1
|
Urushihata T, Takahashi M, Shimojo M, Takado Y, Nitta N, Tajima Y, Masamoto K, Kanno I, Tomita Y, Sahara N, Takahashi M, Obata T, Ito H, Yamashita T, Suhara T, Higuchi M, Takuwa H. Neuronal deterioration associated with hyperexcitability under mild chronic cerebral hypoperfusion. J Cereb Blood Flow Metab 2025:271678X251328971. [PMID: 40370304 DOI: 10.1177/0271678x251328971] [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] [Indexed: 05/16/2025]
Abstract
Chronic cerebral hypoperfusion (CCH) has been indicated to impair cognitive and diverse brain functions. However, the neural mechanisms linking these cerebrovascular and phenotypic alterations remain unclear. Here, we investigated the effect of CCH on neuronal activity in male mice with unilateral common carotid artery occlusion using optical imaging and MRI. Our examinations revealed enhanced neuronal activity in concurrence with increased glutamate and tissue acidosis up to seven days after occlusion. At 21-28 days after occlusion, neuronal activity decreased below baseline, while the acidotic but not the hyperglutamatergic state persisted. Notably, pharmacological blockade of the N-methyl-D-aspartate-type glutamate receptor, initiated at an early stage of CCH, suppressed the onset of neuronal hyperexcitation and subsequent deficits in neuronal activity. Altogether, we provide experimental evidence that CCH induces a glutamate surge and results in neuronal hyperexcitation at an early phase, which thereafter gives rise to a non-lethal but progressive deterioration of neuronal functions.
Collapse
Affiliation(s)
- Takuya Urushihata
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, QST, Chiba, Japan
- Department of Integrative Physiology, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Manami Takahashi
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, QST, Chiba, Japan
- Quantum Neuromapping and Neuromodulation Group, Institute for Quantum Life Science, QST, Chiba, Japan
| | - Masafumi Shimojo
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, QST, Chiba, Japan
| | - Yuhei Takado
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, QST, Chiba, Japan
| | - Nobuhiro Nitta
- Applied MRI Research, Department of Molecular Imaging and Theranostics, Institute for Quantum Medical Science, QST, Chiba, Japan
| | - Yosuke Tajima
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, QST, Chiba, Japan
| | - Kazuto Masamoto
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, QST, Chiba, Japan
- Brain Science Inspired Life Support Research Center, University of Electro-Communications, Tokyo, Japan
| | - Iwao Kanno
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, QST, Chiba, Japan
| | - Yutaka Tomita
- Department of Neurology, Keio University School of Medicine, Tokyo, Japan
| | - Naruhiko Sahara
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, QST, Chiba, Japan
| | - Masaya Takahashi
- Department of Radiology, Juntendo University School of Medicine, Tokyo, Japan
| | - Takayuki Obata
- Applied MRI Research, Department of Molecular Imaging and Theranostics, Institute for Quantum Medical Science, QST, Chiba, Japan
| | - Hiroshi Ito
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, QST, Chiba, Japan
- Department of Radiology and Nuclear Medicine, Fukushima Medical University, Fukushima, Japan
| | - Tetsuro Yamashita
- Department of Biological Chemistry, Faculty of Agriculture, Iwate University, Morioka, Japan
| | - Tetsuya Suhara
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, QST, Chiba, Japan
- Quantum Neuromapping and Neuromodulation Group, Institute for Quantum Life Science, QST, Chiba, Japan
| | - Makoto Higuchi
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, QST, Chiba, Japan
| | - Hiroyuki Takuwa
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, QST, Chiba, Japan
- Quantum Neuromapping and Neuromodulation Group, Institute for Quantum Life Science, QST, Chiba, Japan
| |
Collapse
|
2
|
Ziabska K, Gewartowska M, Frontczak-Baniewicz M, Sypecka J, Ziemka-Nalecz M. The Impact of the Histone Deacetylase Inhibitor-Sodium Butyrate on Complement-Mediated Synapse Loss in a Rat Model of Neonatal Hypoxia-Ischemia. Mol Neurobiol 2025; 62:5216-5233. [PMID: 39531190 PMCID: PMC11880148 DOI: 10.1007/s12035-024-04591-w] [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: 07/05/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024]
