|
5
|
Ray A, Loghinov I, Ravindranath V, Barth AL. Early hippocampal hyperexcitability and synaptic reorganization in mouse models of amyloidosis. iScience 2024; 27:110629. [PMID: 39262788 PMCID: PMC11388185 DOI: 10.1016/j.isci.2024.110629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 05/09/2024] [Accepted: 07/29/2024] [Indexed: 09/13/2024] Open
Abstract
The limited success of plaque-reducing therapies in Alzheimer's disease suggests that early treatment might be more effective in delaying or reversing memory impairments. Toward this end, it is important to establish the progression of synaptic and circuit changes before onset of plaques or cognitive deficits. Here, we used quantitative, fluorescence-based methods for synapse detection in CA1 pyramidal neurons to investigate the interaction between abnormal circuit activity, measured by Fos-immunoreactivity, and synapse reorganization in mouse models of amyloidosis. Using a genetically encoded, fluorescently labeled synaptic marker in juvenile mice (prior to sexual maturity), we find both synapse gain and loss depending on dendritic location. This progresses to broad synapse loss in aged mice. Elevated hippocampal activity in both CA3 and CA1 was present at weaning and preceded this reorganization. Thus, Aβ overproduction may initiate abnormal activity and subsequent input-specific synapse plasticity. These findings indicate that sustained amyloidosis drives heterogeneous and progressive circuit-wide abnormalities.
Collapse
Affiliation(s)
- Ajit Ray
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Iulia Loghinov
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Vijayalakshmi Ravindranath
- Centre for Neuroscience, Indian Institute of Science, Bengaluru, Karnataka 560012, India
- Centre for Brain Research, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Alison L. Barth
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| |
Collapse
|
6
|
Leung LS, Gill RS, Shen B, Chu L. Cholinergic and behavior-dependent beta and gamma waves are coupled between olfactory bulb and hippocampus. Hippocampus 2024; 34:464-490. [PMID: 38949057 DOI: 10.1002/hipo.23622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 04/16/2024] [Accepted: 06/10/2024] [Indexed: 07/02/2024]
Abstract
Olfactory oscillations may enhance cognitive processing through coupling with beta (β, 15-30 Hz) and gamma (γ, 30-160 Hz) activity in the hippocampus (HPC). We hypothesize that coupling between olfactory bulb (OB) and HPC oscillations is increased by cholinergic activation in control rats and is reduced in kainic-acid-treated epileptic rats, a model of temporal lobe epilepsy. OB γ2 (63-100 Hz) power was higher during walking and immobility-awake (IMM) compared to sleep, while γ1 (30-57 Hz) power was higher during grooming than other behavioral states. Muscarinic cholinergic agonist pilocarpine (25 mg/kg ip) with peripheral muscarinic blockade increased OB power and OB-HPC coherence at β and γ1 frequency bands. A similar effect was found after physostigmine (0.5 mg/kg ip) but not scopolamine (10 mg/kg ip). Pilocarpine increased bicoherence and cross-frequency coherence (CFC) between OB slow waves (SW, 1-5 Hz) and hippocampal β, γ1 and γ2 waves, with stronger coherence at CA1 alveus and CA3c than CA1 stratum radiatum. Bicoherence further revealed a nonlinear interaction of β waves in OB with β waves at the CA1-alveus. Beta and γ1 waves in OB or HPC were segregated at one phase of the OB-SW, opposite to the phase of γ2 and γ3 (100-160 Hz) waves, suggesting independent temporal processing of β/γ1 versus γ2/γ3 waves. At CA1 radiatum, kainic-acid-treated epileptic rats compared to control rats showed decreased theta power, theta-β and theta-γ2 CFC during baseline walking, decreased CFC of HPC SW with γ2 and γ3 waves during baseline IMM, and decreased coupling of OB SW with β and γ2 waves at CA1 alveus after pilocarpine. It is concluded that β and γ waves in the OB and HPC are modulated by a slow respiratory rhythm, in a cholinergic and behavior-dependent manner, and OB-HPC functional connectivity at β and γ frequencies may enhance cognitive functions.
Collapse
Affiliation(s)
- L Stan Leung
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
- Graduate Program in Neuroscience, University of Western Ontario, London, Ontario, Canada
| | - Ravnoor Singh Gill
- Graduate Program in Neuroscience, University of Western Ontario, London, Ontario, Canada
| | - Bixia Shen
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
| | - Liangwei Chu
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
7
|
Stevens NA, Lankisch K, Draguhn A, Engelhardt M, Both M, Thome C. Increased Interhemispheric Connectivity of a Distinct Type of Hippocampal Pyramidal Cells. J Neurosci 2024; 44:e0440232023. [PMID: 38123997 PMCID: PMC10869156 DOI: 10.1523/jneurosci.0440-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 11/02/2023] [Accepted: 11/19/2023] [Indexed: 12/23/2023] Open
Abstract
Neurons typically generate action potentials at their axon initial segment based on the integration of synaptic inputs. In many neurons, the axon extends from the soma, equally weighting dendritic inputs. A notable exception is found in a subset of hippocampal pyramidal cells where the axon emerges from a basal dendrite. This structure allows these axon-carrying dendrites (AcDs) a privileged input route. We found that in male mice, such cells in the CA1 region receive stronger excitatory input from the contralateral CA3, compared with those with somatic axon origins. This is supported by a higher count of putative synapses from contralateral CA3 on the AcD. These findings, combined with prior observations of their distinct role in sharp-wave ripple firing, suggest a key role of this neuron subset in coordinating bi-hemispheric hippocampal activity during memory-centric oscillations.
Collapse
Affiliation(s)
- Nikolas Andreas Stevens
- Institute of Physiology and Pathophysiology, Heidelberg University, 69120 Heidelberg, Germany
| | - Katja Lankisch
- Institute of Physiology and Pathophysiology, Heidelberg University, 69120 Heidelberg, Germany
| | - Andreas Draguhn
- Institute of Physiology and Pathophysiology, Heidelberg University, 69120 Heidelberg, Germany
| | - Maren Engelhardt
- Institute of Anatomy and Cell Biology, Medical Faculty, Johannes Kepler University Linz, 4020 Linz, Austria
| | - Martin Both
- Institute of Physiology and Pathophysiology, Heidelberg University, 69120 Heidelberg, Germany
| | - Christian Thome
- Institute of Physiology and Pathophysiology, Heidelberg University, 69120 Heidelberg, Germany
- Institute of Anatomy and Cell Biology, Medical Faculty, Johannes Kepler University Linz, 4020 Linz, Austria
- Institute of Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California 94305
| |
Collapse
|
8
|
Zhao F, Behnisch T. The Enigmatic CA2: Exploring the Understudied Region of the Hippocampus and Its Involvement in Parkinson's Disease. Biomedicines 2023; 11:1996. [PMID: 37509636 PMCID: PMC10377725 DOI: 10.3390/biomedicines11071996] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/12/2023] [Accepted: 07/13/2023] [Indexed: 07/30/2023] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disease that affects both motor and non-motor functions. Although motor impairment is a prominent clinical sign of PD, additional neurological symptoms may also occur, particularly in the preclinical and prodromal stages. Among these symptoms, social cognitive impairment is common and detrimental. This article aims to review non-motor symptoms in PD patients, focusing on social cognitive deficits. It also examines the specific characteristics of the CA2 region and its involvement in social behavior, highlighting recent advances and perspectives. Additionally, this review provides critical insights into and analysis of research conducted in rodents and humans, which may help improve the understanding of the current status of putative therapeutic strategies for social cognitive dysfunction in PD and potential avenues related to the function of the hippocampal CA2 region.
Collapse
Affiliation(s)
- Fang Zhao
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Thomas Behnisch
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| |
Collapse
|
9
|
Shi HJ, Wang S, Wang XP, Zhang RX, Zhu LJ. Hippocampus: Molecular, Cellular, and Circuit Features in Anxiety. Neurosci Bull 2023; 39:1009-1026. [PMID: 36680709 PMCID: PMC10264315 DOI: 10.1007/s12264-023-01020-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 11/13/2022] [Indexed: 01/22/2023] Open
Abstract
Anxiety disorders are currently a major psychiatric and social problem, the mechanisms of which have been only partially elucidated. The hippocampus serves as a major target of stress mediators and is closely related to anxiety modulation. Yet so far, its complex anatomy has been a challenge for research on the mechanisms of anxiety regulation. Recent advances in imaging, virus tracking, and optogenetics/chemogenetics have permitted elucidation of the activity, connectivity, and function of specific cell types within the hippocampus and its connected brain regions, providing mechanistic insights into the elaborate organization of the hippocampal circuitry underlying anxiety. Studies of hippocampal neurotransmitter systems, including glutamatergic, GABAergic, cholinergic, dopaminergic, and serotonergic systems, have contributed to the interpretation of the underlying neural mechanisms of anxiety. Neuropeptides and neuroinflammatory factors are also involved in anxiety modulation. This review comprehensively summarizes the hippocampal mechanisms associated with anxiety modulation, based on molecular, cellular, and circuit properties, to provide tailored targets for future anxiety treatment.
Collapse
Affiliation(s)
- Hu-Jiang Shi
- Key Laboratory of Developmental Genes and Human Diseases, MOE, Department of Histology and Embryology, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Shuang Wang
- Key Laboratory of Developmental Genes and Human Diseases, MOE, Department of Histology and Embryology, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Xin-Ping Wang
- Key Laboratory of Developmental Genes and Human Diseases, MOE, Department of Histology and Embryology, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Rui-Xin Zhang
- Key Laboratory of Developmental Genes and Human Diseases, MOE, Department of Histology and Embryology, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Li-Juan Zhu
- Key Laboratory of Developmental Genes and Human Diseases, MOE, Department of Histology and Embryology, School of Medicine, Southeast University, Nanjing, 210009, China.
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 201108, China.
| |
Collapse
|
10
|
Dudek SM, Alexander GM, Farris S. Introduction to the special issue on: A new view of hippocampal area CA2. Hippocampus 2023; 33:127-132. [PMID: 36826426 DOI: 10.1002/hipo.23514] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/31/2023] [Indexed: 02/25/2023]
Affiliation(s)
- Serena M Dudek
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institute of Health, Research Triangle Park, North Carolina, USA
| | - Georgia M Alexander
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institute of Health, Research Triangle Park, North Carolina, USA
| | - Shannon Farris
- Center for Neurobiology Research, Fralin Biomedical Research Institute, Virginia Tech Carilion, Roanoke, Virginia, USA
| |
Collapse
|
11
|
Radzicki D, Chong S, Dudek SM. Morphological and molecular markers of mouse area CA2 along the proximodistal and dorsoventral hippocampal axes. Hippocampus 2023; 33:133-149. [PMID: 36762588 PMCID: PMC10443601 DOI: 10.1002/hipo.23509] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 01/20/2023] [Accepted: 01/23/2023] [Indexed: 02/11/2023]
Abstract
Hippocampal area CA2 is a molecularly and functionally distinct region of the hippocampus that has classically been defined as the area with large pyramidal neurons lacking input from the dentate gyrus and the thorny excrescences (TEs) characteristic of CA3 neurons. A modern definition of CA2, however, makes use of the expression of several molecular markers that distinguish it from neighboring CA3 and CA1. Using immunohistochemistry, we sought to characterize the staining patterns of commonly used CA2 markers along the dorsal-ventral hippocampal axis and determine how these markers align along the proximodistal axis. We used a region of CA2 that stained for both Regulator of G-protein Signaling 14 (RGS14) and Purkinje Cell Protein 4 (PCP4; "double-labeled zone" [DLZ]) as a reference. Here, we report that certain commonly used CA2 molecular markers may be better suited for drawing distinct boundaries between CA2/3 and CA2/1. For example, RGS14+ and STEP+ neurons showed minimal to no extension into area CA1 while areas stained with VGluT2 and Wisteria Floribunda agglutinin were consistently smaller than the DLZ/CA2 borders by ~100 μ on the CA1 or CA3 sides respectively. In addition, these patterns are dependent on position along the dorsal-ventral hippocampal axis such that PCP4 labeling often extended beyond the distal border of the DLZ into CA1. Finally, we found that, consistent with previous findings, mossy fibers innervate a subset of RGS14 positive neurons (~65%-70%) and that mossy fiber bouton number and relative size in CA2 are less than that of boutons in CA3. Unexpectedly, we did find evidence of some complex spines on apical dendrites in CA2, though much fewer in number than in CA3. Our results indicate that certain molecular markers may be better suited than others when defining the proximal and distal borders of area CA2 and that the presence or absence of complex spines alone may not be suitable as a distinguishing feature differentiating CA3 from CA2 neurons.
