1
|
Martins YA, Cardinali CAEF, Torrão AS. Age-related differences in long-term memory performance and astrocyte morphology in rat hippocampus. Neurobiol Aging 2025; 150:19-43. [PMID: 40043468 DOI: 10.1016/j.neurobiolaging.2025.02.006] [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: 10/07/2024] [Revised: 02/10/2025] [Accepted: 02/24/2025] [Indexed: 04/10/2025]
Abstract
Astrocytes are neuromodulator cells. Their complex and dynamic morphology regulates neuronal signaling, synaptic plasticity, and neurogenesis. The impact of aging on astrocyte morphology is still under ongoing debate. Therefore, this study aimed to characterize astrocyte morphology in the hippocampus of older rats. 2-, 18-, and 20-month-old male Wistar rats were submitted to the object recognition test to assess their short- and long-term memories. CA1, CA2, CA3, and the dentate gyrus were collected for immunohistochemistry analysis and glial fibrillary acid protein (GFAP) immunostaining. Our results indicate that 20-month-old rats did not recognize or discriminate the novel object in the long-term memory test. Also, GFAP staining was greater in the oldest group for all analyzed areas. Morphometric and fractal analysis indicated shorter branch lengths and smaller sizes for astrocytes of 20-month-old rats. Overall, our results suggest that 20-month-old rats have long-term memory impairment, increased GFAP staining, and astrocyte dystrophy. These age-related alterations in astrocyte morphology are a resource for future studies exploring the role of astrocytes in age-related cognitive decline and age-related diseases.
Collapse
Affiliation(s)
- Yandara A Martins
- Departamento de Fisiologia e Biofisica, Universidade de São Paulo, Sao Paulo, Brazil.
| | | | - Andréa S Torrão
- Departamento de Fisiologia e Biofisica, Universidade de São Paulo, Sao Paulo, Brazil.
| |
Collapse
|
2
|
Sen M, Karamustafalioglu N, Celikkiran P, Ansen G, Sakul BU, Namlı MN, Yesilkaya UH. Altered Volumes of the Amygdala and Hippocampus in the Brain of Suicidal Patients with First Episode Schizophrenia. Psychiatr Q 2025:10.1007/s11126-025-10124-y. [PMID: 40000589 DOI: 10.1007/s11126-025-10124-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/13/2025] [Indexed: 02/27/2025]
Abstract
Suicide remains a significant cause of premature death in schizophrenia patients. Structural alterations in the brain and neurobiological mechanisms behind suicidal behavior (SB) in first-episode schizophrenia (FES) have received increasing attention. The amygdala and hippocampus regulate behaviors such as risk-taking, impulsivity, and emotional processing. Abnormalities in these regions have been linked with suicidal ideation, behavior, and psychotic symptoms. However, the association remains unclear. The study included 20 FES patients with current SB, 51 FES patients without SB, and 42 healthy controls. All patients were subjected to clinical evaluations to assess psychotic symptoms and suicidal ideation and behavior, both current and lifetime. T1-weighted magnetic resonance imaging scans were processed through two web-based automatic analysis tools, MRICloud, and volBrain. Bilateral amygdala volumes were found to be significantly lower in the patient groups, while schizophrenia and suicidal ideation had opposite effects on amygdala volumes. Hippocampal subfields such as the right Cornu Amnonis (CA) fields varied according to the clinical status of the patients, including the severity of suicidal ideation and behavior. These findings support not only the involvement of the amygdala and hippocampus in SB and schizophrenia but also their roles in the discrimination of SB in patients with schizophrenia.
Collapse
Affiliation(s)
- Meltem Sen
- Department of Psychiatry, Bakirkoy Prof Mazhar Osman Training and Research Hospital for Psychiatry, Neurology, and Neurosurgery, Istanbul, Turkey.
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, USA.
| | - Nesrin Karamustafalioglu
- Department of Psychiatry, Bakirkoy Prof Mazhar Osman Training and Research Hospital for Psychiatry, Neurology, and Neurosurgery, Istanbul, Turkey
| | - Pinar Celikkiran
- Department of Psychiatry, Bakirkoy Prof Mazhar Osman Training and Research Hospital for Psychiatry, Neurology, and Neurosurgery, Istanbul, Turkey
| | - Gamze Ansen
- Department of Anatomy, Medipol University Medical Faculty, Istanbul, Turkey
| | - Bayram Ufuk Sakul
- Department of Anatomy, Medipol University Medical Faculty, Istanbul, Turkey
| | - Mustafa Nuray Namlı
- Department of Psychiatry, Bakirkoy Prof Mazhar Osman Training and Research Hospital for Psychiatry, Neurology, and Neurosurgery, Istanbul, Turkey
| | - Umit Haluk Yesilkaya
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
3
|
Yoshida K, Forrest SL, Ichimata S, Tanaka H, Kon T, Kovacs GG. Co-pathologies modify hippocampal protein accumulation patterns in neurodegenerative diseases. Alzheimers Dement 2025; 21:e14355. [PMID: 39711489 PMCID: PMC11782183 DOI: 10.1002/alz.14355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 08/23/2024] [Accepted: 09/17/2024] [Indexed: 12/24/2024]
Abstract
INTRODUCTION Limited research has extensively analyzed neurodegenerative disease-related protein deposition patterns in the hippocampus. METHODS This study examined the distribution of proteins in hippocampal subregions across major neurodegenerative diseases and explored their relation to each other. The area density of phosphorylated tau (p-tau), amyloid beta (Aβ), α-synuclein, and phosphorylated TDP-43 protein deposits together with pyramidal cell density in each hippocampal subregion, including CA1-4, prosubiculum (ProS), and subiculum was assessed in 166 cases encompassing various neurodegenerative diseases. RESULTS Alzheimer's disease-associated p-tau predominated in ProS, Aβ in the CA1, and Lewy body-related α-synuclein in the CA2. The area density of protein deposits increased with the pathological stage until a peak, then decreased in cases with high pathology stages along with pyramidal cell density. Comorbid protein pathology influenced protein deposition patterns. DISCUSSION This comprehensive evaluation reveals characteristic neurodegenerative disease-related protein accumulation patterns in hippocampal subregions modified by co-pathologies. HIGHLIGHTS Alzheimer's disease-related phosphorylated tau predominates in the prosubiculum. Amyloid beta predominates in the CA1 and Lewy body-related α-synuclein in the CA2. The area density of protein deposition increases with the disease stage up to a peak. In the high pathology stage, protein deposition and pyramidal cell density decreases. Comorbid protein pathology affects the pattern of protein accumulation.
Collapse
Affiliation(s)
- Koji Yoshida
- Department of Laboratory Medicine and Pathobiology and Department of MedicineUniversity of TorontoTorontoOntarioCanada
- Tanz Centre for Research in Neurodegenerative DiseaseKrembil Discovery TowerUniversity of TorontoTorontoOntarioCanada
- Department of Legal MedicineGraduate School of Medicine and Pharmaceutical SciencesUniversity of ToyamaToyamaJapan
| | - Shelley L. Forrest
- Department of Laboratory Medicine and Pathobiology and Department of MedicineUniversity of TorontoTorontoOntarioCanada
- Tanz Centre for Research in Neurodegenerative DiseaseKrembil Discovery TowerUniversity of TorontoTorontoOntarioCanada
- Laboratory Medicine Program & Krembil Brain InstituteUniversity Health NetworkTorontoOntarioCanada
- Department of NeurologyDementia Research CentreMacquarie Medical SchoolFaculty of MedicineHealth and Human SciencesMacquarie UniversitySydneyAustralia
| | - Shojiro Ichimata
- Department of Laboratory Medicine and Pathobiology and Department of MedicineUniversity of TorontoTorontoOntarioCanada
- Tanz Centre for Research in Neurodegenerative DiseaseKrembil Discovery TowerUniversity of TorontoTorontoOntarioCanada
- Department of Legal MedicineGraduate School of Medicine and Pharmaceutical SciencesUniversity of ToyamaToyamaJapan
| | - Hidetomo Tanaka
- Department of Laboratory Medicine and Pathobiology and Department of MedicineUniversity of TorontoTorontoOntarioCanada
- Tanz Centre for Research in Neurodegenerative DiseaseKrembil Discovery TowerUniversity of TorontoTorontoOntarioCanada
| | - Tomoya Kon
- Department of Laboratory Medicine and Pathobiology and Department of MedicineUniversity of TorontoTorontoOntarioCanada
- Tanz Centre for Research in Neurodegenerative DiseaseKrembil Discovery TowerUniversity of TorontoTorontoOntarioCanada
- Graduate School of MedicineHirosaki UniversityHirosakiJapan
| | - Gabor G. Kovacs
- Department of Laboratory Medicine and Pathobiology and Department of MedicineUniversity of TorontoTorontoOntarioCanada
- Tanz Centre for Research in Neurodegenerative DiseaseKrembil Discovery TowerUniversity of TorontoTorontoOntarioCanada
- Laboratory Medicine Program & Krembil Brain InstituteUniversity Health NetworkTorontoOntarioCanada
- Department of NeurologyDementia Research CentreMacquarie Medical SchoolFaculty of MedicineHealth and Human SciencesMacquarie UniversitySydneyAustralia
| |
Collapse
|
4
|
Shivakumar AB, Mehak SF, Jijimon F, Gangadharan G. Extrahippocampal Contributions to Social Memory: The Role of Septal Nuclei. Biol Psychiatry 2024; 96:835-847. [PMID: 38718881 DOI: 10.1016/j.biopsych.2024.04.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 03/22/2024] [Accepted: 04/22/2024] [Indexed: 06/16/2024]
Abstract
Social memory, the ability to recognize and remember individuals within a social group, is crucial for social interactions and relationships. Deficits in social memory have been linked to several neuropsychiatric and neurodegenerative disorders. The hippocampus, especially the circuit that links dorsal CA2 and ventral CA1 neurons, is considered a neural substrate for social memory formation. Recent studies have provided compelling evidence of extrahippocampal contributions to social memory. The septal nuclei, including the medial and lateral septum, make up a basal forebrain region that shares bidirectional neuronal connections with the hippocampus and has recently been identified as critical for social memory. The focus of our review is the neural circuit mechanisms that underlie social memory, with a special emphasis on the septum. We also discuss the social memory dysfunction associated with neuropsychiatric and neurodegenerative disorders.
Collapse
Affiliation(s)
- Apoorva Bettagere Shivakumar
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Sonam Fathima Mehak
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Feyba Jijimon
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Gireesh Gangadharan
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India.
| |
Collapse
|
5
|
Barrera-Conde M, Ramon-Duaso C, González-Parra JA, Veza-Estevez E, Chevaleyre V, Piskorowski RA, de la Torre R, Busquets-García A, Robledo P. Adolescent cannabinoid exposure rescues phencyclidine-induced social deficits through modulation of CA2 transmission. Prog Neurobiol 2024; 240:102652. [PMID: 38955325 DOI: 10.1016/j.pneurobio.2024.102652] [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: 03/07/2024] [Revised: 06/18/2024] [Accepted: 06/24/2024] [Indexed: 07/04/2024]
Abstract
Psychotic disorders entail intricate conditions marked by disruptions in cognition, perception, emotions, and social behavior. Notably, psychotic patients who use cannabis tend to show less severe deficits in social behaviors, such as the misinterpretation of social cues and the inability to interact with others. However, the biological underpinnings of this epidemiological interaction remain unclear. Here, we used the NMDA receptor blocker phencyclidine (PCP) to induce psychotic-like states and to study the impact of adolescent cannabinoid exposure on social behavior deficits and synaptic transmission changes in hippocampal area CA2, a region known to be active during social interactions. In particular, adolescent mice underwent 7 days of subchronic treatment with the synthetic cannabinoid, WIN 55, 212-2 (WIN) followed by one injection of PCP. Using behavioral, biochemical, and electrophysiological approaches, we showed that PCP persistently reduced sociability, decreased GAD67 expression in the hippocampus, and induced GABAergic deficits in proximal inputs from CA3 and distal inputs from the entorhinal cortex (EC) to CA2. Notably, WIN exposure during adolescence specifically restores adult sociability deficits, the expression changes in GAD67, and the GABAergic impairments in the EC-CA2 circuit, but not in the CA3-CA2 circuit. Using a chemogenetic approach to target EC-CA2 projections, we demonstrated the involvement of this specific circuit on sociability deficits. Indeed, enhancing EC-CA2 transmission was sufficient to induce sociability deficits in vehicle-treated mice, but not in animals treated with WIN during adolescence, suggesting a mechanism by which adolescent cannabinoid exposure rescues sociability deficits caused by enhanced EC-CA2 activity in adult mice.