Abstract
Perinatal asphyxia is one of the most important causes of morbidity and mortality in newborns. One of the key pathogenic factors in hypoxic-ischemic (HI) brain injury is the inflammatory reaction including complement system activation. Over-activated complement stimulates cells to release inflammatory molecules and is involved in the post-ischemic degradation of synaptic connections. On the other hand, complement is also involved in regenerative processes. The histone deacetylase inhibitor (HDACi)-sodium butyrate (SB)-provides reduction of inflammation by decreasing the expression of the proinflammatory factors. The main purpose of this study was to examine the effect of SB treatment on complement activation and synapse elimination after HI. Neonatal HI was induced in Wistar rats pups by unilateral ligation of the common carotid artery followed by 60-min hypoxia (7.6% O2). SB (300 mg/kg) was administered on a 5-day regimen. Our study has shown decreased levels of synapsin I, synaptophysin, and PSD-95 in the hypoxic-ischemic hemisphere, indicating synaptic loss after neonatal HI. Transmission electron microscopy revealed injury of the synaptic structures in the brain after HI. SB treatment increased the level of the synaptic proteins, improved tissue ultrastructure, and reduced degradation of the synapses. Neonatal HI induced mRNA expression of the complement C1q, C3, C5, and C9, and their receptors C3aR and C5aR. The effect of SB was different depending on the time after induction of hypoxic-ischemic damage. Our study demonstrated that neuroprotective effect of SB may be related to the modulation of complement activity after HI brain injury.
Collapse
Affiliation(s)
- Karolina Ziabska
- NeuroRepair Department, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 A. Pawinskiego Street, 02-106, Warsaw, Poland
| | - Magdalena Gewartowska
- Electron Microscopy Research Unit, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 A. Pawinskiego Street, 02-106, Warsaw, Poland
| | - Malgorzata Frontczak-Baniewicz
- Electron Microscopy Research Unit, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 A. Pawinskiego Street, 02-106, Warsaw, Poland
- Higher School of Engineering and Health in Warsaw, 18 Bitwy Warszawskiej 1920r. Street, 02-366, Warsaw, Poland
| | - Joanna Sypecka
- NeuroRepair Department, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 A. Pawinskiego Street, 02-106, Warsaw, Poland
| | - Malgorzata Ziemka-Nalecz
- NeuroRepair Department, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 A. Pawinskiego Street, 02-106, Warsaw, Poland.
| |
Collapse
|
3
|
Pollitt SL, Levy AD, Anderson MC, Blanpied TA. Large Donor CRISPR for Whole-Coding Sequence Replacement of Cell Adhesion Molecule LRRTM2. J Neurosci 2025; 45:e1461242024. [PMID: 39824639 PMCID: PMC11823385 DOI: 10.1523/jneurosci.1461-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 11/01/2024] [Accepted: 12/02/2024] [Indexed: 01/20/2025] Open
Abstract
The cell adhesion molecule leucine-rich repeat transmembrane neuronal protein 2 (LRRTM2) is crucial for synapse development and function. However, our understanding of its endogenous trafficking has been limited due to difficulties in manipulating its coding sequence (CDS) using standard genome editing techniques. Instead, we replaced the entire LRRTM2 CDS by adapting a two-guide CRISPR knock-in method, enabling complete control of LRRTM2. In primary rat hippocampal cultures dissociated from embryos of both sexes, N-terminally tagged, endogenous LRRTM2 was found in 80% of synapses, and synaptic LRRTM2 content correlated with PSD-95 and AMPAR levels. LRRTM2 was also enriched with AMPARs outside synapses, demonstrating the sensitivity of this method to detect relevant new biology. Finally, we leveraged total genomic control to increase the synaptic levels of LRRTM2 via simultaneous mutation of its C-terminal domain, which did not correspondingly increase AMPAR enrichment. The coding region of thousands of genes span lengths suitable for whole-CDS replacement, suggesting this simple approach will enable straightforward structure-function analysis in neurons.