Collapse
Affiliation(s)
- Daniel Radzicki
- Neurobiology Laboratory, National Institute of Environmental Health SciencesNational Institute of HealthResearch Triangle ParkNorth CarolinaUSA
| | - Sarah Chong
- Neurobiology Laboratory, National Institute of Environmental Health SciencesNational Institute of HealthResearch Triangle ParkNorth CarolinaUSA
| | - Serena M. Dudek
- Neurobiology Laboratory, National Institute of Environmental Health SciencesNational Institute of HealthResearch Triangle ParkNorth CarolinaUSA
| |
Collapse
|
12
|
Chevaleyre V, Piskorowski R. New hues for CA2. Hippocampus 2023; 33:161-165. [PMID: 36585825 DOI: 10.1002/hipo.23496] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 12/10/2022] [Accepted: 12/16/2022] [Indexed: 01/01/2023]
Affiliation(s)
- Vivien Chevaleyre
- INSERM U1266, Institute of Psychiatry and Neuroscience of Paris (IPNP), Université Paris Cité, Paris, France.,GHU Paris Psychiatrie et Neurosciences, Hôpital Sainte Anne, Paris, France
| | - Rebecca Piskorowski
- INSERM U1266, Institute of Psychiatry and Neuroscience of Paris (IPNP), Université Paris Cité, Paris, France.,GHU Paris Psychiatrie et Neurosciences, Hôpital Sainte Anne, Paris, France
| |
Collapse
|
13
|
Shinohara Y, Kohara K. Projections of hippocampal CA2 pyramidal neurons: Distinct innervation patterns of CA2 compared to CA3 in rodents. Hippocampus 2023; 33:691-699. [PMID: 36855258 DOI: 10.1002/hipo.23519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 02/01/2023] [Accepted: 02/14/2023] [Indexed: 03/02/2023]
Abstract
The hippocampus is a center for spatial and episodic memory formation in rodents. Understanding the composition of subregions and circuitry maps of the hippocampus is essential for elucidating the mechanism of memory formation and recall. For decades, the trisynaptic circuit (entorhinal cortex layer II-dentate gyrus - CA3-CA1) has been considered the neural network substrate responsible for learning and memory. Recently, CA2 has emerged as an important area in the hippocampal circuitry, with distinct functions from those of CA3. In this article, we review the historical definition of the hippocampal area CA2 and the differential projection patterns between CA2 and CA3 pyramidal neurons. We provide a concise and comprehensive map of CA2 outputs by comparing (1) ipsi versus contra projections, (2) septal versus temporal projections, and (3) lamellar structures of CA2 and CA3 pyramidal neurons.
Collapse
Affiliation(s)
- Yoshiaki Shinohara
- Department of Anatomy and Cell Biology, Faculty of Medicine, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Keigo Kohara
- KMU Biobank Center, Institute of Biomedical Science, Kansai Medical University, Hirakata, Osaka, Japan
| |
Collapse
|
14
|
Cholvin T, Bartos M. Hemisphere-specific spatial representation by hippocampal granule cells. Nat Commun 2022; 13:6227. [PMID: 36266288 PMCID: PMC9585038 DOI: 10.1038/s41467-022-34039-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 10/11/2022] [Indexed: 12/24/2022] Open
Abstract
The dentate gyrus (DG) output plays a key role in the emergence of spatial and contextual map representation within the hippocampus during learning. Differences in neuronal network activity have been observed between left and right CA1-3 areas, implying lateralization in spatial coding properties. Whether bilateral differences of DG granule cell (GC) assemblies encoding spatial and contextual information exist remains largely unexplored. Here, we employed two-photon calcium imaging of the left or the right DG to record the activity of GC populations over five consecutive days in head-fixed mice navigating through familiar and novel virtual environments. Imaging revealed similar mean GC activity on both sides. However, spatial tuning, context-selectivity and run-to-run place field reliability was markedly higher for DG place cells in the left than the right hemisphere. Moreover, the proportion of GCs reconfiguring their place fields between contexts was greater in the left DG. Thus, our data suggest that contextual information is differentially processed by GC populations depending on the hemisphere, with higher context discrimination in the left but a bias towards generalization in the right DG.
Collapse
Affiliation(s)
- Thibault Cholvin
- grid.5963.9Institute for Physiology I, University of Freiburg, Medical Faculty, 79104 Freiburg, Germany
| | - Marlene Bartos
- grid.5963.9Institute for Physiology I, University of Freiburg, Medical Faculty, 79104 Freiburg, Germany
| |
Collapse
|
15
|
Bittencourt LO, Damasceno-Silva RD, Aragão WAB, Eiró-Quirino L, Oliveira ACA, Fernandes RM, Freire MAM, Cartágenes SC, Dionizio A, Buzalaf MAR, Cassoli JS, Cirovic A, Cirovic A, Maia CDSF, Lima RR. Global Proteomic Profile of Aluminum-Induced Hippocampal Impairments in Rats: Are Low Doses of Aluminum Really Safe? Int J Mol Sci 2022; 23:ijms232012523. [PMID: 36293377 PMCID: PMC9603961 DOI: 10.3390/ijms232012523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/12/2022] [Accepted: 09/29/2022] [Indexed: 11/21/2022] Open
Abstract
Hippocampus is the brain area where aluminum (Al) accumulates in abundance and is widely associated with learning and memory. In the present study, we evaluate behavioral, tissue, and proteomic changes in the hippocampus of Wistar rats caused by exposure to doses that mimic human consumption of aluminum chloride (AlCl3) in urban areas. For this, male Wistar rats were divided into two groups: Control (distilled water) and AlCl3 (8.3 mg/kg/day), both groups were exposed orally for 60 days. After the Al exposure protocol, cognitive functions were assessed by the Water maze test, followed by a collection for analysis of the global proteomic profile of the hippocampus by mass spectrometry. Aside from proteomic analysis, we performed a histological analysis of the hippocampus, to the determination of cell body density by cresyl violet staining in Cornu Ammonis fields (CA) 1 and 3, and hilus regions. Our results indicated that exposure to low doses of aluminum chloride triggered a decreased cognitive performance in learning and memory, being associated with the deregulation of proteins expression, mainly those related to the regulation of the cytoskeleton, cellular metabolism, mitochondrial activity, redox regulation, nervous system regulation, and synaptic signaling, reduced cell body density in CA1, CA3, and hilus.
Collapse
Affiliation(s)
- Leonardo Oliveira Bittencourt
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém 66075110, Brazil
| | - Rakhel Dayanne Damasceno-Silva
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém 66075110, Brazil
| | - Walessa Alana Bragança Aragão
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém 66075110, Brazil
| | - Luciana Eiró-Quirino
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém 66075110, Brazil
| | - Ana Carolina Alves Oliveira
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém 66075110, Brazil
| | - Rafael Monteiro Fernandes
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém 66075110, Brazil
| | - Marco Aurelio M. Freire
- Graduate Program in Health and Society, Faculty of Health Sciences, State University of Rio Grande do Norte (UERN), Mossoro 59610210, Brazil
| | - Sabrina Carvalho Cartágenes
- Laboratory of Pharmacology of Inflammation and Behavior, Institute of Health Sciences, Federal University of Pará, Belém 66075110, Brazil
| | - Aline Dionizio
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Bauru 05508060, Brazil
| | | | - Juliana Silva Cassoli
- Institute of Biological Sciences, Federal University of Pará, Belém 66075110, Brazil
| | - Ana Cirovic
- Faculty of Medicine, Institute of Anatomy, University of Belgrade, 11000 Belgrade, Serbia
| | - Aleksandar Cirovic
- Faculty of Medicine, Institute of Anatomy, University of Belgrade, 11000 Belgrade, Serbia
| | - Cristiane do Socorro Ferraz Maia
- Laboratory of Pharmacology of Inflammation and Behavior, Institute of Health Sciences, Federal University of Pará, Belém 66075110, Brazil
| | - Rafael Rodrigues Lima
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém 66075110, Brazil
- Correspondence:
| |
Collapse
|
16
|
Bhasin G, Nair IR. Dynamic Hippocampal CA2 Responses to Contextual Spatial Novelty. Front Syst Neurosci 2022; 16:923911. [PMID: 36003545 PMCID: PMC9393711 DOI: 10.3389/fnsys.2022.923911] [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: 04/19/2022] [Accepted: 06/17/2022] [Indexed: 11/13/2022] Open
Abstract
Hippocampal place cells are functional units of spatial navigation and are present in all subregions: CA1, CA2, CA3, and CA4. Recent studies on CA2 have indicated its role in social and contextual memories, but its contribution to spatial novelty detection and encoding remains largely unknown. The current study aims to uncover how CA2 processes spatial novelty and to distinguish its functional role towards the same from CA1. Accordingly, a novel 3-day paradigm was designed where animals were introduced to a completely new environment on the first day, and on subsequent days, novel segments were inserted into the existing spatial environment while the other segments remained the same, allowing us to compare novel and familiar parts of the same closed-loop track on multiple days. We found that spatial novelty leads to dynamic and complex hippocampal place cell firings at both individual neuron and population levels. Place cells in both CA1 and CA2 had strong responses to novel segments, leading to higher average firing rates and increased pairwise cross correlations across all days. However, CA2 place cells that fired for novel areas had lower spatial information scores than CA1 place cells active in the same areas. At the ensemble level, CA1 only responded to spatial novelty on day 1, when the environment was completely novel, whereas CA2 responded to it on all days, each time novelty was introduced. Therefore, CA2 was more sensitive and responsive to novel spatial features even when introduced in a familiar environment, unlike CA1.