Collapse
Affiliation(s)
- Marta Barrera-Conde
- Integrative Pharmacology and Systems Neuroscience, Neuroscience Research Program, Hospital del Mar Research Institute, Barcelona, Spain; Department of Medicine and Life Sciences, University Pompeu Fabra, Barcelona, Spain
| | - Carla Ramon-Duaso
- Cell-Type Mechanisms in Normal and Pathological Behavior Research Group, Neuroscience Programme, Hospital del Mar Research Institute, Barcelona, Spain
| | - Jose Antonio González-Parra
- Cell-Type Mechanisms in Normal and Pathological Behavior Research Group, Neuroscience Programme, Hospital del Mar Research Institute, Barcelona, Spain
| | - Emma Veza-Estevez
- Integrative Pharmacology and Systems Neuroscience, Neuroscience Research Program, Hospital del Mar Research Institute, Barcelona, Spain
| | - Vivien Chevaleyre
- Université Paris Cité, INSERM U1266, Institute of Psychiatry and Neuroscience of Paris, GHU Paris Psychiatry and Neuroscience, Paris, France
| | - Rebecca A Piskorowski
- Université Paris Cité, INSERM U1266, Institute of Psychiatry and Neuroscience of Paris, GHU Paris Psychiatry and Neuroscience, Paris, France
| | - Rafael de la Torre
- Integrative Pharmacology and Systems Neuroscience, Neuroscience Research Program, Hospital del Mar Research Institute, Barcelona, Spain; Department of Medicine and Life Sciences, University Pompeu Fabra, Barcelona, Spain; Centro de Investigación Biomédica en Red - Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Arnau Busquets-García
- Cell-Type Mechanisms in Normal and Pathological Behavior Research Group, Neuroscience Programme, Hospital del Mar Research Institute, Barcelona, Spain.
| | - Patricia Robledo
- Integrative Pharmacology and Systems Neuroscience, Neuroscience Research Program, Hospital del Mar Research Institute, Barcelona, Spain.
| |
Collapse
|
6
|
Severino L, Kim J, Nam MH, McHugh TJ. From synapses to circuits: What mouse models have taught us about how autism spectrum disorder impacts hippocampal function. Neurosci Biobehav Rev 2024; 158:105559. [PMID: 38246230 DOI: 10.1016/j.neubiorev.2024.105559] [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: 11/29/2023] [Revised: 01/16/2024] [Accepted: 01/17/2024] [Indexed: 01/23/2024]
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder that impacts a variety of cognitive and behavioral domains. While a genetic component of ASD has been well-established, none of the numerous syndromic genes identified in humans accounts for more than 1% of the clinical patients. Due to this large number of target genes, numerous mouse models of the disorder have been generated. However, the focus on distinct brain circuits, behavioral phenotypes and diverse experimental approaches has made it difficult to synthesize the overwhelming number of model animal studies into concrete throughlines that connect the data across levels of investigation. Here we chose to focus on one circuit, the hippocampus, and one hypothesis, a shift in excitatory/inhibitory balance, to examine, from the level of the tripartite synapse up to the level of in vivo circuit activity, the key commonalities across disparate models that can illustrate a path towards a better mechanistic understanding of ASD's impact on hippocampal circuit function.
Collapse
Affiliation(s)
- Leandra Severino
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, South Korea; Division of Bio-Medical Science & Technology, KIST-School, University of Science and Technology, Seoul, South Korea
| | - Jinhyun Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, South Korea; Division of Bio-Medical Science & Technology, KIST-School, University of Science and Technology, Seoul, South Korea
| | - Min-Ho Nam
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, South Korea; Division of Bio-Medical Science & Technology, KIST-School, University of Science and Technology, Seoul, South Korea.
| | - Thomas J McHugh
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, South Korea; Laboratory for Circuit and Behavioral Physiology, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako-shi Saitama, Japan.
| |
Collapse
|
7
|
Garamszegi SP, Brzostowicki DJ, Coyne TM, Vontell RT, Davis DA. TDP-43 and Alzheimer's Disease Pathology in the Brain of a Harbor Porpoise Exposed to the Cyanobacterial Toxin BMAA. Toxins (Basel) 2024; 16:42. [PMID: 38251257 PMCID: PMC10821503 DOI: 10.3390/toxins16010042] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 12/30/2023] [Accepted: 01/10/2024] [Indexed: 01/23/2024] Open
Abstract
Cetaceans are well-regarded as sentinels for toxin exposure. Emerging studies suggest that cetaceans can also develop neuropathological changes associated with neurodegenerative disease. The occurrence of neuropathology makes cetaceans an ideal species for examining the impact of marine toxins on the brain across the lifespan. Here, we describe TAR DNA-binding protein 43 (TDP-43) proteinopathy and Alzheimer's disease (AD) neuropathological changes in a beached harbor porpoise (Phocoena phocoena) that was exposed to a toxin produced by cyanobacteria called β-N-methylamino-L-alanine (BMAA). We found pathogenic TDP-43 cytoplasmic inclusions in neurons throughout the cerebral cortex, midbrain and brainstem. P62/sequestosome-1, responsible for the autophagy of misfolded proteins, was observed in the amygdala, hippocampus and frontal cortex. Genes implicated in AD and TDP-43 neuropathology such as APP and TARDBP were expressed in the brain. AD neuropathological changes such as amyloid-β plaques, neurofibrillary tangles, granulovacuolar degeneration and Hirano bodies were present in the hippocampus. These findings further support the development of progressive neurodegenerative disease in cetaceans and a potential causative link to cyanobacterial toxins. Climate change, nutrient pollution and industrial waste are increasing the frequency of harmful cyanobacterial blooms. Cyanotoxins like BMAA that are associated with neurodegenerative disease pose an increasing public health risk.
Collapse
Affiliation(s)
- Susanna P. Garamszegi
- Department of Neurology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Daniel J. Brzostowicki
- Department of Neurology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Thomas M. Coyne
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Regina T. Vontell
- Department of Neurology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - David A. Davis
- Department of Neurology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| |
Collapse
|
8
|
Oakley RH, Riddick NV, Moy SS, Cidlowski JA. Imbalanced glucocorticoid and mineralocorticoid stress hormone receptor function has sex-dependent and independent regulatory effects in the mouse hippocampus. Neurobiol Stress 2024; 28:100589. [PMID: 38075021 PMCID: PMC10709088 DOI: 10.1016/j.ynstr.2023.100589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 10/25/2023] [Accepted: 11/12/2023] [Indexed: 02/12/2024] Open
Abstract
Many stress-related neuropsychiatric disorders display pronounced sex differences in their frequency and clinical symptoms. Glucocorticoids are primary stress hormones that have been implicated in the development of these disorders but whether they contribute to the observed sex bias is poorly understood. Glucocorticoids signal through two closely related nuclear receptors, the glucocorticoid (GR) and mineralocorticoid receptor (MR). To elucidate the sex-specific and independent actions of glucocorticoids in the hippocampus, we developed knockout mice lacking hippocampal GR, MR, or both GR and MR. Mice deficient in hippocampal MR or both GR and MR showed an altered molecular phenotype of CA2 neurons and reduced anxiety-like behavior in both sexes, but altered stress adaptation behavior only in females and enhanced fear-motivated cue learning only in males. All three knockout mouse models displayed reduced sociability but only in male mice. Male and female mice deficient in both hippocampal GR and MR exhibited extensive neurodegeneration in the dentate gyrus. Global transcriptomic analysis revealed a marked expansion in the number of dysregulated genes in the hippocampus of female knockout mice compared to their male counterparts; however, the overall patterns of gene dysregulation were remarkably similar in both sexes. Within and across sex comparisons identified key GR and MR target genes and associated signaling pathways underlying the knockout phenotypes. These findings define major sex-dependent and independent effects of GR/MR imbalances on gene expression and functional profiles in the hippocampus and inform new strategies for treating men and women with stress-related neuropsychiatric disorders.
Collapse
Affiliation(s)
- Robert H. Oakley
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, 27709, USA
| | - Natallia V. Riddick
- Department of Psychiatry and Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Sheryl S. Moy
- Department of Psychiatry and Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - John A. Cidlowski
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, 27709, USA
| |
Collapse
|
9
|
Ossani GP, Riudavets MA, D'Annunzio V, Uceda AM, Ponzo O, Lago NR, Martino DJ. Effect of lithium in pyramidal neurons of Cornu Ammonis in an animal model. J Psychiatr Res 2023; 167:33-36. [PMID: 37826875 DOI: 10.1016/j.jpsychires.2023.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 07/05/2023] [Accepted: 10/04/2023] [Indexed: 10/14/2023]
Abstract
Bipolar disorder has been associated with a decrease in hippocampal size, and lithium appears to reverse this neuroanatomical abnormality. The objective of this work was to evaluate, at a cellular level, the size of both cell body and nucleus of pyramidal neurons located throughout the Cornu Ammonis (CA1 to CA4 regions). To perform this duty, we used 16 rats that were randomized into two groups: control and dietary lithium-treated. After one month, they were sacrificed and their brains removed for histopathological analysis. Serial photos of the entire Cornu Ammonis were taken and, after dividing them into 4 regions of interest, we measured the cell body and nucleus on each pyramidal neuron belonging to the first 5 photos of each region of interest. As a result of this histological analysis, cell body area and nuclear area were significantly larger in the experimental group in a specific area of the Cornu Ammonis that could correspond to CA2 or the transition between CA1 and CA2. These results suggest that the effect of lithium is not homogeneous throughout the hippocampus and allows directing future studies to a specific area of this structure.
Collapse
Affiliation(s)
- G P Ossani
- Centre of Experimental and Applied Pathology, Department of Pathology, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina; Laboratory of Experimental Medicine, Hospital Alemán, Buenos Aires, Argentina; National Council of Scientific and Technical Research (CONICET), Argentina.
| | - M A Riudavets
- Office of the Chief Medical Examiner, Supreme Court of Justice. Buenos Aires, Argentina
| | - V D'Annunzio
- National Council of Scientific and Technical Research (CONICET), Argentina; Institute of Cardiovascular Physiopathology, Department of Pathology and Institute of Biochemistry and Molecular Medicine (IBIMOL UBA-CONICET), School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - A M Uceda
- Centre of Experimental and Applied Pathology, Department of Pathology, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina; Laboratory of Experimental Medicine, Hospital Alemán, Buenos Aires, Argentina
| | - O Ponzo
- Department of Physiology, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - N R Lago
- Centre of Experimental and Applied Pathology, Department of Pathology, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - D J Martino
- National Council of Scientific and Technical Research (CONICET), Argentina; Institute of Cognitive and Translational Neuroscience (INCyT), INECO Foundation, Favaloro University, Buenos Aires, Argentina
| |
Collapse
|
10
|
Hassan SI, Bigler S, Siegelbaum SA. Social odor discrimination and its enhancement by associative learning in the hippocampal CA2 region. Neuron 2023; 111:2232-2246.e5. [PMID: 37192623 PMCID: PMC10524117 DOI: 10.1016/j.neuron.2023.04.026] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/25/2022] [Accepted: 04/21/2023] [Indexed: 05/18/2023]
Abstract
Although the hippocampus is crucial for social memory, how social sensory information is combined with contextual information to form episodic social memories remains unknown. Here, we investigated the mechanisms for social sensory information processing using two-photon calcium imaging from hippocampal CA2 pyramidal neurons (PNs)-which are crucial for social memory-in awake head-fixed mice exposed to social and non-social odors. We found that CA2 PNs represent social odors of individual conspecifics and that these representations are refined during associative social odor-reward learning to enhance the discrimination of rewarded compared with unrewarded odors. Moreover, the structure of the CA2 PN population activity enables CA2 to generalize along categories of rewarded versus unrewarded and social versus non-social odor stimuli. Finally, we found that CA2 is important for learning social but not non-social odor-reward associations. These properties of CA2 odor representations provide a likely substrate for the encoding of episodic social memory.