Collapse
Affiliation(s)
- Stephanie L Pollitt
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland 21201-1509
- University of Maryland-Medicine Institute for Neuroscience Discovery (UM-MIND), University of Maryland School of Medicine, Baltimore, Maryland 21201-1509
| | - Aaron D Levy
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland 21201-1509
- University of Maryland-Medicine Institute for Neuroscience Discovery (UM-MIND), University of Maryland School of Medicine, Baltimore, Maryland 21201-1509
| | - Michael C Anderson
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland 21201-1509
- University of Maryland-Medicine Institute for Neuroscience Discovery (UM-MIND), University of Maryland School of Medicine, Baltimore, Maryland 21201-1509
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland 21201-1509
| | - Thomas A Blanpied
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland 21201-1509
- University of Maryland-Medicine Institute for Neuroscience Discovery (UM-MIND), University of Maryland School of Medicine, Baltimore, Maryland 21201-1509
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland 21201-1509
| |
Collapse
|
4
|
Hoisington ZW, Gangal H, Phamluong K, Shukla C, Ehinger Y, Moffat JJ, Homanics GE, Wang J, Ron D. Prosapip1 in the dorsal hippocampus mediates synaptic protein composition, long-term potentiation, and spatial memory. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.06.13.597459. [PMID: 38915579 PMCID: PMC11195216 DOI: 10.1101/2024.06.13.597459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Prosapip1 is a brain-specific protein localized to the postsynaptic density, where it promotes dendritic spine maturation in primary hippocampal neurons. However, nothing is known about the role of Prosapip1 in vivo. To examine this, we utilized the Cre-loxP system to develop a Prosapip1 neuronal knockout mouse. We found that Prosapip1 controls the synaptic localization of its binding partner SPAR, along with PSD-95 and the GluN2B subunit of the NMDA receptor (NMDAR) in the dorsal hippocampus (dHP). We next sought to identify the potential contribution of Prosapip1 to the activity and function of the NMDAR and found that Prosapip1 plays an important role in NMDAR-mediated transmission and long-term potentiation (LTP) in the CA1 region of the dHP. As LTP is the cellular hallmark of learning and memory, we examined the consequences of neuronal knockout of Prosapip1 on dHP-dependent memory. We found that global or dHP-specific neuronal knockout of Prosapip1 caused a deficit in learning and memory whereas developmental, locomotor, and anxiety phenotypes were normal. Taken together, Prosapip1 in the dHP promotes the proper localization of synaptic proteins which, in turn, facilitates LTP driving recognition, social, and spatial learning and memory.