Collapse
|
17
|
Tsai TC, Fang YS, Hung YC, Hung LC, Hsu KS. A dorsal CA2 to ventral CA1 circuit contributes to oxytocinergic modulation of long-term social recognition memory. J Biomed Sci 2022; 29:50. [PMID: 35811321 PMCID: PMC9272559 DOI: 10.1186/s12929-022-00834-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 07/06/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Social recognition memory (SRM) is the ability to distinguish familiar from novel conspecifics and is crucial for survival and reproductive success across social species. We previously reported that oxytocin (OXT) receptor (OXTR) signaling in the CA2/CA3a of dorsal hippocampus is essential to promote the persistence of long-term SRM, yet how the endogenous OXT system influences CA2 outputs to regulate long-term SRM formation remains unclear. METHODS To achieve a selective deletion of CA2 OXTRs, we crossed Amigo2-Cre mice with Oxtr-floxed mice to generate CA2-specific Oxtr conditional knockout (Oxtr-/-) mice. A three-chamber paradigm test was used for studying SRM in mice. Chemogenetic and optogenetic targeting strategies were employed to manipulate neuronal activity. RESULTS We show that selective ablation of Oxtr in the CA2 suffices to impair the persistence of long-term SRM but has no effect on sociability and social novelty preference in the three-chamber paradigm test. We find that cell-type specific activation of OXT neurons within the hypothalamic paraventricular nucleus enhances long-term SRM and this enhancement is blocked by local application of OXTR antagonist L-368,899 into dorsal hippocampal CA2 (dCA2) region. In addition, chemogenetic neuronal silencing in dCA2 demonstrated that neuronal activity is essential for forming long-term SRM. Moreover, chemogenetic terminal-specific inactivation reveals a crucial role for dCA2 outputs to ventral CA1 (vCA1), but not dorsal lateral septum, in long-term SRM. Finally, targeted activation of the dCA2-to-vCA1 circuit effectively ameliorates long-term SRM deficit observed in Oxtr-/- mice. CONCLUSIONS These findings highlight the importance of hippocampal CA2 OXTR signaling in governing the persistence of long-term SRM and identify a hippocampal circuit linking dCA2 to vCA1 necessary for controlling long-term SRM formation.
Collapse
Affiliation(s)
- Tsung-Chih Tsai
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Yi-Syuan Fang
- Department of Pharmacology, College of Medicine, National Cheng Kung University, No. 1, University Rd., Tainan, 70101, Taiwan
| | - Yu-Chieh Hung
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Ling-Chien Hung
- Division of Neurology, Department of Internal Medicine, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi, 60002, Taiwan.
| | - Kuei-Sen Hsu
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan.
- Department of Pharmacology, College of Medicine, National Cheng Kung University, No. 1, University Rd., Tainan, 70101, Taiwan.
| |
Collapse
|
18
|
Nenninger AW, Willman M, Willman J, Stewart E, Mesidor P, Novoa M, Morrill NK, Alvarez L, Joly-Amado A, Peters MM, Gulick D, Nash KR. Improving Gene Therapy for Angelman Syndrome with Secreted Human UBE3A. Neurotherapeutics 2022; 19:1329-1339. [PMID: 35534672 PMCID: PMC9587189 DOI: 10.1007/s13311-022-01239-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/10/2022] [Indexed: 11/27/2022] Open
Abstract
The rare genetic neurodevelopmental disease Angelman syndrome (AS) is caused by the loss of function of UBE3A, a ubiquitin ligase. The disease results in a lifetime of severe symptoms, including intellectual disability and motor impairments for which there are no effective treatments. One avenue of treatment for AS is the use of gene therapy to reintroduce a functional copy of the UBE3A gene. Our group had previously shown that recombinant adeno-associated virus (rAAV) expressing mouse Ube3a could rescue deficits in a mouse model of AS. Here, we expand on this work and show that this approach could be successfully replicated in a second AS model using the human UBE3A gene. Furthermore, we address the challenge of limited vector distribution in the brain by developing a novel modified form of UBE3A. This modified protein, termed STUB, was designed with a secretion signal and a cell-penetrating peptide. This allowed transduced cells to act as factories for the production of UBE3A protein that could be taken up by neighboring non-transduced cells, thus increasing the number of neurons receiving the therapeutic protein. Combining this construct with intracerebroventricular injections to maximize rAAV distribution within the brain, we demonstrate that this novel approach improves the recovery of behavioral and electrophysiological deficits in the AS rat model. More importantly, a comparison of rAAV-STUB to a rAAV expressing the normal human UBE3A gene showed that STUB was a more effective therapeutic. These data suggest that rAAV-STUB is a new potential approach for the treatment of AS.
Collapse
Affiliation(s)
- Austin W Nenninger
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, FL-33612, USA
| | - Matthew Willman
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, FL-33612, USA
| | - Jonathan Willman
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, FL-33612, USA
| | - Emma Stewart
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, FL-33612, USA
| | - Philippe Mesidor
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, FL-33612, USA
| | - Michelle Novoa
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, FL-33612, USA
| | - Nicole K Morrill
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, FL-33612, USA
| | - Luis Alvarez
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, FL-33612, USA
| | - Aurélie Joly-Amado
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, FL-33612, USA
| | - Melinda M Peters
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, FL-33612, USA
| | - Danielle Gulick
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, FL-33612, USA
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, FL-33612, USA
| | - Kevin R Nash
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, FL-33612, USA.
| |
Collapse
|
19
|
Pimentel GA, Crestani AM, Florindo LH. Do spatial and recognition memories have a lateralized processing by the dorsal hippocampus CA3? Behav Brain Res 2022; 416:113566. [PMID: 34499937 DOI: 10.1016/j.bbr.2021.113566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 08/24/2021] [Accepted: 08/25/2021] [Indexed: 11/02/2022]
Abstract
The present study evaluated the function of the right and left CA3 of the dorsal hippocampus (dHPC) in the processing of (i) recognition memory, (ii) recent and remote spatial memory, (iii) working memory and (iv) navigation strategy. Wistar rats were divided into four experimental groups: vehicle group (VG), animals received a bilateral injection of phosphate-saline buffer (PBS) in both right and left dorsal CA3; dHPC-R group, animals received an injection of ibotenic acid (IBO) in the right dorsal CA3; dHPC-L group, animals received an IBO injection in left dorsal CA3; and dHPC-Bi group, animals received bilateral injections of IBO in both dorsal CA3. Rats were submitted to a sequence of behavioral tests: Morris water maze (MWM), object recognition test (ORT), forced T-maze and MWM 30 days after the first exposure. The results showed no evidence of functional lateralization and the dorsal CA3 does not seem to be essential for learning and memory (recent and remote) processing and allocentric navigation analyzed in the MWM and T-maze, respectively. However, rats with right or bilateral lesions in the dorsal CA3 failed to recognize the familiar object in the ORT, suggesting a lateralized processing of recognition memory. That result is unprecedented and contributes to the knowledge about the compartmentalization of HPC functions.
Collapse
Affiliation(s)
- Gabrielle Araujo Pimentel
- Department of Zoology and Botany, Universidade Estadual Paulista (UNESP), Rua Cristóvão Colombo, 2265, São José do Rio Preto, SP 15054-000, Brazil; Graduate Program in Animal Biology, Universidade Estadual Paulista (UNESP), Rua Cristóvão Colombo, 2265̥, São José do Rio Preto, SP 15054-000, Brazil.
| | - Ariela Maltarolo Crestani
- Department of Zoology and Botany, Universidade Estadual Paulista (UNESP), Rua Cristóvão Colombo, 2265, São José do Rio Preto, SP 15054-000, Brazil; Join Graduate Program in Physiological Sciences, Universidade Federal de São Carlos/Universidade Estadual Paulista (UFSCar/UNESP), Rodovia Washington Luiz, km 235̥, São Carlos, SP13565-905, Brazil.
| | - Luiz Henrique Florindo
- Department of Zoology and Botany, Universidade Estadual Paulista (UNESP), Rua Cristóvão Colombo, 2265, São José do Rio Preto, SP 15054-000, Brazil; Graduate Program in Animal Biology, Universidade Estadual Paulista (UNESP), Rua Cristóvão Colombo, 2265̥, São José do Rio Preto, SP 15054-000, Brazil; Join Graduate Program in Physiological Sciences, Universidade Federal de São Carlos/Universidade Estadual Paulista (UFSCar/UNESP), Rodovia Washington Luiz, km 235̥, São Carlos, SP13565-905, Brazil.
| |
Collapse
|
20
|
Silkis IG. The Role of Hypothalamus in the Formation of Neural Representations of Object–Place Associations in the Hippocampus during Wakefulness and Paradoxical Sleep. NEUROCHEM J+ 2021. [DOI: 10.1134/s1819712421020148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
21
|
Jeon S, Lee Y, Ryu D, Cho YK, Lee Y, Jun SB, Ji CH. Implantable Optrode Array for Optogenetic Modulation and Electrical Neural Recording. MICROMACHINES 2021; 12:mi12060725. [PMID: 34205473 PMCID: PMC8234104 DOI: 10.3390/mi12060725] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/14/2021] [Accepted: 06/16/2021] [Indexed: 02/06/2023]
Abstract
During the last decade, optogenetics has become an essential tool for neuroscience research due to its unrivaled feature of cell-type-specific neuromodulation. There have been several technological advances in light delivery devices. Among them, the combination of optogenetics and electrophysiology provides an opportunity for facilitating optogenetic approaches. In this study, a novel design of an optrode array was proposed for realizing optical modulation and electrophysiological recording. A 4 × 4 optrode array and five-channel recording electrodes were assembled as a disposable part, while a reusable part comprised an LED (light-emitting diode) source and a power line. After the characterization of the intensity of the light delivered at the fiber tips, in vivo animal experiment was performed with transgenic mice expressing channelrhodopsin, showing the effectiveness of optical activation and neural recording.
Collapse
Affiliation(s)
- Saeyeong Jeon
- Department of Electrical and Computer Engineering, Seoul National University, Seoul 08826, Korea; (S.J.); (D.R.)
| | - Youjin Lee
- Department of Electronic and Electrical Engineering, Ewha Womans University, Seoul 03760, Korea; (Y.L.); (Y.K.C.); (Y.L.); (S.B.J.)
- Graduate Program in Smart Factory, Ewha Womans University, Seoul 03760, Korea
| | - Daeho Ryu
- Department of Electrical and Computer Engineering, Seoul National University, Seoul 08826, Korea; (S.J.); (D.R.)
| | - Yoon Kyung Cho
- Department of Electronic and Electrical Engineering, Ewha Womans University, Seoul 03760, Korea; (Y.L.); (Y.K.C.); (Y.L.); (S.B.J.)
| | - Yena Lee
- Department of Electronic and Electrical Engineering, Ewha Womans University, Seoul 03760, Korea; (Y.L.); (Y.K.C.); (Y.L.); (S.B.J.)
| | - Sang Beom Jun
- Department of Electronic and Electrical Engineering, Ewha Womans University, Seoul 03760, Korea; (Y.L.); (Y.K.C.); (Y.L.); (S.B.J.)
- Graduate Program in Smart Factory, Ewha Womans University, Seoul 03760, Korea
- Department of Brain and Cognitive Sciences, Ewha Womans University, Seoul 03760, Korea
| | - Chang-Hyeon Ji
- Department of Electronic and Electrical Engineering, Ewha Womans University, Seoul 03760, Korea; (Y.L.); (Y.K.C.); (Y.L.); (S.B.J.)