Collapse
Affiliation(s)
- Sami I Hassan
- Department of Neuroscience, Mortimer B. Zuckerman Mind Brain Behavior Institute, The Kavli Institute for Brain Science, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10027, USA.
| | - Shivani Bigler
- Department of Neuroscience, Mortimer B. Zuckerman Mind Brain Behavior Institute, The Kavli Institute for Brain Science, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10027, USA
| | - Steven A Siegelbaum
- Department of Neuroscience, Mortimer B. Zuckerman Mind Brain Behavior Institute, The Kavli Institute for Brain Science, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10027, USA.
| |
Collapse
|
11
|
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
|
12
|
Engel K, Lee HN, Tewari BP, Lewkowicz AP, Ireland DDC, Manangeeswaran M, Verthelyi D. Neonatal Zika virus infection causes transient perineuronal net degradation. Front Cell Neurosci 2023; 17:1187425. [PMID: 37496706 PMCID: PMC10366369 DOI: 10.3389/fncel.2023.1187425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 06/21/2023] [Indexed: 07/28/2023] Open
Abstract
Perineuronal nets (PNNs) form a specialized extracellular matrix that predominantly surrounds parvalbumin (PV)-expressing GABAergic inhibitory interneurons and help regulate neuronal activity. Their formation early in the postnatal period is regulated by neuronal signaling and glial activation raising concerns that part of the long-term effects ascribed to perinatal viral infections could be mediated by altered PNN formation. Previously, we developed a model of neonatal Zika virus (ZIKV) infection where mice have lifelong neurological sequelae that includes motor disfunction and reduced anxiety coupled with a persistent low-grade expression in proinflammatory markers despite resolving the acute infection. Here, we demonstrate that ZIKV infection to P1 neonatal mice results in a reduction of PNN formation during the acute disease with significant reduction in Wisteria floribunda agglutinin (WFA) staining at the peak of infection [15 days post infection (dpi)] that persisted after the symptoms resolved (30 dpi). At 60 dpi, when there is residual inflammation in the CNS, the number of WFA+ cells and the level of WFA staining as well as levels of aggrecan and brevican in the brains of convalescent mice were not different from those in uninfected controls, however, there was increased frequency of PNNs with an immature phenotype. Over time the impact of the perinatal infection became less evident and there were no clear differences in PNN morphology between the groups at 1 year post infection. Of note, the reduction in PNNs during acute ZIKV infection was not associated with decreased mRNA levels of aggrecan or brevican, but increased levels of degraded aggrecan and brevican indicating increased PNN degradation. These changes were associated with increased expression of matrix metalloproteinase 12 (MMP12) and MMP19, but not MMP9, a disintegrin and metalloproteinase with thrombospondin motifs 4 (ADAMTS4) or ADAMTS5. Together our findings indicate that infection at the time of PNN development interferes with PNN formation, but the nets can reform once the infection and inflammation subside.
Collapse
Affiliation(s)
- Kaliroi Engel
- Laboratory of Immunology, Center of Excellence in Infectious Disease and Inflammation, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | - Ha-Na Lee
- Laboratory of Immunology, Center of Excellence in Infectious Disease and Inflammation, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | - Bhanu P. Tewari
- Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA, United States
| | - Aaron P. Lewkowicz
- Laboratory of Immunology, Center of Excellence in Infectious Disease and Inflammation, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | - Derek D. C. Ireland
- Laboratory of Immunology, Center of Excellence in Infectious Disease and Inflammation, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | - Mohanraj Manangeeswaran
- Laboratory of Immunology, Center of Excellence in Infectious Disease and Inflammation, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | - Daniela Verthelyi
- Laboratory of Immunology, Center of Excellence in Infectious Disease and Inflammation, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| |
Collapse
|
13
|
Walker JM, Goette W, Farrell K, Iida MA, Karlovich E, White CL, Crary JF, Richardson TE. The relationship between hippocampal amyloid beta burden and spatial distribution of neurofibrillary degeneration. Alzheimers Dement 2023; 19:3158-3170. [PMID: 36738450 PMCID: PMC11100308 DOI: 10.1002/alz.12966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/30/2022] [Accepted: 01/04/2023] [Indexed: 02/05/2023]
Abstract
INTRODUCTION Neurofibrillary degeneration in Alzheimer's disease (AD) typically involves the entorhinal cortex and CA1 subregion of the hippocampus early in the disease process, whereas in primary age-related tauopathy (PART), there is an early selective vulnerability of the CA2 subregion. METHODS Image analysis-based quantitative pixel assessments were used to objectively evaluate amyloid beta (Aβ) burden in the medial temporal lobe in relation to the distribution of hyperphosphorylated-tau (p-tau) in 142 cases of PART and AD. RESULTS Entorhinal, CA1, CA3, and CA4 p-tau deposition levels are significantly correlated with Aβ burden, while CA2 p-tau is not. Furthermore, the CA2/CA1 p-tau ratio is inversely correlated with Aβ burden and distribution. In addition, cognitive impairment is correlated with overall p-tau burden. DISCUSSION These data indicate that the presence and extent of medial temporal lobe Aβ may determine the distribution and spread of neurofibrillary degeneration. The resulting p-tau distribution patterns may discriminate between PART and AD. HIGHLIGHTS Subregional hyperphosphorylated-tau (p-tau) distribution is influenced by hippocampal amyloid beta burden. Higher CA2/CA1 p-tau ratio is predictive of primary age-related tauopathy-like neuropathology. Cognitive function is correlated with the overall hippocampal p-tau burden.
Collapse
Affiliation(s)
- Jamie M. Walker
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Neuropathology Brain Bank & Research CoRE, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - William Goette
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Kurt Farrell
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Neuropathology Brain Bank & Research CoRE, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Artificial Intelligence & Human Health, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Megan A. Iida
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Neuropathology Brain Bank & Research CoRE, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- School of Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Esma Karlovich
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Neuropathology Brain Bank & Research CoRE, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Pathology and Cell Biology, Columbia University, New York, New York, USA
| | - The PART Working Group
- The PART working group is a multi-institutional collaboration. PART working group investigators are listed in the acknowledgments section
| | - Charles L. White
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - John F. Crary
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Neuropathology Brain Bank & Research CoRE, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Artificial Intelligence & Human Health, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Timothy E. Richardson
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Neuropathology Brain Bank & Research CoRE, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
14
|
Huang H, Joffrin AM, Zhao Y, Miller GM, Zhang GC, Oka Y, Hsieh-Wilson LC. Chondroitin 4- O-sulfation regulates hippocampal perineuronal nets and social memory. Proc Natl Acad Sci U S A 2023; 120:e2301312120. [PMID: 37279269 PMCID: PMC10268298 DOI: 10.1073/pnas.2301312120] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 04/26/2023] [Indexed: 06/08/2023] Open
Abstract
Glycan alterations are associated with aging, neuropsychiatric, and neurodegenerative diseases, although the contributions of specific glycan structures to emotion and cognitive functions remain largely unknown. Here, we used a combination of chemistry and neurobiology to show that 4-O-sulfated chondroitin sulfate (CS) polysaccharides are critical regulators of perineuronal nets (PNNs) and synapse development in the mouse hippocampus, thereby affecting anxiety and cognitive abilities such as social memory. Brain-specific deletion of CS 4-O-sulfation in mice increased PNN densities in the area CA2 (cornu ammonis 2), leading to imbalanced excitatory-to-inhibitory synaptic ratios, reduced CREB activation, elevated anxiety, and social memory dysfunction. The impairments in PNN densities, CREB activity, and social memory were recapitulated by selective ablation of CS 4-O-sulfation in the CA2 region during adulthood. Notably, enzymatic pruning of the excess PNNs reduced anxiety levels and restored social memory, while chemical manipulation of CS 4-O-sulfation levels reversibly modulated PNN densities surrounding hippocampal neurons and the balance of excitatory and inhibitory synapses. These findings reveal key roles for CS 4-O-sulfation in adult brain plasticity, social memory, and anxiety regulation, and they suggest that targeting CS 4-O-sulfation may represent a strategy to address neuropsychiatric and neurodegenerative diseases associated with social cognitive dysfunction.
Collapse
Affiliation(s)
- Huiqian Huang
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA91125
- Clinical Research Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou310000, China
| | - Amélie M. Joffrin
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA91125
| | - Yuan Zhao
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA91125
| | - Gregory M. Miller
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA91125
| | - Grace C. Zhang
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA91125
| | - Yuki Oka
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA91125
| | - Linda C. Hsieh-Wilson
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA91125
| |
Collapse
|
15
|
Oliva A, Fernandez-Ruiz A, Karaba LA. CA2 orchestrates hippocampal network dynamics. Hippocampus 2023; 33:241-251. [PMID: 36575880 PMCID: PMC9974898 DOI: 10.1002/hipo.23495] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 11/25/2022] [Accepted: 12/11/2022] [Indexed: 12/29/2022]
Abstract
The hippocampus is composed of various subregions: CA1, CA2, CA3, and the dentate gyrus (DG). Despite the abundant hippocampal research literature, until recently, CA2 received little attention. The development of new genetic and physiological tools allowed recent studies characterizing the unique properties and functional roles of this hippocampal subregion. Despite its small size, the cellular content of CA2 is heterogeneous at the molecular and physiological levels. CA2 has been heavily implicated in social behaviors, including social memory. More generally, the mechanisms by which the hippocampus is involved in memory include the reactivation of neuronal ensembles following experience. This process is coordinated by synchronous network events known as sharp-wave ripples (SWRs). Recent evidence suggests that CA2 plays an important role in the generation of SWRs. The unique connectivity and physiological properties of CA2 pyramidal cells make this region a computational hub at the core of hippocampal information processing. Here, we review recent findings that support the role of CA2 in coordinating hippocampal network dynamics from a systems neuroscience perspective.
Collapse
Affiliation(s)
- Azahara Oliva
- Department of Neurobiology and Behavior, Cornell University, Ithaca, New York, USA
| | | | - Lindsay A Karaba
- Department of Neurobiology and Behavior, Cornell University, Ithaca, New York, USA
| |
Collapse
|
16
|
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
|
17
|
Bienkowski MS. Further refining the boundaries of the hippocampus CA2 with gene expression and connectivity: Potential subregions and heterogeneous cell types. Hippocampus 2023; 33:150-160. [PMID: 36786207 PMCID: PMC9987718 DOI: 10.1002/hipo.23508] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 01/20/2023] [Accepted: 01/23/2023] [Indexed: 02/15/2023]
Abstract
Over the last two decades, the definition of hippocampal area CA2 has evolved from Lorente de Nó's original Golgi-based morphological description with the discovery of specific CA2 gene expression markers. Exploiting the specificity of these molecules has allowed for the genetic dissection of CA2 structure and function in transgenic mice. With this change in criteria, the anatomical boundaries of the CA2 have expanded across the hippocampal axis but the CA2's full rostrocaudal extent is not consistently delineated across atlases. The Hippocampus Gene Expression Atlas (HGEA) provides a comprehensive map of 20 gene expression domains across the entire mouse hippocampus including the CA2. In this commentary, I will review the consensus gene expression patterns that demarcate the expanded CA2 boundaries in the HGEA. Using DropViz single-cell transcriptomics and Mouse Connectome Project connectomics data, I will then suggest potential differences in CA2 cell type heterogeneity and connectivity that may identify and characterize further CA2 subregions.
Collapse
Affiliation(s)
- Michael S Bienkowski
- USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, California, USA
- Zilkha Neurogenetic Institute, Keck School of Medicine of University of Southern California, Los Angeles, California, USA
- USC Center for Integrative Connectomics, Keck School of Medicine of University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
18
|
Piskorowski RA, Chevaleyre V. Hippocampal area CA2: interneuron disfunction during pathological states. Front Neural Circuits 2023; 17:1181032. [PMID: 37180763 PMCID: PMC10174260 DOI: 10.3389/fncir.2023.1181032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 04/07/2023] [Indexed: 05/16/2023] Open
Abstract
Hippocampal area CA2 plays a critical role in social recognition memory and has unique cellular and molecular properties that distinguish it from areas CA1 and CA3. In addition to having a particularly high density of interneurons, the inhibitory transmission in this region displays two distinct forms of long-term synaptic plasticity. Early studies on human hippocampal tissue have reported unique alteration in area CA2 with several pathologies and psychiatric disorders. In this review, we present recent studies revealing changes in inhibitory transmission and plasticity of area CA2 in mouse models of multiple sclerosis, autism spectrum disorder, Alzheimer's disease, schizophrenia and the 22q11.2 deletion syndrome and propose how these changes could underly deficits in social cognition observed during these pathologies.
Collapse
Affiliation(s)
- Rebecca A. Piskorowski
- Université Paris Cité, INSERM UMRS 1266, Institute of Psychiatry and Neuroscience of Paris, GHU Paris Psychiatrie et Neurosciences, Paris, France
- Institute of Biology Paris Seine, Neuroscience Paris Seine, CNRS UMR 8246, INSERM U1130, Sorbonne Université, Paris, France
- *Correspondence: Rebecca A. Piskorowski,
| | - Vivien Chevaleyre
- Université Paris Cité, INSERM UMRS 1266, Institute of Psychiatry and Neuroscience of Paris, GHU Paris Psychiatrie et Neurosciences, Paris, France
- Institute of Biology Paris Seine, Neuroscience Paris Seine, CNRS UMR 8246, INSERM U1130, Sorbonne Université, Paris, France
| |
Collapse
|
19
|
Oliva A. CA2 physiology underlying social memory. Curr Opin Neurobiol 2022; 77:102642. [PMID: 36215845 DOI: 10.1016/j.conb.2022.102642] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 08/23/2022] [Accepted: 09/11/2022] [Indexed: 01/10/2023]
Abstract
In recent years, convergent evidence has emerged in support of the idea of social brain networks, specific brain regions that are interconnected and support social behaviors. One of these regions is the CA2 area of the hippocampus, a small region strongly connected with cortical and subcortical areas implicated in social behaviors. Furthermore, CA2 area is enriched in receptors for several neuromodulators that are related to various aspects of social behaviors, suggesting that this area could be a key component of social information processing in the brain. In this review, recent findings related to the physiological mechanisms underlying the role of CA2 in social memory are discussed.
Collapse
Affiliation(s)
- Azahara Oliva
- Department of Neurobiology and Behavior, Cornell University, Ithaca, NY, 14853, USA.
| |
Collapse
|
20
|
The 18 kDa translocator protein is associated with microglia in the hippocampus of non-demented elderly subjects. AGING BRAIN 2022; 2:100045. [PMID: 36908874 PMCID: PMC9997180 DOI: 10.1016/j.nbas.2022.100045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 06/22/2022] [Accepted: 06/24/2022] [Indexed: 11/23/2022] Open
Abstract
Increase in the brain expression of the 18 kDa translocator protein (TSPO) is considered as a marker of neuroinflammation in the context of brain diseases, such as Alzheimer's disease (AD). However, in non-demented subjects with Alzheimer's neuropathology, TSPO accumulation in hippocampus subdivisions has not been fully characterized. To determine if TSPO is associated with the presence of amyloid β plaques and/or phosphorylated Tau accumulation, we analyzed hippocampal sections using immunohistochemistry of 14 non-demented subjects with positive staining for Aβ and/or phosphorylated Tau. TSPO expression was heterogenous with higher accumulation in the CA2/3 and subiculum subfields of the hippocampus. Its distribution closely resembled that of the microglial IBA1 marker and of the Aβ42 amyloid form. In addition, positive correlations were observed between TSPO and IBA1 densities in CA4, CA2/3 and the subiculum but not with either the astrocyte GFAP marker or the AD-type Aβ and Tau markers. This study sustains the hypothesis that TSPO is mainly associated with microglia and in Aβ42-rich subdivisions in the hippocampus of non-demented elderly individuals.
Collapse
|
21
|
Lee S, Kim TD, Kim RY, Joo Y, Chung YA, Lim SM, Lyoo IK, Kim J, Yoon S. Hippocampal subregional alterations and verbal fluency in the early stage of type 2 diabetes mellitus. Eur J Neurosci 2021; 54:7550-7559. [PMID: 34687097 DOI: 10.1111/ejn.15505] [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: 04/15/2021] [Revised: 10/16/2021] [Accepted: 10/21/2021] [Indexed: 11/30/2022]
Abstract
Growing evidence indicates that type 2 diabetes mellitus (T2DM)-related cognitive dysfunction may develop in the early stage of the disease and is often accompanied by hippocampal structural alterations. In the current study, we investigated volume and shape alterations of the hippocampus at a subregional level in patients with T2DM. With the use of high-resolution brain structural images that were obtained from 30 T2DM patients with less than 5 years of disease duration and 30 healthy individuals, volumetric and shape analyses were performed. We also assessed the relationship between T2DM-related hippocampal structural alterations and performance on verbal fluency. In volumetric analysis, total hippocampal volume was smaller in the T2DM group, relative to the control group. At a subregional level, T2DM patients showed significant inward deformation and volume reduction of the right dentate gyrus and cornu ammonis 2/3 subregions as compared with healthy individuals. In particular, T2DM patients with lower performance on verbal fluency had smaller right dentate gyrus volumes relative to those with higher performance. These findings suggest that the hippocampus may undergo atrophy at a subregional level even in the early stage of T2DM, and this subregion-specific atrophy may be associated with reduced performance on verbal fluency.
Collapse
Affiliation(s)
- Suji Lee
- Ewha Brain Institute, Ewha Womans University, Seoul, South Korea
| | - Tammy D Kim
- Ewha Brain Institute, Ewha Womans University, Seoul, South Korea
| | - Rye Young Kim
- Ewha Brain Institute, Ewha Womans University, Seoul, South Korea.,Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, South Korea
| | - Yoonji Joo
- Ewha Brain Institute, Ewha Womans University, Seoul, South Korea.,Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, South Korea
| | - Yong-An Chung
- Department of Radiology, Incheon St. Mary's Hospital, The Catholic University of Korea College of Medicine, Incheon, South Korea
| | - Soo Mee Lim
- Department of Radiology, College of Medicine, Ewha Womans University, Seoul, South Korea
| | - In Kyoon Lyoo
- Ewha Brain Institute, Ewha Womans University, Seoul, South Korea.,Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, South Korea.,Department of Brain and Cognitive Sciences, Ewha Womans University, Seoul, South Korea.,The Brain Institute and Department of Psychiatry, University of Utah, Salt Lake City, Utah, USA
| | - Jungyoon Kim
- Ewha Brain Institute, Ewha Womans University, Seoul, South Korea.,Department of Brain and Cognitive Sciences, Ewha Womans University, Seoul, South Korea
| | - Sujung Yoon
- Ewha Brain Institute, Ewha Womans University, Seoul, South Korea.,Department of Brain and Cognitive Sciences, Ewha Womans University, Seoul, South Korea
| |
Collapse
|
22
|
Complement-associated loss of CA2 inhibitory synapses in the demyelinated hippocampus impairs memory. Acta Neuropathol 2021; 142:643-667. [PMID: 34170374 PMCID: PMC8423657 DOI: 10.1007/s00401-021-02338-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 06/03/2021] [Accepted: 06/14/2021] [Indexed: 12/27/2022]
Abstract
The complement system is implicated in synapse loss in the MS hippocampus, but the functional consequences of synapse loss remain poorly understood. Here, in post-mortem MS hippocampi with demyelination we find that deposits of the complement component C1q are enriched in the CA2 subfield, are linked to loss of inhibitory synapses and are significantly higher in MS patients with cognitive impairments compared to those with preserved cognitive functions. Using the cuprizone mouse model of demyelination, we corroborated that C1q deposits are highest within the demyelinated dorsal hippocampal CA2 pyramidal layer and co-localized with inhibitory synapses engulfed by microglia/macrophages. In agreement with the loss of inhibitory perisomatic synapses, we found that Schaffer collateral feedforward inhibition but not excitation was impaired in CA2 pyramidal neurons and accompanied by intrinsic changes and a reduced spike output. Finally, consistent with excitability deficits, we show that cuprizone-treated mice exhibit impaired encoding of social memories. Together, our findings identify CA2 as a critical circuit in demyelinated intrahippocampal lesions and memory dysfunctions in MS.
Collapse
|
23
|
RGS14 Regulation of Post-Synaptic Signaling and Spine Plasticity in Brain. Int J Mol Sci 2021; 22:ijms22136823. [PMID: 34201943 PMCID: PMC8268017 DOI: 10.3390/ijms22136823] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 06/12/2021] [Accepted: 06/14/2021] [Indexed: 02/06/2023] Open
Abstract
The regulator of G-protein signaling 14 (RGS14) is a multifunctional signaling protein that regulates post synaptic plasticity in neurons. RGS14 is expressed in the brain regions essential for learning, memory, emotion, and stimulus-induced behaviors, including the basal ganglia, limbic system, and cortex. Behaviorally, RGS14 regulates spatial and object memory, female-specific responses to cued fear conditioning, and environmental- and psychostimulant-induced locomotion. At the cellular level, RGS14 acts as a scaffolding protein that integrates G protein, Ras/ERK, and calcium/calmodulin signaling pathways essential for spine plasticity and cell signaling, allowing RGS14 to naturally suppress long-term potentiation (LTP) and structural plasticity in hippocampal area CA2 pyramidal cells. Recent proteomics findings indicate that RGS14 also engages the actomyosin system in the brain, perhaps to impact spine morphogenesis. Of note, RGS14 is also a nucleocytoplasmic shuttling protein, where its role in the nucleus remains uncertain. Balanced nuclear import/export and dendritic spine localization are likely essential for RGS14 neuronal functions as a regulator of synaptic plasticity. Supporting this idea, human genetic variants disrupting RGS14 localization also disrupt RGS14’s effects on plasticity. This review will focus on the known and unexplored roles of RGS14 in cell signaling, physiology, disease and behavior.
Collapse
|
24
|
Robert V, Therreau L, Davatolhagh MF, Bernardo-Garcia FJ, Clements KN, Chevaleyre V, Piskorowski RA. The mechanisms shaping CA2 pyramidal neuron action potential bursting induced by muscarinic acetylcholine receptor activation. J Gen Physiol 2021; 152:133812. [PMID: 32069351 PMCID: PMC7141590 DOI: 10.1085/jgp.201912462] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 12/18/2019] [Accepted: 01/13/2020] [Indexed: 01/04/2023] Open
Abstract
Recent studies have revealed that hippocampal area CA2 plays an important role in hippocampal network function. Disruption of this region has been implicated in neuropsychiatric disorders. It is well appreciated that cholinergic input to the hippocampus plays an important role in learning and memory. While the effect of elevated cholinergic tone has been well studied in areas CA1 and CA3, it remains unclear how changes in cholinergic tone impact synaptic transmission and the intrinsic properties of neurons in area CA2. In this study, we applied the cholinergic agonist carbachol and performed on-cell, whole-cell, and extracellular recordings in area CA2. We observed that under conditions of high cholinergic tone, CA2 pyramidal neurons depolarized and rhythmically fired bursts of action potentials. This depolarization depended on the activation of M1 and M3 cholinergic receptors. Furthermore, we examined how the intrinsic properties and action-potential firing were altered in CA2 pyramidal neurons treated with 10 µM carbachol. While this intrinsic burst firing persisted in the absence of synaptic transmission, bursts were shaped by synaptic inputs in the intact network. We found that both excitatory and inhibitory synaptic transmission were reduced upon carbachol treatment. Finally, we examined the contribution of different channels to the cholinergic-induced changes in neuronal properties. We found that a conductance from Kv7 channels partially contributed to carbachol-induced changes in resting membrane potential and membrane resistance. We also found that D-type potassium currents contributed to controlling several properties of the bursts, including firing rate and burst kinetics. Furthermore, we determined that T-type calcium channels and small conductance calcium-activated potassium channels play a role in regulating bursting activity.