- Graduate Program in Smart Factory, Ewha Womans University, Seoul 03760, Korea
- Correspondence: ; Tel.: +82-2-3277-3895
| |
Collapse
|
22
|
Tao S, Wang Y, Peng J, Zhao Y, He X, Yu X, Liu Q, Jin S, Xu F. Whole-Brain Mapping the Direct Inputs of Dorsal and Ventral CA1 Projection Neurons. Front Neural Circuits 2021; 15:643230. [PMID: 33935658 PMCID: PMC8079783 DOI: 10.3389/fncir.2021.643230] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 03/01/2021] [Indexed: 11/13/2022] Open
Abstract
The CA1, an important subregion of the hippocampus, is anatomically and functionally heterogeneous in the dorsal and ventral hippocampus. Here, to dissect the distinctions between the dorsal (dCA1) and ventral CA1 (vCA1) in anatomical connections, we systematically analyzed the direct inputs to dCA1 and vCA1 projection neurons (PNs) with the rabies virus-mediated retrograde trans-monosynaptic tracing system in Thy1-Cre mice. Our mapping results revealed that the input proportions and distributions of dCA1 and vCA1 PNs varied significantly. Inside the hippocampal region, dCA1 and vCA1 PNs shared the same upstream brain regions, but with distinctive distribution patterns along the rostrocaudal axis. The intrahippocampal inputs to the dCA1 and vCA1 exhibited opposite trends, decreasing and increasing gradually along the dorsoventral axis, respectively. For extrahippocampal inputs, dCA1 and vCA1 shared some monosynaptic projections from certain regions such as pallidum, striatum, hypothalamus, and thalamus. However, vCA1, not dCA1, received innervations from the subregions of olfactory areas and amygdala nuclei. Characterization of the direct input networks of dCA1 and vCA1 PNs may provide a structural basis to understand the differential functions of dCA1 and vCA1.
Collapse
Affiliation(s)
- Sijue Tao
- Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China.,State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Center for Magnetic Resonance, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, China
| | - Yihang Wang
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Center for Magnetic Resonance, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, China
| | - Jundan Peng
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Center for Magnetic Resonance, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, China
| | - Yang Zhao
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Center for Magnetic Resonance, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, China
| | - Xiaobin He
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Center for Magnetic Resonance, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Xuefeng Yu
- Materials and Interfaces Center, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Qing Liu
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Center for Magnetic Resonance, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, China.,Shenzhen Key Lab of Neuropsychiatric Modulation, Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.,Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
| | - Sen Jin
- Materials and Interfaces Center, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Fuqiang Xu
- Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China.,State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Center for Magnetic Resonance, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, China.,Shenzhen Key Lab of Neuropsychiatric Modulation, Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.,Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China.,Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
23
|
Lehr AB, Kumar A, Tetzlaff C, Hafting T, Fyhn M, Stöber TM. CA2 beyond social memory: Evidence for a fundamental role in hippocampal information processing. Neurosci Biobehav Rev 2021; 126:398-412. [PMID: 33775693 DOI: 10.1016/j.neubiorev.2021.03.020] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 03/16/2021] [Accepted: 03/18/2021] [Indexed: 01/16/2023]
Abstract
Hippocampal region CA2 has received increased attention due to its importance in social recognition memory. While its specific function remains to be identified, there are indications that CA2 plays a major role in a variety of situations, widely extending beyond social memory. In this targeted review, we highlight lines of research which have begun to converge on a more fundamental role for CA2 in hippocampus-dependent memory processing. We discuss recent proposals that speak to the computations CA2 may perform within the hippocampal circuit.
Collapse
Affiliation(s)
- Andrew B Lehr
- Department of Computational Neuroscience, University of Göttingen, Germany; Bernstein Center for Computational Neuroscience, University of Göttingen, Germany; Department of Computational Physiology, Simula Research Laboratory, Lysaker, Norway; Centre for Integrative Neuroplasticity, University of Oslo, Norway.
| | - Arvind Kumar
- Department of Computational Science and Technology, KTH Royal Institute of Technology, Sweden
| | - Christian Tetzlaff
- Department of Computational Neuroscience, University of Göttingen, Germany; Bernstein Center for Computational Neuroscience, University of Göttingen, Germany
| | - Torkel Hafting
- Centre for Integrative Neuroplasticity, University of Oslo, Norway; Institute of Basic Medical Sciences, University of Oslo, Norway
| | - Marianne Fyhn
- Centre for Integrative Neuroplasticity, University of Oslo, Norway; Department of Biosciences, University of Oslo, Norway
| | - Tristan M Stöber
- Department of Computational Physiology, Simula Research Laboratory, Lysaker, Norway; Centre for Integrative Neuroplasticity, University of Oslo, Norway; Department of Informatics, University of Oslo, Norway.
| |
Collapse
|
24
|
Wu Z, Xie C, Kuang H, Wu J, Chen X, Liu H, Liu T. Oxytocin mediates neuroprotection against hypoxic-ischemic injury in hippocampal CA1 neuron of neonatal rats. Neuropharmacology 2021; 187:108488. [PMID: 33556384 DOI: 10.1016/j.neuropharm.2021.108488] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 01/26/2021] [Accepted: 01/31/2021] [Indexed: 12/13/2022]
Abstract
Neonatal hypoxic-ischemic encephalopathy (NHIE) is one of the most prevalent causes of death during the perinatal period. The lack of exposure to oxytocin is associated with NHIE-mediated severe brain injury. However, the underlying mechanism is not fully understood. This study combined immunohistochemistry with electrophysiological recordings of hippocampal CA1 neurons to investigate the role of oxytocin in an in vitro model of hypoxic-ischemic (HI) injury (oxygen and glucose deprivation, OGD) in postnatal day 7-10 rats. Immunohistochemical analysis showed that oxytocin largely reduced the relative intensity of TOPRO-3 staining following OGD in the hippocampal CA1 region. Whole-cell patch-clamp recording revealed that the OGD-induced onset time of anoxic depolarization (AD) was significantly delayed by oxytocin. This protective effect of oxytocin was blocked by pretreatment with [d(CH2)51, Tyr (Me)2, Thr4, Orn8, des-Gly-NH29] vasotocin (dVOT, an oxytocin receptor antagonist) or bicuculline (a GABAA receptor antagonist). Interestingly, oxytocin enhanced inhibitory postsynaptic currents in CA1 pyramidal neurons, which were abolished by tetrodotoxin or dVOT. In contrast, oxytocin had no effect on excitatory postsynaptic currents but induced an inward current in 86% of the pyramidal neurons tested. Taken together, these results demonstrate that oxytocin receptor signaling plays a critical role in attenuating neonatal neural death by facilitating GABAergic transmission, which may help to regulate the excitatory-inhibitory balance in local neuronal networks in NHIE patients.
Collapse
Affiliation(s)
- Zhihong Wu
- Department of Pediatrics, The First Affiliated Hospital of Nanchang University, 17 St. Yongwaizheng, Nanchang, Jiangxi, 330006, PR China
| | - Changning Xie
- Department of Pediatrics, The First Affiliated Hospital of Nanchang University, 17 St. Yongwaizheng, Nanchang, Jiangxi, 330006, PR China
| | - Haixia Kuang
- Department of Pediatrics, The First Affiliated Hospital of Nanchang University, 17 St. Yongwaizheng, Nanchang, Jiangxi, 330006, PR China
| | - Jian Wu
- Department of Pediatrics, The First Affiliated Hospital of Nanchang University, 17 St. Yongwaizheng, Nanchang, Jiangxi, 330006, PR China
| | - Xiao Chen
- Department of Pediatrics, The First Affiliated Hospital of Nanchang University, 17 St. Yongwaizheng, Nanchang, Jiangxi, 330006, PR China
| | - Huibao Liu
- Department of Pediatrics, Xinyu Maternal and Child Health Hospital, 292 S. Laodong, Xinyu, Jiangxi, 338025, PR China.
| | - Tao Liu
- Department of Pediatrics, The First Affiliated Hospital of Nanchang University, 17 St. Yongwaizheng, Nanchang, Jiangxi, 330006, PR China.
| |
Collapse
|
25
|
Liu H, Tufa U, Zahra A, Chow J, Sivanenthiran N, Cheng C, Liu Y, Cheung P, Lim S, Jin Y, Mao M, Sun Y, Wu C, Wennberg R, Bardakjian B, Carlen PL, Eubanks JH, Song H, Zhang L. Electrographic Features of Spontaneous Recurrent Seizures in a Mouse Model of Extended Hippocampal Kindling. Cereb Cortex Commun 2021; 2:tgab004. [PMID: 34296153 PMCID: PMC8152854 DOI: 10.1093/texcom/tgab004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 01/08/2021] [Accepted: 01/13/2021] [Indexed: 01/14/2023] Open
Abstract
Epilepsy is a chronic neurological disorder characterized by spontaneous recurrent seizures (SRS) and comorbidities. Kindling through repetitive brief stimulation of a limbic structure is a commonly used model of temporal lobe epilepsy. Particularly, extended kindling over a period up to a few months can induce SRS, which may simulate slowly evolving epileptogenesis of temporal lobe epilepsy. Currently, electroencephalographic (EEG) features of SRS in rodent models of extended kindling remain to be detailed. We explored this using a mouse model of extended hippocampal kindling. Intracranial EEG recordings were made from the kindled hippocampus and unstimulated hippocampal, neocortical, piriform, entorhinal, or thalamic area in individual mice. Spontaneous EEG discharges with concurrent low-voltage fast onsets were observed from the two corresponding areas in nearly all SRS detected, irrespective of associated motor seizures. Examined in brain slices, epileptiform discharges were induced by alkaline artificial cerebrospinal fluid in the hippocampal CA3, piriform and entorhinal cortical areas of extended kindled mice but not control mice. Together, these in vivo and in vitro observations suggest that the epileptic activity involving a macroscopic network may generate concurrent discharges in forebrain areas and initiate SRS in hippocampally kindled mice.
Collapse
Affiliation(s)
- Haiyu Liu
- Departments of Neurosurgery, The First Hospital of Jilin University, Changchun, Jilin 130021 China.,Krembil Research Institute, University Health Network, Toronto, Ontario, Canada M5T 2S8
| | - Uilki Tufa
- Krembil Research Institute, University Health Network, Toronto, Ontario, Canada M5T 2S8.,Department of Electrical and Computer Engineering, University of Toronto, Toronto, Ontario M5S 3H5, Canada
| | - Anya Zahra
- Krembil Research Institute, University Health Network, Toronto, Ontario, Canada M5T 2S8
| | - Jonathan Chow
- Krembil Research Institute, University Health Network, Toronto, Ontario, Canada M5T 2S8
| | - Nila Sivanenthiran
- Krembil Research Institute, University Health Network, Toronto, Ontario, Canada M5T 2S8
| | - Chloe Cheng
- Krembil Research Institute, University Health Network, Toronto, Ontario, Canada M5T 2S8
| | - Yapg Liu
- Krembil Research Institute, University Health Network, Toronto, Ontario, Canada M5T 2S8
| | - Phinehas Cheung
- Krembil Research Institute, University Health Network, Toronto, Ontario, Canada M5T 2S8
| | - Stellar Lim
- Krembil Research Institute, University Health Network, Toronto, Ontario, Canada M5T 2S8
| | - Yaozhong Jin
- Krembil Research Institute, University Health Network, Toronto, Ontario, Canada M5T 2S8
| | - Min Mao
- Krembil Research Institute, University Health Network, Toronto, Ontario, Canada M5T 2S8
| | - Yuqing Sun
- Krembil Research Institute, University Health Network, Toronto, Ontario, Canada M5T 2S8
| | - Chiping Wu
- Krembil Research Institute, University Health Network, Toronto, Ontario, Canada M5T 2S8
| | - Richard Wennberg
- Krembil Research Institute, University Health Network, Toronto, Ontario, Canada M5T 2S8.,Department of Medicine, University of Toronto, Toronto, Ontario M2K 1E2, Canada
| | - Berj Bardakjian
- Department of Electrical and Computer Engineering, University of Toronto, Toronto, Ontario M5S 3H5, Canada
| | - Peter L Carlen
- Krembil Research Institute, University Health Network, Toronto, Ontario, Canada M5T 2S8.,Department of Medicine, University of Toronto, Toronto, Ontario M2K 1E2, Canada.,Department of Physiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - James H Eubanks
- Krembil Research Institute, University Health Network, Toronto, Ontario, Canada M5T 2S8.,Department of Surgery, University of Toronto, Toronto, Ontario M5G 1X5, Canada
| | - Hongmei Song
- Departments of Neurosurgery, The First Hospital of Jilin University, Changchun, Jilin 130021 China.,Krembil Research Institute, University Health Network, Toronto, Ontario, Canada M5T 2S8
| | - Liang Zhang
- Krembil Research Institute, University Health Network, Toronto, Ontario, Canada M5T 2S8.,Department of Medicine, University of Toronto, Toronto, Ontario M2K 1E2, Canada
| |
Collapse
|
26
|
Abbasi S, Nasehi M, Ebrahimi-Ghiri M, Zarrindast MR. Anodal tDCS applied to the left frontal cortex abrogates scopolamine-induced fear memory deficit via the dopaminergic system. Acta Neurobiol Exp (Wars) 2021. [DOI: 10.21307/ane-2021-016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
27
|
Ikawa F, Tanaka S, Harada K, Hide I, Maruyama H, Sakai N. Detailed neuronal distribution of GPR3 and its co-expression with EF-hand calcium-binding proteins in the mouse central nervous system. Brain Res 2020; 1750:147166. [PMID: 33075309 DOI: 10.1016/j.brainres.2020.147166] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 10/07/2020] [Accepted: 10/12/2020] [Indexed: 12/11/2022]
Abstract
The G-protein coupled receptor 3 (GPR3), a member of the class A rhodopsin-type GPR family, constitutively activates Gαs proteins without any ligands. Although there have been several reports concerning the functions of GPR3 in neurons, the physiological roles of GPR3 have not been fully elucidated. To address this issue, we analyzed GPR3 distribution in detail using fluorescence-based X-gal staining in heterozygous GPR3 knockout/LacZ knock-in mice, and further investigated the types of GPR3-expressing neurons using fluorescent double labeling with various EF-hand Ca2+-binding proteins. In addition to the previously reported GPR3-expressing areas, we identified GPR3 expression in the basal ganglia and in many nuclei of the cranial nerves, in regions related to olfactory, auditory, emotional, and motor functions. In addition, GPR3 was not only observed in excitatory neurons in layer V of the cerebral cortex, the CA2 region of the hippocampus, and the lateral nucleus of the thalamus, but also in γ-aminobutyric acid (GABA)-ergic interneurons in the cortex, hippocampus, thalamus, striatum, and cerebellum. GPR3 was frequently co-expressed with neuronal Ca2+-binding protein 2 (NECAB2) in neurons in various regions of the central nervous system, especially in the hippocampal CA2, medial habenular nucleus, lateral thalamic nucleus, dorsolateral striatum, brainstem, and spinal cord anterior horn. Furthermore, GPR3 also co-localized with NECAB2 at the tips of neurites in differentiated PC12 cells. These results suggest that GPR3 and NECAB2 are highly co-expressed in specific neurons, and that GPR3 may modulate Ca2+ signaling by interacting with NECAB2 in specific areas of the central nervous system.
Collapse
Affiliation(s)
- Fumiaki Ikawa
- Department of Molecular and Pharmacological Neuroscience, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan; Department of Neurology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Shigeru Tanaka
- Department of Molecular and Pharmacological Neuroscience, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan.
| | - Kana Harada
- Department of Molecular and Pharmacological Neuroscience, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Izumi Hide
- Department of Molecular and Pharmacological Neuroscience, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Hirofumi Maruyama
- Department of Neurology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Norio Sakai
- Department of Molecular and Pharmacological Neuroscience, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
28
|
Kleindienst D, Montanaro J, Bhandari P, Case MJ, Fukazawa Y, Shigemoto R. Deep Learning-Assisted High-Throughput Analysis of Freeze-Fracture Replica Images Applied to Glutamate Receptors and Calcium Channels at Hippocampal Synapses. Int J Mol Sci 2020; 21:E6737. [PMID: 32937911 PMCID: PMC7555218 DOI: 10.3390/ijms21186737] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 09/07/2020] [Accepted: 09/10/2020] [Indexed: 12/15/2022] Open
Abstract
The molecular anatomy of synapses defines their characteristics in transmission and plasticity. Precise measurements of the number and distribution of synaptic proteins are important for our understanding of synapse heterogeneity within and between brain regions. Freeze-fracture replica immunogold electron microscopy enables us to analyze them quantitatively on a two-dimensional membrane surface. Here, we introduce Darea software, which utilizes deep learning for analysis of replica images and demonstrate its usefulness for quick measurements of the pre- and postsynaptic areas, density and distribution of gold particles at synapses in a reproducible manner. We used Darea for comparing glutamate receptor and calcium channel distributions between hippocampal CA3-CA1 spine synapses on apical and basal dendrites, which differ in signaling pathways involved in synaptic plasticity. We found that apical synapses express a higher density of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors and a stronger increase of AMPA receptors with synaptic size, while basal synapses show a larger increase in N-methyl-D-aspartate (NMDA) receptors with size. Interestingly, AMPA and NMDA receptors are segregated within postsynaptic sites and negatively correlated in density among both apical and basal synapses. In the presynaptic sites, Cav2.1 voltage-gated calcium channels show similar densities in apical and basal synapses with distributions consistent with an exclusion zone model of calcium channel-release site topography.
Collapse
Affiliation(s)
- David Kleindienst
- Institute of Science and Technology (IST )Austria, 3400 Klosterneuburg, Austria; (J.M.); (P.B.); (M.J.C.)
| | - Jacqueline Montanaro
- Institute of Science and Technology (IST )Austria, 3400 Klosterneuburg, Austria; (J.M.); (P.B.); (M.J.C.)
| | - Pradeep Bhandari
- Institute of Science and Technology (IST )Austria, 3400 Klosterneuburg, Austria; (J.M.); (P.B.); (M.J.C.)
| | - Matthew J. Case
- Institute of Science and Technology (IST )Austria, 3400 Klosterneuburg, Austria; (J.M.); (P.B.); (M.J.C.)
| | - Yugo Fukazawa
- Department of Histological and Physiological Sciences, Faculty of Medical Science, University of Fukui, Yoshida, Fukui 910-1193, Japan;
| | - Ryuichi Shigemoto
- Institute of Science and Technology (IST )Austria, 3400 Klosterneuburg, Austria; (J.M.); (P.B.); (M.J.C.)
| |
Collapse
|
29
|
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
|
30
|
Brzdak P, Wójcicka O, Zareba-Koziol M, Minge D, Henneberger C, Wlodarczyk J, Mozrzymas JW, Wójtowicz T. Synaptic Potentiation at Basal and Apical Dendrites of Hippocampal Pyramidal Neurons Involves Activation of a Distinct Set of Extracellular and Intracellular Molecular Cues. Cereb Cortex 2020; 29:283-304. [PMID: 29228131 DOI: 10.1093/cercor/bhx324] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 11/07/2017] [Indexed: 12/12/2022] Open
Abstract
In the central nervous system, several forms of experience-dependent plasticity, learning and memory require the activity-dependent control of synaptic efficacy. Despite substantial progress in describing synaptic plasticity, mechanisms related to heterogeneity of synaptic functions at local circuits remain elusive. Here we studied the functional and molecular aspects of hippocampal circuit plasticity by analyzing excitatory synapses at basal and apical dendrites of mouse hippocampal pyramidal cells (CA1 region) in acute brain slices. In the past decade, activity of metalloproteinases (MMPs) has been implicated as a widespread and critical factor in plasticity mechanisms at various projections in the CNS. However, in the present study we discovered that in striking contrast to apical dendrites, synapses located within basal dendrites undergo MMP-independent synaptic potentiation. We demonstrate that synapse-specific molecular pathway allowing MMPs to rapidly upregulate function of NMDARs in stratum radiatum involved protease activated receptor 1 and intracellular kinases and GTPases activity. In contrast, MMP-independent scaling of synaptic strength in stratum oriens involved dopamine D1/D5 receptors and Src kinases. Results of this study reveal that 2 neighboring synaptic systems differ significantly in extracellular and intracellular cascades that control synaptic gain and provide long-searched transduction pathways relevant for MMP-dependent synaptic plasticity.
Collapse
Affiliation(s)
- Patrycja Brzdak
- Laboratory of Neuroscience, Department of Biophysics, Wroclaw Medical University, Wroclaw, Poland.,Department of Molecular Physiology and Neurobiology, Wroclaw University, Wroclaw, Poland
| | - Olga Wójcicka
- Laboratory of Neuroscience, Department of Biophysics, Wroclaw Medical University, Wroclaw, Poland
| | - Monika Zareba-Koziol
- Laboratory of Cell Biophysics, Department of Molecular and Cellular Neurobiology, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Daniel Minge
- Institute of Cellular Neurosciences, University of Bonn Medical School, Bonn, Germany
| | - Christian Henneberger
- Institute of Cellular Neurosciences, University of Bonn Medical School, Bonn, Germany.,Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Institute of Neurology, University College London, London, UK
| | - Jakub Wlodarczyk
- Laboratory of Cell Biophysics, Department of Molecular and Cellular Neurobiology, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Jerzy W Mozrzymas
- Laboratory of Neuroscience, Department of Biophysics, Wroclaw Medical University, Wroclaw, Poland.,Department of Molecular Physiology and Neurobiology, Wroclaw University, Wroclaw, Poland
| | - Tomasz Wójtowicz
- Laboratory of Neuroscience, Department of Biophysics, Wroclaw Medical University, Wroclaw, Poland
| |
Collapse
|
31
|
Abstract
Although Lorente de No' recognized the anatomical distinction of the hippocampal Cornu Ammonis (CA) 2 region, it had, until recently, been assigned no unique function. Its location between the key players of the circuit, CA3 and CA1, which along with the entorhinal cortex and dentate gyrus compose the classic trisynaptic circuit, further distracted research interest. However, the connectivity of CA2 pyramidal cells, together with unique patterns of gene expression, hints at a much larger contribution to hippocampal information processing than has been ascribed. Here we review recent advances that have identified new roles for CA2 in hippocampal centric processing, together with specialized functions in social memory and, potentially, as a broadcaster of novelty. These new data, together with CA2's role in disease, justify a closer look at how this small region exerts its influence and how it might best be exploited to understand and treat disease-related circuit dysfunctions.