Collapse
Affiliation(s)
- Vincent Robert
- Université Paris Descartes, Inserm UMR1266, Institute of Psychiatry and Neuroscience of Paris, Team Synaptic Plasticity and Neural Networks, Paris, France
| | - Ludivine Therreau
- Université Paris Descartes, Inserm UMR1266, Institute of Psychiatry and Neuroscience of Paris, Team Synaptic Plasticity and Neural Networks, Paris, France
| | - M Felicia Davatolhagh
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - F Javier Bernardo-Garcia
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA
| | | | - Vivien Chevaleyre
- Université Paris Descartes, Inserm UMR1266, Institute of Psychiatry and Neuroscience of Paris, Team Synaptic Plasticity and Neural Networks, Paris, France
| | - Rebecca A Piskorowski
- Université Paris Descartes, Inserm UMR1266, Institute of Psychiatry and Neuroscience of Paris, Team Synaptic Plasticity and Neural Networks, Paris, France
| |
Collapse
|
25
|
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
|
26
|
Walker JM, Richardson TE, Farrell K, Iida MA, Foong C, Shang P, Attems J, Ayalon G, Beach TG, Bigio EH, Budson A, Cairns NJ, Corrada M, Cortes E, Dickson DW, Fischer P, Flanagan ME, Franklin E, Gearing M, Glass J, Hansen LA, Haroutunian V, Hof PR, Honig L, Kawas C, Keene CD, Kofler J, Kovacs GG, Lee EB, Lutz MI, Mao Q, Masliah E, McKee AC, McMillan CT, Mesulam MM, Murray M, Nelson PT, Perrin R, Pham T, Poon W, Purohit DP, Rissman RA, Sakai K, Sano M, Schneider JA, Stein TD, Teich AF, Trojanowski JQ, Troncoso JC, Vonsattel JP, Weintraub S, Wolk DA, Woltjer RL, Yamada M, Yu L, White CL, Crary JF. Early Selective Vulnerability of the CA2 Hippocampal Subfield in Primary Age-Related Tauopathy. J Neuropathol Exp Neurol 2021; 80:102-111. [PMID: 33367843 PMCID: PMC8453611 DOI: 10.1093/jnen/nlaa153] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Primary age-related tauopathy (PART) is a neurodegenerative entity defined as Alzheimer-type neurofibrillary degeneration primarily affecting the medial temporal lobe with minimal to absent amyloid-β (Aβ) plaque deposition. The extent to which PART can be differentiated pathoanatomically from Alzheimer disease (AD) is unclear. Here, we examined the regional distribution of tau pathology in a large cohort of postmortem brains (n = 914). We found an early vulnerability of the CA2 subregion of the hippocampus to neurofibrillary degeneration in PART, and semiquantitative assessment of neurofibrillary degeneration in CA2 was significantly greater than in CA1 in PART. In contrast, subjects harboring intermediate-to-high AD neuropathologic change (ADNC) displayed relative sparing of CA2 until later stages of their disease course. In addition, the CA2/CA1 ratio of neurofibrillary degeneration in PART was significantly higher than in subjects with intermediate-to-high ADNC burden. Furthermore, the distribution of tau pathology in PART diverges from the Braak NFT staging system and Braak stage does not correlate with cognitive function in PART as it does in individuals with intermediate-to-high ADNC. These findings highlight the need for a better understanding of the contribution of PART to cognitive impairment and how neurofibrillary degeneration interacts with Aβ pathology in AD and PART.
Collapse
Affiliation(s)
- Jamie M Walker
- From the Department of Pathology, University of Texas Health Science Center, San Antonio, Texas, USA
- Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, University of Texas Health Science Center, San Antonio, Texas, USA
| | - Timothy E Richardson
- From the Department of Pathology, University of Texas Health Science Center, San Antonio, Texas, USA
- Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, University of Texas Health Science Center, San Antonio, Texas, USA
- Department of Pathology, State University of New York, Upstate Medical University, Syracuse, New York, USA
| | - Kurt Farrell
- Department of Pathology and Nash Family Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Neuropathology Brain Bank & Research Core, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Megan A Iida
- Department of Pathology and Nash Family Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Neuropathology Brain Bank & Research Core, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Chan Foong
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Ping Shang
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Johannes Attems
- Institute for Ageing and Health, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Gai Ayalon
- Department of Neuroscience, Genentech Inc., South San Francisco, California, USA
| | - Thomas G Beach
- Neuropathology, Banner Sun Health Research Institute, Sun City, Arizona, USA
| | - Eileen H Bigio
- Department of Pathology, Northwestern Cognitive Neurology and Alzheimer Disease Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Andrew Budson
- Department of Pathology, VA Medical Center & Boston University School of Medicine, Boston, Massachusetts, USA
| | - Nigel J Cairns
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - María Corrada
- Institute for Memory Impairments and Neurological Disorders, UC Irvine, Irvine, California, USA
| | - Etty Cortes
- Department of Pathology and Nash Family Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Dennis W Dickson
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | - Peter Fischer
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Laboratory Medicine Program, University Health Network, and Tanz Centre for Research in Neurodegenerative Disease, Krembil Brain Institute, Toronto, Ontario, Canada
| | - Margaret E Flanagan
- Department of Pathology, Northwestern Cognitive Neurology and Alzheimer Disease Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Erin Franklin
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Marla Gearing
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Jonathan Glass
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Lawrence A Hansen
- Departments of Neurosciences and Pathology, University of California, San Diego, La Jolla, California, USA
| | - Vahram Haroutunian
- Department of Psychiatry and Alzheimer’s Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Patrick R Hof
- Department of Pathology and Nash Family Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Neuropathology Brain Bank & Research Core, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Lawrence Honig
- Department of Neurology, Columbia University Irving Medical Center, New York, New York, USA
| | - Claudia Kawas
- Institute for Memory Impairments and Neurological Disorders, UC Irvine, Irvine, California, USA
| | - C Dirk Keene
- Department of Pathology, University of Washington, Seattle, Washington, USA
| | - Julia Kofler
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Gabor G Kovacs
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Laboratory Medicine Program, University Health Network, and Tanz Centre for Research in Neurodegenerative Disease, Krembil Brain Institute, Toronto, Ontario, Canada
| | - Edward B Lee
- Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Mirjam I Lutz
- Institute of Neurology, Medical University of Vienna, Vienna, Austria
| | - Qinwen Mao
- Neuropathology, Banner Sun Health Research Institute, Sun City, Arizona, USA
| | - Eliezer Masliah
- Departments of Neurosciences and Pathology, University of California, San Diego, La Jolla, California, USA
| | - Ann C McKee
- Department of Pathology, VA Medical Center & Boston University School of Medicine, Boston, Massachusetts, USA
| | - Corey T McMillan
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - M Marsel Mesulam
- Department of Pathology, Northwestern Cognitive Neurology and Alzheimer Disease Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Melissa Murray
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | - Peter T Nelson
- Department of Pathology and Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
| | - Richard Perrin
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Thao Pham
- Department of Pathology, Oregon Health Sciences University, Portland, Oregon, USA
| | - Wayne Poon
- Institute for Memory Impairments and Neurological Disorders, UC Irvine, Irvine, California, USA
| | - Dushyant P Purohit
- Department of Pathology and Nash Family Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Robert A Rissman
- Departments of Neurosciences and Pathology, University of California, San Diego, La Jolla, California, USA
| | - Kenji Sakai
- Department of Neurology and Neurobiology of Aging, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Mary Sano
- Department of Psychiatry and Alzheimer’s Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Julie A Schneider
- Departments of Pathology and Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA
| | - Thor D Stein
- Department of Pathology, VA Medical Center & Boston University School of Medicine, Boston, Massachusetts, USA
| | - Andrew F Teich
- Department of Pathology & Cell Biology and the Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York, New York, USA
| | - John Q Trojanowski
- Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Juan C Troncoso
- Division of Neuropathology, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jean-Paul Vonsattel
- Department of Pathology & Cell Biology and the Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York, New York, USA
| | - Sandra Weintraub
- Department of Pathology, Northwestern Cognitive Neurology and Alzheimer Disease Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - David A Wolk
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Randall L Woltjer
- Department of Pathology, Oregon Health Sciences University, Portland, Oregon, USA
| | - Masahito Yamada
- Department of Neurology and Neurobiology of Aging, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Lei Yu
- Departments of Pathology and Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA
| | - Charles L White
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - John F Crary
- Department of Pathology and Nash Family Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Neuropathology Brain Bank & Research Core, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
27
|
Leblanc H, Ramirez S. Linking Social Cognition to Learning and Memory. J Neurosci 2020; 40:8782-8798. [PMID: 33177112 PMCID: PMC7659449 DOI: 10.1523/jneurosci.1280-20.2020] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 09/30/2020] [Accepted: 10/02/2020] [Indexed: 12/16/2022] Open
Abstract
Many mammals have evolved to be social creatures. In humans, the ability to learn from others' experiences is essential to survival; and from an early age, individuals are surrounded by a social environment that helps them develop a variety of skills, such as walking, talking, and avoiding danger. Similarly, in rodents, behaviors, such as food preference, exploration of novel contexts, and social approach, can be learned through social interaction. Social encounters facilitate new learning and help modify preexisting memories throughout the lifespan of an organism. Moreover, social encounters can help buffer stress or the effects of negative memories, as well as extinguish maladaptive behaviors. Given the importance of such interactions, there has been increasing work studying social learning and applying its concepts in a wide range of fields, including psychotherapy and medical sociology. The process of social learning, including its neural and behavioral mechanisms, has also been a rapidly growing field of interest in neuroscience. However, the term "social learning" has been loosely applied to a variety of psychological phenomena, often without clear definition or delineations. Therefore, this review gives a definition for specific aspects of social learning, provides an overview of previous work at the circuit, systems, and behavioral levels, and finally, introduces new findings on the social modulation of learning. We contextualize such social processes in the brain both through the role of the hippocampus and its capacity to process "social engrams" as well as through the brainwide realization of social experiences. With the integration of new technologies, such as optogenetics, chemogenetics, and calcium imaging, manipulating social engrams will likely offer a novel therapeutic target to enhance the positive buffering effects of social experiences or to inhibit fear-inducing social stimuli in models of anxiety and post-traumatic stress disorder.
Collapse
Affiliation(s)
- Heloise Leblanc
- Department of Psychological and Brain Sciences, Boston University, Boston, Massachusetts, 02119
- Boston University School of Medicine, Boston, Massachusetts, 02118
| | - Steve Ramirez
- Department of Psychological and Brain Sciences, Boston University, Boston, Massachusetts, 02119
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, 02119
- Neurophotonics Center at Boston University, Boston, Massachusetts, 02119
- Center for Systems Neuroscience at Boston University, Boston, Massachusetts, 02119
| |
Collapse
|
28
|
Dual prophylactic/therapeutic potential of date seed, and nigella and olive oils-based nutraceutical formulation in rats with experimentally-induced Alzheimer's disease: A mechanistic insight. J Chem Neuroanat 2020; 110:101878. [PMID: 33144183 DOI: 10.1016/j.jchemneu.2020.101878] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 10/24/2020] [Indexed: 11/21/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder with a multifactorial etiology and significantly increasing incidence during the last decade. Hence, developing an effective therapy is crucial for public health. The current study aimed to examine the dual prophylactic/therapeutic potential of a nutraceutical formula based on aqueous extract of roasted date seeds, and nigella and virgin-olive oils against experimentally-induced Alzheimer's disease in rats. Alzheimer's disease-like pathology was induced in male Wistar rats using oral CuSO4 (200 mg/Kg/day for two months). The nutraceutical formula was given orally to experimental animals (10 mL/kg/d) for 14 days before (as prophylaxis) and after Alzheimer's disease induction and its therapeutic effect in both cases is tested in comparison to donepezil (0.5 mg/kg/d). The nutraceutical formula was found to ameliorate the CuSO4-induced neuronal damage and regenerate the affected hippocampus tissue and significantly improvemed in learning ability. The formula was also effective in decreasing brain amyloid-β, tau protein, TNF-α level, iNOS level in hippocampus, oxidative stress level, and inhibiting acetylcholinesterase activity and expression in brain and hippocampus, respectively. Further, an increase in GSH levels, activities of SOD, and GST and levels of hippocampus ADAM 17 and brain phospholipids was observed. In conclusion, the studied nutraceutical formula is proved to be effective in ameliorating Alzheimer's neurodegenerative progression with added-prophylactic potential.