Collapse
Affiliation(s)
- Steven J Middleton
- Laboratory for Circuit and Behavioral Physiology, RIKEN Center for Brain Science, Wako-shi, Saitama 351-0198, Japan; ,
| | - Thomas J McHugh
- Laboratory for Circuit and Behavioral Physiology, RIKEN Center for Brain Science, Wako-shi, Saitama 351-0198, Japan; ,
| |
Collapse
|
32
|
Lee SL(T, Lew D, Wickenheisser V, Markus EJ. Interdependence between dorsal and ventral hippocampus during spatial navigation. Brain Behav 2019; 9:e01410. [PMID: 31571397 PMCID: PMC6790314 DOI: 10.1002/brb3.1410] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 08/15/2019] [Indexed: 11/10/2022] Open
Abstract
INTRODUCTION The hippocampus is linked to the formation and retrieval of episodic memories and spatial navigation. In rats, it is an elongated structure divided into dorsal (septal) and ventral (temporal) regions paralleling the respective division in the posterior and anterior hippocampus in humans. The dorsal hippocampus has been suggested to be more important for spatial processing and the ventral to processing anxiety-based behaviors. Far less is known regarding the degree to which these different regions interact during information processing. The anatomical connectivity suggests a flow of information between the dorsal and ventral regions; conversely, there are also commissural connections to the contralateral hippocampus. The current study examined the extent to which information from the dorsal hippocampus interacts with processing in the ipsilateral and contralateral ventral hippocampus following the acquisition of a spatial task. METHODS Rats were well-trained on a spatial reference version of the water maze, followed by muscimol inactivation of different hippocampal subregions in a within-animal repeated design. Various combinations of bilateral, ipsilateral, and contralateral infusions were used. RESULTS Combined dorsal and ventral inactivation produced a severe impairment in spatial performance. Inactivation of only the dorsal or ventral regions resulted in intermediate impairment with performance levels falling between controls and combined inactivation. Performance was impaired during contralateral inactivation and was almost equivalent to bilateral dorsal and ventral hippocampus inactivation, while ipsilateral inactivation resulted in little impairment. CONCLUSIONS Taken together, results indicate that for spatial processing, the hippocampus functions as a single integrated structure along the longitudinal axis.
Collapse
Affiliation(s)
- Shang Lin (Tommy) Lee
- Behavioral Neuroscience DivisionDepartment of Psychological SciencesUniversity of ConnecticutStorrsCTUSA
| | - Dana Lew
- Behavioral Neuroscience DivisionDepartment of Psychological SciencesUniversity of ConnecticutStorrsCTUSA
| | - Victoria Wickenheisser
- Behavioral Neuroscience DivisionDepartment of Psychological SciencesUniversity of ConnecticutStorrsCTUSA
| | - Etan J. Markus
- Behavioral Neuroscience DivisionDepartment of Psychological SciencesUniversity of ConnecticutStorrsCTUSA
| |
Collapse
|
33
|
Cox BM, Cox CD, Gunn BG, Le AA, Inshishian VC, Gall CM, Lynch G. Acquisition of temporal order requires an intact CA3 commissural/associational (C/A) feedback system in mice. Commun Biol 2019; 2:251. [PMID: 31286068 PMCID: PMC6610080 DOI: 10.1038/s42003-019-0494-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 05/30/2019] [Indexed: 12/31/2022] Open
Abstract
Episodic memory, an essential element of orderly thinking, requires the organization of serial events into narratives about the identity of cues along with their locations and temporal order (what, where, and when). The hippocampus plays a central role in the acquisition and retrieval of episodes with two of its subsystems being separately linked to what and where information. The substrates for the third element are poorly understood. Here we report that in hippocampal slices field CA3 maintains self-sustained activity for remarkable periods following a brief input and that this effect is extremely sensitive to minor network perturbations. Using behavioral tests, that do not involve training or explicit rewards, we show that partial silencing of the CA3 commissural/associational network in mice blocks acquisition of temporal order, but not the identity or location, of odors. These results suggest a solution to the question of how hippocampus adds time to episodic memories.
Collapse
Affiliation(s)
- Brittney M. Cox
- Department of Anatomy and Neurobiology, University of California, Irvine, CA 92697 USA
| | - Conor D. Cox
- Department of Anatomy and Neurobiology, University of California, Irvine, CA 92697 USA
| | - Benjamin G. Gunn
- Department of Anatomy and Neurobiology, University of California, Irvine, CA 92697 USA
| | - Aliza A. Le
- Department of Anatomy and Neurobiology, University of California, Irvine, CA 92697 USA
| | | | - Christine M. Gall
- Department of Anatomy and Neurobiology, University of California, Irvine, CA 92697 USA
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697 USA
| | - Gary Lynch
- Department of Anatomy and Neurobiology, University of California, Irvine, CA 92697 USA
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA 92697 USA
| |
Collapse
|
34
|
O'Riordan KJ, Hu NW, Rowan MJ. Aß Facilitates LTD at Schaffer Collateral Synapses Preferentially in the Left Hippocampus. Cell Rep 2019; 22:2053-2065. [PMID: 29466733 DOI: 10.1016/j.celrep.2018.01.085] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 12/16/2017] [Accepted: 01/26/2018] [Indexed: 01/03/2023] Open
Abstract
Promotion of long-term depression (LTD) mechanisms by synaptotoxic soluble oligomers of amyloid-β (Aß) has been proposed to underlie synaptic dysfunction in Alzheimer's disease (AD). Previously, LTD was induced by relatively non-specific electrical stimulation. Exploiting optogenetics, we studied LTD using a more physiologically diffuse spatial pattern of selective pathway activation in the rat hippocampus in vivo. This relatively sparse synaptic LTD requires both the ion channel function and GluN2B subunit of the NMDA receptor but, in contrast to electrically induced LTD, is not facilitated by boosting endogenous muscarinic acetylcholine or metabotropic glutamate 5 receptor activation. Although in the absence of Aß, there is no evidence of hippocampal LTD asymmetry, in the presence of Aß, the induction of LTD is preferentially enhanced in the left hippocampus in an mGluR5-dependent manner. This circuit-selective disruption of synaptic plasticity by Aß provides a route to understanding the development of aberrant brain lateralization in AD.
Collapse
Affiliation(s)
- Kenneth J O'Riordan
- Department of Pharmacology and Therapeutics and Institute of Neuroscience, Watts Building, Trinity College, Dublin 2, Ireland
| | - Neng-Wei Hu
- Department of Pharmacology and Therapeutics and Institute of Neuroscience, Watts Building, Trinity College, Dublin 2, Ireland; Department of Gerontology, Yijishan Hospital, Wannan Medical College, Wuhu, China.
| | - Michael J Rowan
- Department of Pharmacology and Therapeutics and Institute of Neuroscience, Watts Building, Trinity College, Dublin 2, Ireland.
| |
Collapse
|
35
|
Valeeva G, Nasretdinov A, Rychkova V, Khazipov R. Bilateral Synchronization of Hippocampal Early Sharp Waves in Neonatal Rats. Front Cell Neurosci 2019; 13:29. [PMID: 30792630 PMCID: PMC6374346 DOI: 10.3389/fncel.2019.00029] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 01/22/2019] [Indexed: 11/17/2022] Open
Abstract
In the neonatal rodent hippocampus, the first and predominant pattern of correlated neuronal network activity is early sharp waves (eSPWs). Whether and how eSPWs are organized bilaterally remains unknown. Here, using simultaneous silicone probe recordings from the left and right hippocampus in neonatal rats in vivo we found that eSPWs are highly synchronized bilaterally with nearly zero time lag between the two sides. The amplitudes of eSPWs in the left and right hippocampi were also highly correlated. eSPWs also supported bilateral synchronization of multiple unit activity (MUA). We suggest that bilateral correlated activity supported by synchronized eSPWs participates in the formation of bilateral connections in the hippocampal system.
Collapse
Affiliation(s)
- Guzel Valeeva
- Laboratory of Neurobiology, Kazan Federal University, Kazan, Russia
| | - Azat Nasretdinov
- Laboratory of Neurobiology, Kazan Federal University, Kazan, Russia
| | | | - Roustem Khazipov
- Laboratory of Neurobiology, Kazan Federal University, Kazan, Russia.,Aix-Marseille University, INMED, Institut National de la Santé et de la Recherche Médicale (INSERM), Marseille, France
| |
Collapse
|
36
|
Blázquez G, Castañé A, Saavedra A, Masana M, Alberch J, Pérez-Navarro E. Social Memory and Social Patterns Alterations in the Absence of STriatal-Enriched Protein Tyrosine Phosphatase. Front Behav Neurosci 2019; 12:317. [PMID: 30760987 PMCID: PMC6362413 DOI: 10.3389/fnbeh.2018.00317] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 12/04/2018] [Indexed: 01/23/2023] Open
Abstract
STriatal-Enriched protein tyrosine Phosphatase (STEP) is a neural-specific protein that opposes the development of synaptic strengthening and whose levels are altered in several neurodegenerative and psychiatric disorders. Since STEP is expressed in brain regions implicated in social behavior, namely the striatum, the CA2 region of the hippocampus, cortex and amygdala, here we investigated whether social memory and social patterns were altered in STEP knockout (KO) mice. Our data robustly demonstrated that STEP KO mice presented specific social memory impairment as indicated by the three-chamber sociability test, the social discrimination test, the 11-trial habituation/dishabituation social recognition test, and the novel object recognition test (NORT). This affectation was not related to deficiencies in the detection of social olfactory cues, altered sociability or anxiety levels. However, STEP KO mice showed lower exploratory activity, reduced interaction time with an intruder, less dominant behavior and higher immobility time in the tail suspension test than controls, suggesting alterations in motivation. Moreover, the extracellular levels of dopamine (DA), but not serotonin (5-HT), were increased in the dorsal striatum of STEP KO mice. Overall, our results indicate that STEP deficiency disrupts social memory and other social behaviors as well as DA homeostasis in the dorsal striatum.
Collapse
Affiliation(s)
- Gloria Blázquez
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - Anna Castañé
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Department of Neurochemistry and Neuropharmacology, CSIC-Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Barcelona, Spain.,Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Barcelona, Spain
| | - Ana Saavedra
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - Mercè Masana
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - Jordi Alberch
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - Esther Pérez-Navarro
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| |
Collapse
|
37
|
Helton TD, Zhao M, Farris S, Dudek SM. Diversity of dendritic morphology and entorhinal cortex synaptic effectiveness in mouse CA2 pyramidal neurons. Hippocampus 2018; 29:78-92. [PMID: 30067288 DOI: 10.1002/hipo.23012] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 07/09/2018] [Accepted: 07/11/2018] [Indexed: 11/06/2022]
Abstract
Excitatory synaptic inputs from specific brain regions are often targeted to distinct dendritic arbors on hippocampal pyramidal neurons. Recent work has suggested that CA2 pyramidal neurons respond robustly and preferentially to excitatory input into the stratum lacunosum moleculare (SLM), with a relatively modest response to Schaffer collateral excitatory input into stratum radiatum (SR) in acute mouse hippocampal slices, but the extent to which this difference may be explained by morphology is unknown. In an effort to replicate these findings and to better understand the role of dendritic morphology in shaping responses from proximal and distal synaptic sites, we measured excitatory postsynaptic currents and action potentials in CA2 pyramidal cells in response to SR and SLM stimulation and subsequently analyzed confocal images of the filled cells. We found that, in contrast to previous reports, SR stimulation evoked substantial responses in all recorded CA2 pyramidal cells. Strikingly, however, we found that not all neurons responded to SLM stimulation, and in those neurons that did, responses evoked by SLM and SR were comparable in size and effectiveness in inducing action potentials. In a comprehensive morphometric analysis of CA2 pyramidal cell apical dendrites, we found that the neurons that were unresponsive to SLM stimulation were the same ones that lacked substantial apical dendritic arborization in the SLM. Neurons responsive to both SR and SLM stimulation had roughly equal amounts of dendritic branching in each layer. Remarkably, our study in mouse CA2 generally replicates the work characterizing the diversity of CA2 pyramidal cells in the guinea pig hippocampus. We conclude, then, that like in guinea pig, mouse CA2 pyramidal cells have a diverse apical dendrite morphology that is likely to be reflective of both the amount and source of excitatory input into CA2 from the entorhinal cortex and CA3.
Collapse
Affiliation(s)
- Thomas D Helton
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Meilan Zhao
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Shannon Farris
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Serena M Dudek
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| |
Collapse
|
38
|
Long-Term Potentiation and Excitability in the Hippocampus Are Modulated Differently by θ Rhythm. eNeuro 2018; 5:eN-CFN-0236-18. [PMID: 30627662 PMCID: PMC6325566 DOI: 10.1523/eneuro.0236-18.2018] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 11/02/2018] [Accepted: 11/05/2018] [Indexed: 01/27/2023] Open
Abstract
Oscillations in the brain facilitate neural processing and cognitive functions. This study investigated the dependence of long-term potentiation (LTP), a neural correlate of memory, on the phase of the hippocampal θ rhythm, a prominent brain oscillation. Multichannel field potentials and current source-sinks were analyzed in hippocampal CA1 of adult male rats under urethane anesthesia. A single burst (five pulses at 200 Hz) stimulation of stratum oriens (OR) induced LTP of the basal dendritic excitatory sink (ES), which was maximal when the burst was delivered at ∼340° and ∼160° of the distal dendritic θ rhythm. Apical dendritic sink evoked by stratum radiatum (RAD) stimulation also showed biphasic maxima at ∼30° and ∼210° of the distal dendritic θ rhythm, about 50° phase delay to basal dendritic LTP. By contrast, maximal population spike (PS) excitability, following single-pulse excitation of the basal or mid-apical dendrites, occurred at a θ phase of ∼140°, and maximal basal dendritic ES occurred at ∼20°; γ (30–57 Hz) activity recorded in CA1 RAD had maximal power at ∼300° of the distal dendritic θ rhythm, different from the phases of maximal LTP. LTP induced during the rising θ phase was NMDA receptor sensitive. It is suggested that the θ phase modulation of CA1 PS excitability is mainly provided by θ-rhythmic proximal inhibition, while dendritic LTP is also modulated by dendritic inhibition and excitation, specific to basal and apical dendrites. In summary, basal and apical dendritic synaptic plasticity and spike excitability are facilitated at different θ phases in a compartmental fashion.
Collapse
|
39
|
Tirko NN, Eyring KW, Carcea I, Mitre M, Chao MV, Froemke RC, Tsien RW. Oxytocin Transforms Firing Mode of CA2 Hippocampal Neurons. Neuron 2018; 100:593-608.e3. [PMID: 30293821 DOI: 10.1016/j.neuron.2018.09.008] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2016] [Revised: 01/03/2018] [Accepted: 09/04/2018] [Indexed: 01/30/2023]
Abstract
Oxytocin is an important neuromodulator in the mammalian brain that increases information salience and circuit plasticity, but its signaling mechanisms and circuit effect are not fully understood. Here we report robust oxytocinergic modulation of intrinsic properties and circuit operations in hippocampal area CA2, a region of emerging importance for hippocampal function and social behavior. Upon oxytocin receptor activation, CA2 pyramidal cells depolarize and fire bursts of action potentials, a consequence of phospholipase C signaling to modify two separate voltage-dependent ionic processes. A reduction of potassium current carried by KCNQ-based M channels depolarizes the cell; protein kinase C activity attenuates spike rate of rise and overshoot, dampening after-hyperpolarizations. These actions, in concert with activation of fast-spiking interneurons, promote repetitive firing and CA2 bursting; bursting then governs short-term plasticity of CA2 synaptic transmission onto CA1 and, thus, efficacy of information transfer in the hippocampal network.
Collapse
Affiliation(s)
- Natasha N Tirko
- NYU Neuroscience Institute, New York University School of Medicine, New York, NY 10016, USA
| | - Katherine W Eyring
- NYU Neuroscience Institute, New York University School of Medicine, New York, NY 10016, USA
| | - Ioana Carcea
- NYU Neuroscience Institute, New York University School of Medicine, New York, NY 10016, USA; Skirball Institute, New York University School of Medicine, New York, NY 10016, USA; Department of Otolaryngology, New York University School of Medicine, New York, NY 10016, USA
| | - Mariela Mitre
- NYU Neuroscience Institute, New York University School of Medicine, New York, NY 10016, USA; Skirball Institute, New York University School of Medicine, New York, NY 10016, USA; Department of Otolaryngology, New York University School of Medicine, New York, NY 10016, USA
| | - Moses V Chao
- NYU Neuroscience Institute, New York University School of Medicine, New York, NY 10016, USA; Skirball Institute, New York University School of Medicine, New York, NY 10016, USA
| | - Robert C Froemke
- NYU Neuroscience Institute, New York University School of Medicine, New York, NY 10016, USA; Skirball Institute, New York University School of Medicine, New York, NY 10016, USA; Department of Otolaryngology, New York University School of Medicine, New York, NY 10016, USA
| | - Richard W Tsien
- NYU Neuroscience Institute, New York University School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
40
|
García-Forn M, Martínez-Torres S, García-Díaz Barriga G, Alberch J, Milà M, Azkona G, Pérez-Navarro E. Pharmacogenetic modulation of STEP improves motor and cognitive function in a mouse model of Huntington's disease. Neurobiol Dis 2018; 120:88-97. [PMID: 30176350 DOI: 10.1016/j.nbd.2018.08.024] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 07/31/2018] [Accepted: 08/30/2018] [Indexed: 12/27/2022] Open
Abstract
Huntington's disease (HD) is a hereditary neurodegenerative disorder caused by an expansion of a CAG repeat in the huntingtin (htt) gene, which results in an aberrant form of the protein (mhtt). This leads to motor and cognitive deficits associated with corticostriatal and hippocampal alterations. The levels of STriatal-Enriched protein tyrosine Phosphatase (STEP), a neural-specific tyrosine phosphatase that opposes the development of synaptic strengthening, are decreased in the striatum of HD patients and also in R6/1 mice, thereby contributing to the resistance to excitotoxicity described in this HD mouse model. Here, we aimed to analyze whether STEP inactivation plays a role in the pathophysiology of HD by investigating its effect on motor and cognitive impairment in the R6/1 mouse model of HD. We found that genetic deletion of STEP delayed the onset of motor dysfunction and prevented the appearance of cognitive deficits in R6/1 mice. This phenotype was accompanied by an increase in pERK1/2 levels, a delay in the decrease of striatal DARPP-32 levels and a reduction in the size of mhtt aggregates, both in the striatum and CA1 hippocampal region. We also found that acute pharmacological inhibition of STEP with TC-2153 improved cognitive function in R6/1 mice. In conclusion, our results show that deletion of STEP has a beneficial effect on motor coordination and cognition in a mouse model of HD suggesting that STEP inhibition could be a good therapeutic strategy in HD patients.
Collapse
Affiliation(s)
- Marta García-Forn
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, Barcelona, Catalonia, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Sara Martínez-Torres
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Gerardo García-Díaz Barriga
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, Barcelona, Catalonia, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Jordi Alberch
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, Barcelona, Catalonia, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Montse Milà
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia, Spain; Departament de Genètica, Hospital Clínic de Barcelona, Barcelona, Catalonia, Spain
| | - Garikoitz Azkona
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, Barcelona, Catalonia, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Esther Pérez-Navarro
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, Barcelona, Catalonia, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain.
| |
Collapse
|
41
|
Janz P, Hauser P, Heining K, Nestel S, Kirsch M, Egert U, Haas CA. Position- and Time-Dependent Arc Expression Links Neuronal Activity to Synaptic Plasticity During Epileptogenesis. Front Cell Neurosci 2018; 12:244. [PMID: 30154698 PMCID: PMC6102356 DOI: 10.3389/fncel.2018.00244] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 07/18/2018] [Indexed: 12/14/2022] Open
Abstract
In mesial temporal lobe epilepsy (mTLE) an initial precipitating injury can trigger aberrant wiring of neuronal circuits causing seizure activity. While circuit reorganization is known to be largely activity-dependent, the interactions between neuronal activity and synaptic plasticity during the development of mTLE remain poorly understood. Therefore, the present study aimed at delineating the spatiotemporal relationship between epileptic activity, activity-dependent gene expression and synaptic plasticity during kainic acid-induced epileptogenesis in mice. We show that during epileptogenesis the sclerotic hippocampus differed from non-sclerotic regions by displaying a consistently lower power of paroxysmal discharges. However, the power of these discharges steadily increased during epileptogenesis. This increase was paralleled by the upregulation of the activity-related cytoskeleton protein (Arc) gene expression in dentate granule cells (DGCs) of the sclerotic hippocampus. Importantly, we found that Arc mRNA-upregulating DGCs exhibited increased spine densities and spine sizes, but at the same time decreased AMPA-type glutamate receptor (AMPAR) densities. Finally, we show that in vivo optogenetic stimulation of DGC synapses evoked robust seizure activity in epileptic mice, but failed to induce dendritic translocation of Arc mRNA as under healthy conditions, supporting the theory of a breakdown of the dentate gate in mTLE. We conclude that during epileptogenesis epileptic activity emerges early and persists in the whole hippocampus, however, only the sclerotic part shows modulation of discharge amplitudes accompanied by plasticity of DGCs. In this context, we identified Arc as a putative mediator between seizure activity and synaptic plasticity.
Collapse
Affiliation(s)
- Philipp Janz
- Experimental Epilepsy Research, Department of Neurosurgery, University Medical Center, University of Freiburg, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Pascal Hauser
- Experimental Epilepsy Research, Department of Neurosurgery, University Medical Center, University of Freiburg, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Katharina Heining
- Faculty of Biology, University of Freiburg, Freiburg, Germany.,Laboratory for Biomicrotechnology, Department of Microsystems Engineering, University of Freiburg, Freiburg, Germany.,Bernstein Center Freiburg, University of Freiburg, Freiburg, Germany
| | - Sigrun Nestel
- Institute for Anatomy and Cell Biology, Department of Neuroanatomy, University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Matthias Kirsch
- Institute for Anatomy and Cell Biology, Department of Neuroanatomy, University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany.,BrainLinks-BrainTools Cluster of Excellence, University of Freiburg, Freiburg, Germany
| | - Ulrich Egert
- Laboratory for Biomicrotechnology, Department of Microsystems Engineering, University of Freiburg, Freiburg, Germany.,Bernstein Center Freiburg, University of Freiburg, Freiburg, Germany.,BrainLinks-BrainTools Cluster of Excellence, University of Freiburg, Freiburg, Germany
| | - Carola A Haas
- Experimental Epilepsy Research, Department of Neurosurgery, University Medical Center, University of Freiburg, Freiburg, Germany.,Bernstein Center Freiburg, University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany.,BrainLinks-BrainTools Cluster of Excellence, University of Freiburg, Freiburg, Germany
| |
Collapse
|
42
|
Carstens KE, Dudek SM. Regulation of synaptic plasticity in hippocampal area CA2. Curr Opin Neurobiol 2018; 54:194-199. [PMID: 30120016 DOI: 10.1016/j.conb.2018.07.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 07/26/2018] [Accepted: 07/31/2018] [Indexed: 11/15/2022]
Abstract
Synaptic plasticity in the hippocampus is thought to play a vital role in both the refinement of neuronal circuits during development and in learning in the mature brain. Synapses in hippocampal area CA1 are known for a robust capacity for long-term potentiation (LTP), whereas synapses in the stratum radiatum of hippocampal area CA2 are particularly resistant to such changes. Although we have yet to fully understand the mechanisms behind this resistance to plasticity, a number of genes and extracellular matrix components highly expressed in CA2 appear to function as molecular brakes on plasticity and develop postnatally in the rodent brain. Curiously, the developmental profile of several CA2-enriched molecules is suggestive of a still undefined critical window of plasticity in the hippocampus.