Collapse
|
29
|
Schultheiss NW, Schlecht M, Jayachandran M, Brooks DR, McGlothan JL, Guilarte TR, Allen TA. Awake delta and theta-rhythmic hippocampal network modes during intermittent locomotor behaviors in the rat. Behav Neurosci 2020; 134:529-546. [PMID: 32672989 DOI: 10.1037/bne0000409] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Delta-frequency network activity is commonly associated with sleep or behavioral disengagement accompanied by a dearth of cortical spiking, but delta in awake behaving animals is not well understood. We show that hippocampal (HC) synchronization in the delta frequency band (1-4 Hz) is related to animals' locomotor behavior using detailed analyses of the HC local field potential (LFP) and simultaneous head- and body-tracking data. In contrast to running-speed modulation of the theta rhythm (6-10 Hz), delta was most prominent when animals were stationary or moving slowly, that is, when theta and fast gamma (65-120 Hz) were weak, and often developed rapidly when animals paused briefly between runs. We next combined time-frequency decomposition of the LFP with hierarchical clustering algorithms to categorize momentary estimations of the power spectral density (PSD) into putative modes of HC activity. Delta and theta power were strikingly orthogonal across spectral modes, as well as across bouts of precisely defined running and stationary behavior. Delta-band and theta-band coherences between HC recording sites were monotonically related to theta-delta ratios across modes; and whereas theta coherence between HC and medial prefrontal cortex (mPFC) increased during running, delta-band coherence between mPFC and HC increased during stationary bouts. Taken together, our findings suggest that delta-dominated network modes (and corresponding mPFC-HC couplings) represent functionally distinct circuit dynamics that are temporally and behaviorally interspersed among theta-dominated modes during navigation. As such, delta modes could play a fundamental role in coordinating encoding and retrieval mechanisms or decision-making processes at a timescale that segments event sequences within behavioral episodes. (PsycInfo Database Record (c) 2021 APA, all rights reserved).
Collapse
Affiliation(s)
| | | | | | - Deborah R Brooks
- Department of Environmental Health Sciences, Robert Stempel College of Public Health, Florida International University
| | - Jennifer L McGlothan
- Department of Environmental Health Sciences, Robert Stempel College of Public Health, Florida International University
| | - Tomás R Guilarte
- Department of Environmental Health Sciences, Robert Stempel College of Public Health, Florida International University
| | - Timothy A Allen
- Cognitive Neuroscience Program, Robert Stempel College of Public Health, Florida International University
| |
Collapse
|
30
|
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
|
31
|
Mrdjen D, Fox EJ, Bukhari SA, Montine KS, Bendall SC, Montine TJ. The basis of cellular and regional vulnerability in Alzheimer's disease. Acta Neuropathol 2019; 138:729-749. [PMID: 31392412 PMCID: PMC6802290 DOI: 10.1007/s00401-019-02054-4] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 07/24/2019] [Accepted: 07/31/2019] [Indexed: 12/11/2022]
Abstract
Alzheimer's disease (AD) differentially and specifically affects brain regions and neuronal cell types in a predictable pattern. Damage to the brain appears to spread and worsens with time, taking over more regions and activating multiple stressors that can converge to promote vulnerability of certain cell types. At the same time, other cell types and brain regions remain intact in the face of this onslaught of neuropathology. Although neuropathologic descriptions of AD have been extensively expanded and mapped over the last several decades, our understanding of the mechanisms underlying how certain regions and cell populations are specifically vulnerable or resistant has lagged behind. In this review, we detail what is known about the selectivity of local initiation of AD pathology in the hippocampus, its proposed spread via synaptic connections, and the diversity of clinical phenotypes and brain atrophy patterns that may arise from different fibrillar strains of pathologic proteins or genetic predispositions. We summarize accumulated and emerging knowledge of the cellular and molecular basis for neuroanatomic selectivity, consider potential disease-relevant differences between vulnerable and resistant neuronal cell types and isolate molecular markers to identify them.
Collapse
Affiliation(s)
- Dunja Mrdjen
- Department of Pathology, School of Medicine, Stanford University, Palo Alto, CA, USA
| | - Edward J Fox
- Department of Pathology, School of Medicine, Stanford University, Palo Alto, CA, USA
| | - Syed A Bukhari
- Department of Pathology, School of Medicine, Stanford University, Palo Alto, CA, USA
| | - Kathleen S Montine
- Department of Pathology, School of Medicine, Stanford University, Palo Alto, CA, USA
| | - Sean C Bendall
- Department of Pathology, School of Medicine, Stanford University, Palo Alto, CA, USA
| | - Thomas J Montine
- Department of Pathology, School of Medicine, Stanford University, Palo Alto, CA, USA.
| |
Collapse
|
32
|
Chang J, Yu R. Hippocampal connectivity in the aftermath of acute social stress. Neurobiol Stress 2019; 11:100195. [PMID: 31832509 PMCID: PMC6889252 DOI: 10.1016/j.ynstr.2019.100195] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 08/06/2019] [Accepted: 09/13/2019] [Indexed: 12/12/2022] Open
Abstract
The hippocampus is a core brain region that responds to stress. Previous studies have found a dysconnectivity between hippocampus and other brain regions under acute and chronic stress. However, whether and how acute social stress influences the directed connectivity patterns from and to the hippocampus remains unclear. In this study, using a within-subject design and Granger causal analysis (GCA), we investigated the alterations of resting state effective connectivity from and to hippocampal subregions after an acute social stressor (the Trier Social Stress Test). Participants were engaged in stress and control conditions spaced approximately one month apart. Our findings showed that stress altered the information flows in the thalamus-hippocampus-insula/midbrain circuit. The changes in this circuit could also predict with high accuracy the stress and control conditions at the subject level. These hippocampus-related brain networks have been documented to be involved in emotional information processing and storage, as well as habitual responses. We speculate that alterations of the effective connectivity between these brain regions may be associated with the registering and encoding of threatening stimuli under stress. Our investigation of hippocampal functional connectivity at a subregional level may help elucidate the functional neurobiology of stress-related psychiatric disorders.
Collapse
Affiliation(s)
- Jingjing Chang
- Guangdong Key Laboratory of Mental Health and Cognitive Science, Center for Studies of Psychological Application, School of Psychology, South China Normal University, Guangzhou, China
| | - Rongjun Yu
- Department of Psychology, National University of Singapore, Singapore
| |
Collapse
|
33
|
Todorov OS, Weisbecker V, Gilissen E, Zilles K, de Sousa AA. Primate hippocampus size and organization are predicted by sociality but not diet. Proc Biol Sci 2019; 286:20191712. [PMID: 31662078 DOI: 10.1098/rspb.2019.1712] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The hippocampus is well known for its roles in spatial navigation and memory, but it is organized into regions that have different connections and functional specializations. Notably, the region CA2 has a role in social and not spatial cognition, as is the case for the regions CA1 and CA3 that surround it. Here, we investigated the evolution of the hippocampus in terms of its size and organization in relation to the evolution of social and ecological variables in primates, namely home range, diet and different measures of group size. We found that the volumes within the whole cornu ammonis coevolve with group size, while only the volume of CA1 and subiculum can also be predicted by home range. On the other hand, diet, expressed as a shift from folivory towards frugivory, was shown to not be related to hippocampal volume. Interestingly, CA2 was shown to exhibit phylogenetic signal only against certain measures of group size, but not with ecological factors. We also found that sex differences in the hippocampus are related to body size sex dimorphism. This is in line with reports of sex differences in hippocampal volume in non-primates that are related to social structure and sex differences in behaviour. Our findings support the notion that in primates, the hippocampus is a mosaic structure evolving in line with social pressures, where certain subsections evolve in line with spatial ability too.
Collapse
Affiliation(s)
- Orlin S Todorov
- School of Biological Sciences, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Vera Weisbecker
- School of Biological Sciences, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Emmanuel Gilissen
- Department of African Zoology, Royal Museum for Central Africa, Leuvensesteenweg, 3080 Tervuren, Belgium.,Laboratory of Histology and Neuropathology, Université Libre de Bruxelles, Brussels, Belgium
| | - Karl Zilles
- Research Centre Jülich, Institute of Neuroscience and Medicine (INM-1), Jülich, Germany
| | | |
Collapse
|
34
|
Domínguez S, Rey CC, Therreau L, Fanton A, Massotte D, Verret L, Piskorowski RA, Chevaleyre V. Maturation of PNN and ErbB4 Signaling in Area CA2 during Adolescence Underlies the Emergence of PV Interneuron Plasticity and Social Memory. Cell Rep 2019; 29:1099-1112.e4. [DOI: 10.1016/j.celrep.2019.09.044] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 07/31/2019] [Accepted: 09/13/2019] [Indexed: 12/28/2022] Open
|
35
|
Jiang L, Cao X, Jiang J, Li T, Wang J, Yang Z, Li C. Atrophy of hippocampal subfield CA2/3 in healthy elderly men is related to educational attainment. Neurobiol Aging 2019; 80:21-28. [PMID: 31077957 DOI: 10.1016/j.neurobiolaging.2019.03.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 03/10/2019] [Accepted: 03/27/2019] [Indexed: 11/16/2022]
Abstract
A higher education level is a protective factor against cognitive decline in elders; however, the underlying neural mechanisms remain unclear. Modulated by both aging and education, the hippocampus is a starting point for understanding the long-lasting effect of education on the aging of human brain. Because the hippocampus possesses functionally heterogeneous subfields and exhibits sex differences, we examined hippocampal subfields in men and women separately. We performed both cross-sectional (n = 143) and longitudinal (n = 51) analyses on healthy participants aged 65-75 years, who underwent structural magnetic resonance imaging. Volumes of the hippocampi and their subfields were estimated by automated segmentation. We found significantly positive correlations between educational attainment and the volume of hippocampal CA2/3 in men but not in women. The longitudinal analysis focusing on this region validated the above results by showing that a higher education level attenuated the progression of atrophy during a 15-month follow-up period in the CA2/3 region in men. These findings suggest that, in men, education plays a role in the aging of specific hippocampal subfields.
Collapse
Affiliation(s)
- Lijuan Jiang
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinyi Cao
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiangling Jiang
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ting Li
- Department of Geriatric Psychiatry, Shanghai Changning Mental Health Center, Shanghai, China
| | - Jijun Wang
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Center for Excellence in Brain Science and Intelligence Technology (CEBSIT), Chinese Academy of Science, Shanghai, China; Institute of Psychology and Behavioral Science, Shanghai Jiao Tong Universit, Shanghai, China
| | - Zhi Yang
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Institute of Psychology and Behavioral Science, Shanghai Jiao Tong Universit, Shanghai, China; Brain Science and Technology Research Center, Shanghai Jiao Tong University, Shanghai, China.
| | - Chunbo Li
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Center for Excellence in Brain Science and Intelligence Technology (CEBSIT), Chinese Academy of Science, Shanghai, China; Institute of Psychology and Behavioral Science, Shanghai Jiao Tong Universit, Shanghai, China; Brain Science and Technology Research Center, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
36
|
Liu AKL, Chau TW, Lim EJ, Ahmed I, Chang RCC, Kalaitzakis ME, Graeber MB, Gentleman SM, Pearce RKB. Hippocampal CA2 Lewy pathology is associated with cholinergic degeneration in Parkinson's disease with cognitive decline. Acta Neuropathol Commun 2019; 7:61. [PMID: 31023342 PMCID: PMC6485180 DOI: 10.1186/s40478-019-0717-3] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Accepted: 04/10/2019] [Indexed: 01/06/2023] Open
Abstract
Although the precise neuropathological substrates of cognitive decline in Parkinson's disease (PD) remain elusive, it has long been regarded that pathology in the CA2 hippocampal subfield is characteristic of Lewy body dementias, including dementia in PD (PDD). Early non-human primate tracer studies demonstrated connections from the nucleus of the vertical limb of the diagonal band of Broca (nvlDBB, Ch2) to the hippocampus. However, the relationship between Lewy pathology of the CA2 subfield and cholinergic fibres has not been explored. Therefore, in this study, we investigated the burden of pathology in the CA2 subsector of PD cases with varying degrees of cognitive impairment and correlated this with the extent of septohippocampal cholinergic deficit. Hippocampal sections from 67 PD, 34 PD with mild cognitive impairment and 96 PDD cases were immunostained for tau and alpha-synuclein, and the respective pathology burden was assessed semi-quantitatively. In a subset of cases, the degree of CA2 cholinergic depletion was quantified using confocal microscopy and correlated with cholinergic neuronal loss in Ch2. We found that only cases with dementia have a significantly greater Lewy pathology, whereas cholinergic fibre depletion was evident in cases with mild cognitive impairment and this was significantly correlated with loss of cholinergic neurons in Ch2. In addition, multiple antigen immunofluorescence demonstrated colocalisation between cholinergic fibres and alpha-synuclein but not tau pathology. Such specific Lewy pathology targeting the cholinergic system within the CA2 subfield may contribute to the unique memory retrieval deficit seen in patients with Lewy body disorders, as distinct from the memory storage deficit seen in Alzheimer's disease.
Collapse
Affiliation(s)
- Alan King Lun Liu
- Neuropathology Unit, Division of Brain Sciences, Department of Medicine, Imperial College London, 4/F, Burlington Danes Building, Du Cane Road, London, W12 0NN, UK.
| | - Tsz Wing Chau
- Neuropathology Unit, Division of Brain Sciences, Department of Medicine, Imperial College London, 4/F, Burlington Danes Building, Du Cane Road, London, W12 0NN, UK
| | - Ernest Junwei Lim
- Neuropathology Unit, Division of Brain Sciences, Department of Medicine, Imperial College London, 4/F, Burlington Danes Building, Du Cane Road, London, W12 0NN, UK
| | - Idil Ahmed
- Neuropathology Unit, Division of Brain Sciences, Department of Medicine, Imperial College London, 4/F, Burlington Danes Building, Du Cane Road, London, W12 0NN, UK
| | - Raymond Chuen-Chung Chang
- Laboratory of Neurodegenerative Diseases, School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR
- State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Pokfulam, Hong Kong, Special Administrative Region of China
| | - Michail E Kalaitzakis
- Neuropathology Unit, Division of Brain Sciences, Department of Medicine, Imperial College London, 4/F, Burlington Danes Building, Du Cane Road, London, W12 0NN, UK
| | - Manuel B Graeber
- Brain and Mind Centre, Bosch Institute, Discipline of Anatomy and Embryology, and Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, 94 Mallett Street, Camperdown, NSW, 2050, Australia
| | - Steve M Gentleman
- Neuropathology Unit, Division of Brain Sciences, Department of Medicine, Imperial College London, 4/F, Burlington Danes Building, Du Cane Road, London, W12 0NN, UK
| | - Ronald K B Pearce
- Neuropathology Unit, Division of Brain Sciences, Department of Medicine, Imperial College London, 4/F, Burlington Danes Building, Du Cane Road, London, W12 0NN, UK
| |
Collapse
|
37
|
Do multiple system atrophy and Parkinson's disease show distinct patterns of volumetric alterations across hippocampal subfields? An exploratory study. Eur Radiol 2019; 29:4948-4956. [PMID: 30796577 DOI: 10.1007/s00330-019-06043-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 12/25/2018] [Accepted: 01/24/2019] [Indexed: 12/21/2022]
Abstract
OBJECTIVES To investigate the volumetric alterations of hippocampal subfields and identify which subfields contribute to mild cognitive impairment (MCI) in multiple system atrophy (MSA) and Parkinson's disease (PD). METHODS Thirty MSA-MCI, 26 PD-MCI, and 30 healthy controls were administered cognitive assessment, along with hippocampal segmentation using FreeSurfer 6.0 after a 3-T MRI scan. Regression analyses were performed between the volumes of hippocampal subfields and cognitive variables. RESULTS Compared with healthy controls, the volume of the hippocampal fissure was enlarged in PD-MCI patients, while left Cornu Ammonis (CA2-CA3), bilateral molecular layer, bilateral hippocampus-amygdala transition area, right subiculum, right CA1, right presubiculum, right parasubiculum, and bilateral whole hippocampus were reduced in the MSA-MCI group. Moreover, volumetric reductions of the bilateral hippocampal tail, bilateral CA1, bilateral presubiculum, bilateral molecular layer, left CA2-CA3, left hippocampus-amygdala transition area, right parasubiculum, and bilateral whole hippocampus were found in MSA-MCI relative to the PD-MCI group. The volumes of the left CA2-CA3 (B = - 11.34, p = 0.006) and left parasubiculum (B = 4.63, p = 0.01) were respectively correlated with language and abstraction functions. The volumes of the left fimbria (B = 6.99, p = 0.002) and left hippocampus-amygdala transition area (B = 2.28, p = 0.009) were correlated with visuospatial/executive function. CONCLUSIONS The MSA-MCI patients showed more widespread impairment of hippocampal subfields compared with the PD-MCI group, involving trisynaptic loop and amygdala-hippocampus interactions. The alteration of CA, hippocampus-amygdala transition area, and fimbria still requires further comparison between the two patient groups. KEY POINTS • The atrophy patterns of hippocampal subfields differed between MSA and PD patients. • MSA has widespread change in trisynaptic loop and amygdala-hippocampus interactions. • The atrophy patterns may help to understand the differences of cognitive impairment in MSA and PD.
Collapse
|
38
|
Chen J, Yan Y, Yuan F, Cao J, Li S, Eickhoff SB, Zhang J. Brain grey matter volume reduction and anxiety-like behavior in lipopolysaccharide-induced chronic pulmonary inflammation rats: A structural MRI study with histological validation. Brain Behav Immun 2019; 76:182-197. [PMID: 30472482 DOI: 10.1016/j.bbi.2018.11.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 10/01/2018] [Accepted: 11/21/2018] [Indexed: 12/26/2022] Open
Abstract
While there have been multiple fMRI studies into the brain functional changes after acutely stimulated peripheral infection, knowledge for the effect of chronic peripheral infection on whole brain morphology is still quite limited. The present study was designed to investigate the brain structural and emotional changes after peripheral local infection initiated chronic systemic inflammation and the relationship between circulating inflammatory markers and brain grey matter. Specifically, in-vivo T2-weighted MRI was performed on rats with lipopolysaccharide (LPS)-induced chronic pulmonary inflammation (CPI) and those without. Grey matter volume was quantified using diffeomorphic anatomical registration through exponentiated lie (DARTEL) enhanced voxel-based morphometry followed by between-group comparison. Open field experiment was conducted to test the potential anxiety-like behaviors after CPI, along with the ELISA estimated inflammatory markers were correlated to grey matter volume. Guided by image findings, we undertook a focused histological investigation with immunefluorescence and Nissl staining. A widespread decrease of grey matter volume in CPI-model rats was revealed. 8 of the 12 measured inflammatory markers presented differential neuroanatomical correlation patterns with three of the pro-inflammatory cytokines (IL-1β, IL-6 and TNF-α) and CRP being the most notable. Lower grey matter volumes in some of the inflammatory markers related regions (amygdala, CA2 and cingulate cortex) were associated with more-severe anxiety-like behaviors. Furthermore, grey matter volumes in amygdala and CA3 were correlated negatively with the expressions of glial proteins (S100β and Nogo-A), while the grey matter volume in hypo-thalamus was changing positively with neural cell area. Overall, the neuroanatomical association patterns and the histopathology underpinning the MRI observations we demonstrated here would probably serve as one explanation for the cerebral and emotional deficits presented in the patients with CPI, which would furthermore yield new insights into the adverse effects the many other systemic inflammation and inflammatory autoimmune diseases would pose on brain morphology.
Collapse
Affiliation(s)
- Ji Chen
- Institute of Brain Diseases and Cognition, Medical College of Xiamen University, Xiamen, China; Institute of Neuroscience and Medicine, Brain and Behaviour (INM-7), Research Centre Jülich, Jülich, Germany; Institute of Systems Neuroscience, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
| | - Ya Yan
- Institute of Brain Diseases and Cognition, Medical College of Xiamen University, Xiamen, China
| | - Fengjuan Yuan
- Institute of Brain Diseases and Cognition, Medical College of Xiamen University, Xiamen, China
| | - Jianbo Cao
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, China; Medical College of Xiamen University, Xiamen, China; Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Shanhua Li
- Institute of Brain Diseases and Cognition, Medical College of Xiamen University, Xiamen, China
| | - Simon B Eickhoff
- Institute of Neuroscience and Medicine, Brain and Behaviour (INM-7), Research Centre Jülich, Jülich, Germany; Institute of Systems Neuroscience, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Jiaxing Zhang
- Institute of Brain Diseases and Cognition, Medical College of Xiamen University, Xiamen, China.
| |
Collapse
|
39
|
Sex-Specific Proteomic Changes Induced by Genetic Deletion of Fibroblast Growth Factor 14 (FGF14), a Regulator of Neuronal Ion Channels. Proteomes 2019; 7:proteomes7010005. [PMID: 30678040 PMCID: PMC6473632 DOI: 10.3390/proteomes7010005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 01/16/2019] [Accepted: 01/17/2019] [Indexed: 12/18/2022] Open
Abstract
Fibroblast growth factor 14 (FGF14) is a member of the intracellular FGFs, which is a group of proteins involved in neuronal ion channel regulation and synaptic transmission. We previously demonstrated that male Fgf14−/− mice recapitulate the salient endophenotypes of synaptic dysfunction and behaviors that are associated with schizophrenia (SZ). As the underlying etiology of SZ and its sex-specific onset remain elusive, the Fgf14−/− model may provide a valuable tool to interrogate pathways related to disease mechanisms. Here, we performed label-free quantitative proteomics to identify enriched pathways in both male and female hippocampi from Fgf14+/+ and Fgf14−/− mice. We discovered that all of the differentially expressed proteins measured in Fgf14−/− animals, relative to their same-sex wildtype counterparts, are associated with SZ based on genome-wide association data. In addition, measured changes in the proteome were predominantly sex-specific, with the male Fgf14−/− mice distinctly enriched for pathways associated with neuropsychiatric disorders. In the male Fgf14−/− mouse, we found molecular characteristics that, in part, may explain a previously described neurotransmission and behavioral phenotype. This includes decreased levels of ALDH1A1 and protein kinase A (PRKAR2B). ALDH1A1 has been shown to mediate an alternative pathway for gamma-aminobutyric acid (GABA) synthesis, while PRKAR2B is essential for dopamine 2 receptor signaling, which is the basis of current antipsychotics. Collectively, our results provide new insights in the role of FGF14 and support the use of the Fgf14−/− mouse as a useful preclinical model of SZ for generating hypotheses on disease mechanisms, sex-specific manifestation, and therapy.