Collapse
Affiliation(s)
- Kelly E Carstens
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Serena M Dudek
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA.
| |
Collapse
|
43
|
Booker SA, Vida I. Morphological diversity and connectivity of hippocampal interneurons. Cell Tissue Res 2018; 373:619-641. [PMID: 30084021 PMCID: PMC6132631 DOI: 10.1007/s00441-018-2882-2] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 07/03/2018] [Indexed: 12/21/2022]
Abstract
The mammalian forebrain is constructed from ensembles of neurons that form local microcircuits giving rise to the exquisite cognitive tasks the mammalian brain can perform. Hippocampal neuronal circuits comprise populations of relatively homogenous excitatory neurons, principal cells and exceedingly heterogeneous inhibitory neurons, the interneurons. Interneurons release GABA from their axon terminals and are capable of controlling excitability in every cellular compartment of principal cells and interneurons alike; thus, they provide a brake on excess activity, control the timing of neuronal discharge and provide modulation of synaptic transmission. The dendritic and axonal morphology of interneurons, as well as their afferent and efferent connections within hippocampal circuits, is central to their ability to differentially control excitability, in a cell-type- and compartment-specific manner. This review aims to provide an up-to-date compendium of described hippocampal interneuron subtypes, with respect to their morphology, connectivity, neurochemistry and physiology, a full understanding of which will in time help to explain the rich diversity of neuronal function.
Collapse
Affiliation(s)
- Sam A Booker
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH8 9XD, UK.
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, EH8 9XD, UK.
| | - Imre Vida
- Institute for Integrative Neuroanatomy, Charité - Universitätmedizin Berlin, Berlin, Germany.
| |
Collapse
|
44
|
Soluble Aβ Oligomers Impair Dipolar Heterodendritic Plasticity by Activation of mGluR in the Hippocampal CA1 Region. iScience 2018; 6:138-150. [PMID: 30240608 PMCID: PMC6137707 DOI: 10.1016/j.isci.2018.07.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 07/05/2018] [Accepted: 07/19/2018] [Indexed: 11/20/2022] Open
Abstract
Soluble Aβ oligomers (oAβs) contribute importantly to synaptotoxicity in Alzheimer disease (AD), but the mechanisms related to heterogeneity of synaptic functions at local circuits remain elusive. Nearly all studies of the effects of oAβs on hippocampal synaptic plasticity have only examined homosynaptic plasticity. Here we stimulated the Schaffer collaterals and then simultaneously recorded in stratum radiatum (apical dendrites) and stratum oriens (basal dendrites) of CA1 neurons. We found that the apical dendrites are significantly more vulnerable to oAβ-mediated synaptic dysfunction: the heterosynaptic basal dendritic long-term potentiation (LTP) remained unchanged, whereas the homosynaptic apical LTP was impaired. However, the heterosynaptic basal dendritic plasticity induced by either spaced 10-Hz bursts or low-frequency (1-Hz) stimulation was disrupted by oAβs in a mGluR5-dependent manner. These results suggest that different firing patterns in the same neurons may be selectively altered by soluble oAβs in an early phase of AD, before frank neurodegeneration.
Collapse
|
45
|
You M, Dong J, Fu Y, Cong Z, Fu H, Wei L, Wang Y, Wang Y, Chen J. Exposure to Di-(2-ethylhexyl) Phthalate During Perinatal Period Gender-Specifically Impairs the Dendritic Growth of Pyramidal Neurons in Rat Offspring. Front Neurosci 2018; 12:444. [PMID: 30087586 PMCID: PMC6066609 DOI: 10.3389/fnins.2018.00444] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 06/12/2018] [Indexed: 01/09/2023] Open
Abstract
Di-(2-ethylhexyl) phthalate (DEHP), as a prevalent xenoestrogen endocrine disrupter, is omnipresent in the environment and commonly used in polyethylene plastic products. Although DEHP has potential adverse effects on multisystem organs, damage to the central nervous system is more significant. However, the consequences and mechanisms of DEHP exposure remain to be explored. The aim of this study was to investigate the effects and related mechanisms of maternal DEHP exposure on dendritic development of hippocampal pyramidal neurons in a rat model. Pregnant Wistar rats were intragastrically administrated either vehicle or DEHP (30, 300, and 750 mg/kg/d) from gestation day 0 to postnatal day (PN) 21. The dendritic length and complexity of dendritic arbors' pattern in pyramidal neurons of the hippocampus were measured using Golgi-Cox staining and Sholl analysis. The expression of dendritic development-related proteins was detected using western blot and immunofluorescence staining. DEHP-treated male but not female pups showed an obvious decrease in the total length and branching numbers of basal dendrites on PN7, PN14, and PN21. The phosphorylation of MAP2c, stathmin, and JNK1 in the male pup hippocampus was significantly decreased in DEHP treatment groups compared to controls. However, protein expression alteration in the hippocampus of female offspring was not observed. In summary, our study indicated that DEHP has a gender-specific negative impact on the dendritic growth of CA1 pyramidal neurons in male offspring of a rat model of DEHP exposure. The adverse impact may be related to the dysregulation of phosphorylated and total MAP2c and stathmin mediated by JNK1.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Jie Chen
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, China
| |
Collapse
|
46
|
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
|
47
|
Visualization of Altered Hippocampal Connectivity in an Animal Model of Alzheimer's Disease. Mol Neurobiol 2018; 55:7886-7899. [PMID: 29488134 PMCID: PMC6132739 DOI: 10.1007/s12035-018-0918-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 01/21/2018] [Indexed: 12/22/2022]
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disorder characterized by cognitive decline and neurodegeneration in the hippocampus. Despite the pathological importance of the hippocampal degeneration in AD, little topographical evidence exists of impaired hippocampal connectivity in patients with AD. To investigate the anatomical connections of the hippocampus, we injected the neurotracer 1,1′-dioctadecyl-3,3,3′3,3′-tetramethyl-indocarbocyanine perchlorate (DiI) into the hippocampi of 5XFAD mice, which were used as an animal model of AD. In wild-type controls, DiI-containing cells were found in the entorhinal cortex, medial septum, locus coeruleus, dorsal raphe, substantia nigra pars compacta, and olfactory bulb. Hippocampal inputs were decreased in multiple brain regions in the 5XFAD mice compared to wild-type littermate mice. These results are the first to reveal alterations at the cellular level in hippocampal connectivity in the brains of 5XFAD mice. These results suggest that anatomical mapping of hippocampal connectivity will elucidate new pathogenic mechanisms and therapeutic targets for AD treatment.
Collapse
|
48
|
Conditional Deletion of Hippocampal CA2/CA3a Oxytocin Receptors Impairs the Persistence of Long-Term Social Recognition Memory in Mice. J Neurosci 2017; 38:1218-1231. [PMID: 29279308 DOI: 10.1523/jneurosci.1896-17.2017] [Citation(s) in RCA: 115] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 11/17/2017] [Accepted: 12/18/2017] [Indexed: 11/21/2022] Open
Abstract
Oxytocin (OXT) receptors (OXTRs) are prominently expressed in hippocampal CA2 and CA3 pyramidal neurons, but little is known about its physiological function. As the functional necessity of hippocampal CA2 for social memory processing, we tested whether CA2 OXTRs may contribute to long-term social recognition memory (SRM) formation. Here, we found that conditional deletion of Oxtr from forebrain (Oxtr-/-) or CA2/CA3a-restricted excitatory neurons in adult male mice impaired the persistence of long-term SRM but had no effect on sociability and preference for social novelty. Conditional deletion of CA2/CA3a Oxtr showed no changes in anxiety-like behavior assessed using the open-field, elevated plus maze and novelty-suppressed feeding tests. Application of a highly selective OXTR agonist [Thr4,Gly7]-OXT to hippocampal slices resulted in an acute and lasting potentiation of excitatory synaptic responses in CA2 pyramidal neurons that relied on N-methyl-d-aspartate receptor activation and calcium/calmodulin-dependent protein kinase II activity. In addition, Oxtr-/- mice displayed a defect in the induction of long-term potentiation, but not long-term depression, at the synapses between the entorhinal cortex and CA2 pyramidal neurons. Furthermore, Oxtr deletion led to a reduced complexity of basal dendritic arbors of CA2 pyramidal neurons, but caused no alteration in the density of apical dendritic spines. Considering that the methodologies we have used to delete Oxtr do not rule out targeting the neighboring CA3a region, these findings suggest that OXTR signaling in the CA2/CA3a is crucial for the persistence of long-term SRM.SIGNIFICANCE STATEMENT Oxytocin receptors (OXTRs) are abundantly expressed in hippocampal CA2 and CA3 regions, but there are little known about their physiological function. Taking advantage of the conditional Oxtr knock-out mice, the present study highlights the importance of OXTR signaling in the induction of long-term potentiation at the synapses between the entorhinal cortex and CA2 pyramidal neurons and the persistence of long-term social recognition memory. Thus, OXTRs in the CA2/CA3a may provide a new target for therapeutic approaches to the treatment of social cognition deficits, which are often observed in patients with neuropsychiatric disorders.
Collapse
|
49
|
Interplay between global and pathway-specific synaptic plasticity in CA1 pyramidal cells. Sci Rep 2017; 7:17040. [PMID: 29213058 PMCID: PMC5719010 DOI: 10.1038/s41598-017-17161-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 11/22/2017] [Indexed: 12/22/2022] Open
Abstract
Mechanisms underlying information storage have been depicted for global cell-wide and pathway-specific synaptic plasticity. Yet, little is known how these forms of plasticity interact to enhance synaptic competition and network stability. We examined synaptic interactions between apical and basal dendrites of CA1 pyramidal neurons in mouse hippocampal slices. Bursts (50 Hz) of three action potentials (AP-bursts) paired with preceding presynaptic stimulation in stratum radiatum specifically led to LTP of the paired pathway in adult mice (P75). At adolescence (P28), an increase in burst frequency (>50 Hz) was required to gain timing-dependent LTP. Surprisingly, paired radiatum and unpaired oriens pathway potentiated, unless the pre-post delay was shortened from 10 to 5 ms, which selectively potentiated paired radiatum pathway, since unpaired oriens pathway decreased back to baseline. Conversely, the exact same 5 ms pairing in stratum oriens potentiated both pathways, as did AP-bursts alone, which potentiated synaptic efficacy as well as current-evoked postsynaptic spiking. L-type voltage-gated Ca2+ channels were involved in mediating synaptic potentiation in oriens, whereas NMDA and adenosine receptors counteracted unpaired stratum oriens potentiation following pairing in stratum radiatum. This asymmetric plasticity uncovers important insights into alterations of synaptic efficacy and intrinsic neuronal excitability for pathways that convey hippocampal and extra-hippocampal information.
Collapse
|
50
|
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: 137] [Impact Index Per Article: 17.1] [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
|