Collapse
|
40
|
Muñoz MD, Solís JM. Characterisation of the mechanisms underlying the special sensitivity of the CA2 hippocampal area to adenosine receptor antagonists. Neuropharmacology 2019; 144:9-18. [DOI: 10.1016/j.neuropharm.2018.10.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 09/17/2018] [Accepted: 10/12/2018] [Indexed: 12/28/2022]
|
41
|
Das T, Hwang JJ, Poston KL. Episodic recognition memory and the hippocampus in Parkinson's disease: A review. Cortex 2018; 113:191-209. [PMID: 30660957 DOI: 10.1016/j.cortex.2018.11.021] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Revised: 10/02/2018] [Accepted: 11/15/2018] [Indexed: 01/09/2023]
Abstract
Parkinson's disease is a progressive neurodegenerative disorder of aging. The hallmark pathophysiology includes the development of neuronal Lewy bodies in the substantia nigra of the midbrain with subsequent loss of dopaminergic neurons. These neuronal losses lead to the characteristic motor symptoms of bradykinesia, rigidity, and rest tremor. In addition to these cardinal motor symptoms patients with PD experience a wide range of non-motor symptoms, the most important being cognitive impairments that in many circumstances lead to dementia. People with PD experience a wide range of cognitive impairments; in this review we will focus on memory impairment in PD and specifically episodic memory, which are memories of day-to-day events of life. Importantly, these memory impairments severely impact the lives of patients and caregivers alike. Traditionally episodic memory is considered to be markedly dependent on the hippocampus; therefore, it is important to understand the exact nature of PD episodic memory deficits in relation to hippocampal function and dysfunction. In this review, we discuss an aspect of episodic memory called recognition memory and its subcomponents called recollection and familiarity. Recognition memory is believed to be impaired in PD; thus, we discuss what aspects of the hippocampus are expected to be deficient in function as they relate to these recognition memory impairments. In addition to the hippocampus as a whole, we will discuss the role of hippocampal subfields in recognition memory impairments.
Collapse
Affiliation(s)
- Tanusree Das
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.
| | - Jaclyn J Hwang
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA; Department of Neuroscience, University of Pittsburgh, USA.
| | - Kathleen L Poston
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA; Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
42
|
Cattaud V, Bezzina C, Rey CC, Lejards C, Dahan L, Verret L. Early disruption of parvalbumin expression and perineuronal nets in the hippocampus of the Tg2576 mouse model of Alzheimer's disease can be rescued by enriched environment. Neurobiol Aging 2018; 72:147-158. [DOI: 10.1016/j.neurobiolaging.2018.08.024] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 08/28/2018] [Accepted: 08/28/2018] [Indexed: 11/30/2022]
|
43
|
Meira T, Leroy F, Buss EW, Oliva A, Park J, Siegelbaum SA. A hippocampal circuit linking dorsal CA2 to ventral CA1 critical for social memory dynamics. Nat Commun 2018; 9:4163. [PMID: 30301899 PMCID: PMC6178349 DOI: 10.1038/s41467-018-06501-w] [Citation(s) in RCA: 173] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 09/06/2018] [Indexed: 11/09/2022] Open
Abstract
Recent results suggest that social memory requires the dorsal hippocampal CA2 region as well as a subset of ventral CA1 neurons. However, it is unclear whether dorsal CA2 and ventral CA1 represent parallel or sequential circuits. Moreover, because evidence implicating CA2 in social memory comes largely from long-term inactivation experiments, the dynamic role of CA2 in social memory remains unclear. Here, we use pharmacogenetics and optogenetics in mice to acutely and reversibly silence dorsal CA2 and its projections to ventral hippocampus. We show that dorsal CA2 activity is critical for encoding, consolidation, and recall phases of social memory. Moreover, dorsal CA2 contributes to social memory by providing strong excitatory input to the same subregion of ventral CA1 that contains the subset of neurons implicated in social memory. Thus, our studies provide new insights into a dorsal CA2 to ventral CA1 circuit whose dynamic activity is necessary for social memory.
Collapse
Affiliation(s)
- Torcato Meira
- Department of Neuroscience, Zuckerman and Kavli Institutes, Vagelos College of Physicians and Surgeons, Columbia University, 3227 Broadway, New York, NY, 10027, USA
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, 4710-057, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, 4806-909, Portugal
| | - Felix Leroy
- Department of Neuroscience, Zuckerman and Kavli Institutes, Vagelos College of Physicians and Surgeons, Columbia University, 3227 Broadway, New York, NY, 10027, USA
| | - Eric W Buss
- Department of Neuroscience, Zuckerman and Kavli Institutes, Vagelos College of Physicians and Surgeons, Columbia University, 3227 Broadway, New York, NY, 10027, USA
| | - Azahara Oliva
- Department of Neuroscience, Zuckerman and Kavli Institutes, Vagelos College of Physicians and Surgeons, Columbia University, 3227 Broadway, New York, NY, 10027, USA
| | - Jung Park
- Department of Neuroscience, Zuckerman and Kavli Institutes, Vagelos College of Physicians and Surgeons, Columbia University, 3227 Broadway, New York, NY, 10027, USA
| | - Steven A Siegelbaum
- Department of Neuroscience, Zuckerman and Kavli Institutes, Vagelos College of Physicians and Surgeons, Columbia University, 3227 Broadway, New York, NY, 10027, USA.
| |
Collapse
|
44
|
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
|
45
|
Piskorowski RA, Chevaleyre V. Memory circuits: CA2. Curr Opin Neurobiol 2018; 52:54-59. [DOI: 10.1016/j.conb.2018.04.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 04/09/2018] [Indexed: 01/01/2023]
|
46
|
Perić I, Costina V, Stanisavljević A, Findeisen P, Filipović D. Proteomic characterization of hippocampus of chronically socially isolated rats treated with fluoxetine: Depression-like behaviour and fluoxetine mechanism of action. Neuropharmacology 2018; 135:268-283. [DOI: 10.1016/j.neuropharm.2018.03.034] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 03/22/2018] [Accepted: 03/24/2018] [Indexed: 12/20/2022]
|
47
|
Hayani H, Song I, Dityatev A. Increased Excitability and Reduced Excitatory Synaptic Input Into Fast-Spiking CA2 Interneurons After Enzymatic Attenuation of Extracellular Matrix. Front Cell Neurosci 2018; 12:149. [PMID: 29899690 PMCID: PMC5988902 DOI: 10.3389/fncel.2018.00149] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 05/14/2018] [Indexed: 12/21/2022] Open
Abstract
The neural extracellular matrix (ECM) is enriched with hyaluronic acid, chondroitin sulfate proteoglycans (CSPGs) and the glycoprotein tenascin-R, which play important roles in synaptic plasticity, as shown by studies of the CA1 region of the hippocampus. However, ECM molecules are strongly expressed in the CA2 region, which harbors a high number of fast-spiking interneurons (FSIs) surrounded by a particularly condensed form of ECM, perineuronal nets. Despite this intriguing peculiarity, the functional role of ECM in the CA2 region is mostly unknown. Here, we investigate the acute and delayed effects of chondroitinase ABC (ChABC), an enzyme that digests chondroitin sulfate side chains of CSPGs and greatly attenuates neural ECM, on neuronal excitability and excitatory transmission in the CA2 region. Whole-cell patch clamp recordings of CA2 pyramidal cells (PCs) and FSIs in hippocampal slices revealed that 7 days after injection of ChABC into the CA2 region in vivo, there are alterations in excitability of FSIs and PCs. FSIs generated action potentials with larger amplitudes and longer durations in response to less depolarizing currents compared to controls. PCs were excited at less depolarized membrane potentials, resulted in lower latency of spike generation. The frequency of excitatory postsynaptic currents in FSIs was selectively reduced, while the frequency of inhibitory postsynaptic currents was selectively increased. Acute treatment of hippocampal slices with ChABC did not result in any of these effects. This increase in excitability and changes in synaptic inputs to FSIs after attenuation of ECM suggests a crucial role for perineuronal nets associated with FSIs in regulation of synaptic and electrical properties of these cells.
Collapse
Affiliation(s)
- Hussam Hayani
- Molecular Neuroplasticity, German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Inseon Song
- Molecular Neuroplasticity, German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Alexander Dityatev
- Molecular Neuroplasticity, German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany.,Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany.,Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| |
Collapse
|
48
|
Bian C, Huang Y, Zhu H, Zhao Y, Zhao J, Zhang J. Steroid Receptor Coactivator-1 Knockdown Decreases Synaptic Plasticity and Impairs Spatial Memory in the Hippocampus of Mice. Neuroscience 2018. [PMID: 29524638 DOI: 10.1016/j.neuroscience.2018.02.034] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Steroids have been demonstrated to play profound roles in the regulation of hippocampal function by acting on their receptors, which need coactivators for their transcriptional activities. Previous studies have shown that steroid receptor coactivator-1 (SRC-1) is the predominant coactivator in the hippocampus, but its exact role and the underlying mechanisms remain unclear. In this study, we constructed SRC-1 RNA interference (RNAi) lentiviruses, injected them into the hippocampus of male mice, and then examined the changes in the expression of selected synaptic proteins, CA1 synapse density, postsynaptic density (PSD) thickness, and in vivo long-term potentiation (LTP). Spatial learning and memory behavior changes were investigated using the Morris water maze. We then transfected the lentiviruses into cultured hippocampal cells and examined the changes in synaptic protein and phospho-cyclic AMP response element-binding protein (pCREB) expression. The in vivo results showed that SRC-1 knockdown significantly decreased the expression of synaptic proteins and CA1 synapse density as well as PSD thickness; SRC-1 knockdown also significantly impaired in vivo LTP and disrupted spatial learning and memory. The in vitro results showed that while the expression of synaptic proteins was significantly decreased by SRC-1 knockdown, pCREB expression was also significantly decreased. The above results suggest a pivotal role of SRC-1 in the regulation of hippocampal synaptic plasticity and spatial learning and memory, strongly indicating SRC-1 may serve as a novel therapeutic target for hippocampus-dependent memory disorders.
Collapse
Affiliation(s)
- Chen Bian
- Department of Military Psychology, College of Psychology, Third Military Medical University, Chongqing 400038, China; Department of Neurobiology, Third Military Medical University, Chongqing 400038, China
| | - Yan Huang
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Haitao Zhu
- Department of Military Psychology, College of Psychology, Third Military Medical University, Chongqing 400038, China; Medical Company, Troops 95848 of People's Liberation Army, Xiaogan, Hubei 432100, China
| | - Yangang Zhao
- Department of Neurobiology, Third Military Medical University, Chongqing 400038, China
| | - Jikai Zhao
- Department of Neurobiology, Third Military Medical University, Chongqing 400038, China
| | - Jiqiang Zhang
- Department of Neurobiology, Third Military Medical University, Chongqing 400038, China.
| |
Collapse
|
49
|
Benoy A, Dasgupta A, Sajikumar S. Hippocampal area CA2: an emerging modulatory gateway in the hippocampal circuit. Exp Brain Res 2018; 236:919-931. [DOI: 10.1007/s00221-018-5187-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Accepted: 01/22/2018] [Indexed: 12/18/2022]
|
50
|
Hippocampal area CA2: properties and contribution to hippocampal function. Cell Tissue Res 2018; 373:525-540. [PMID: 29335778 DOI: 10.1007/s00441-017-2769-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 12/07/2017] [Indexed: 12/30/2022]
Abstract
This review focuses on area CA2 of the hippocampus, as recent results have revealed the unique properties and surprising role of this region in encoding social, temporal and contextual aspects of memory. Originally identified and described by Lorente de No, in 1934, this region of the hippocampus has unique intra-and extra-hippocampal connectivity, sending and receiving input to septal and hypothalamic regions. Recent in vivo studies have indicated that CA2 pyramidal neurons encode spatial information during immobility and play an important role in the generation of sharp-wave ripples. Furthermore, CA2 neurons act to control overall excitability in the hippocampal network and have been found to be consistently altered in psychiatric diseases, indicating that normal function of this region is necessary for normal cognition. With its unique role, area CA2 has a unique molecular profile, interneuron density and composition. Furthermore, this region has an unusual manifestation of synaptic plasticity that does not occur post-synaptically at pyramidal neuron dendrities but through the local network of inhibitory neurons. While much progress has recently been made in understanding the large contribution of area CA2 to social memory formation, much still needs to be learned.
Collapse
|