Collapse
|
5
|
Abstract
In animals, memory formation and recall are essential for their survival and for adaptations to a complex and often dynamically changing environment. During memory formation, experiences prompt the activation of a selected and sparse population of cells (engram cells) that undergo persistent physical and/or chemical changes allowing long-term memory formation, which can last for decades. Over the past few decades, important progress has been made on elucidating signaling mechanisms by which synaptic transmission leads to the induction of activity-dependent gene regulation programs during the different phases of learning (acquisition, consolidation, and recall). But what are the molecular mechanisms that govern the expression of immediate-early genes (IEGs; c-fos, Npas4) and plasticity-related genes (PRGs; Dlg4/PSD95 and Grin2b/NR2B) in memory ensemble? Studies in relatively simple in vitro and in vivo neuronal model systems have demonstrated that synaptic activity during development, or when induced by chemical stimuli (i.e., cLTP, KCl, picrotoxin), activates the NMDAR-Ca2+-CREB signaling pathway that upregulates gene expression through changes in the epigenetic landscape (i.e., histone marks and DNA methylation) and/or 3D chromatin organization. The data support a model in which epigenetic modifications in promoters and enhancers facilitate the priming and activation of these regulatory regions, hence leading to the formation of enhancer-promoter interactions (EPIs) through chromatin looping. The exploration of whether similar molecular mechanisms drive gene expression in learning and memory has presented notable challenges due to the distinct phases of learning and the activation of only sparse population of cells (the engram). Consequently, such studies demand precise temporal and spatial control. By combining activity-dependent engram tagging strategies (i.e., TRAP mice) with multi-omics analyses (i.e., RNA-seq, ChiP-seq, ATAC-seq, and Hi-C), it has been recently possible to associate changes in the epigenomic landscape and/or 3D genome architecture with transcriptional waves in engram cells of mice subjected to contextual fear conditioning (CFC), a relevant one-shot Pavlovian learning task. These studies support the role of specific epigenetic mechanisms and of the 3D chromatin organization during the control of gene transcription waves in engram cells. Advancements in our comprehension of the molecular mechanisms driving memory ensemble will undoubtedly play a crucial role in the development of better-targeted strategies to tackle cognitive diseases, including Alzheimer's disease and frontotemporal dementia, among other information-processing disorders.
Collapse
Affiliation(s)
- Brigitte van Zundert
- Faculty of Medicine and Faculty of Life Sciences, Institute of Biomedical Sciences (ICB), Universidad Andres Bello, Santiago, Chile.
- Millennium Nucleus of Neuroepigenetics and Plasticity (EpiNeuro), Santiago, Chile.
- Department of Neurology, University of Massachusetts Chan Medical School (UMMS), Worcester, MA, USA.
| | - Martin Montecino
- Faculty of Medicine and Faculty of Life Sciences, Institute of Biomedical Sciences (ICB), Universidad Andres Bello, Santiago, Chile.
- Millennium Nucleus of Neuroepigenetics and Plasticity (EpiNeuro), Santiago, Chile.
| |
Collapse
|
6
|
Vinci E, Beretta S, Colombo V, Zippo A, Catanese A, Wiegreffe C, Britsch S, Boeckers T, Verpelli C, Sala C. Regulation of Dendrite and Dendritic Spine Formation by TCF20. J Neurochem 2025; 169:e16297. [PMID: 39801227 PMCID: PMC11725998 DOI: 10.1111/jnc.16297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 10/24/2024] [Accepted: 12/11/2024] [Indexed: 01/16/2025]
Abstract
Mutations in the Transcription Factor 20 (TCF20) have been identified in patients with autism spectrum disorders (ASDs), intellectual disabilities (IDs), and other neurological issues. Recently, a new syndrome called TCF20-associated neurodevelopmental disorders (TAND) has been described, with specific clinical features. While TCF20's role in the neurogenesis of mouse embryos has been reported, little is known about its molecular function in neurons. In this study, we demonstrate that TCF20 is expressed in all analyzed brain regions in mice, and its expression increases during brain development but decreases in muscle tissue. Our findings suggest that TCF20 plays a central role in dendritic arborization and dendritic spine formation processes. RNA sequencing analysis revealed a downregulation of pre- and postsynaptic pathways in TCF20 knockdown neurons. We also found decreased levels of GABRA1, BDNF, PSD-95, and c-Fos in total homogenates and in synaptosomal preparations of knockdown TCF20 rat cortical cultures. Furthermore, synaptosomal preparations of knockdown TCF20 rat cortical cultures showed significant downregulation of GluN2B and GABRA5, while GluA2 was significantly upregulated. Overall, our data suggest that TCF20 plays an essential role in neuronal development and function by modulating the expression of proteins involved in dendrite and synapse formation and function.
Collapse
Affiliation(s)
- Ersilia Vinci
- CNR Neuroscience Institute, MilanoVedano al LambroItaly
| | | | | | - Antonio Zippo
- CNR Neuroscience Institute, MilanoVedano al LambroItaly
| | - Alberto Catanese
- Institute of Anatomy and Cell BiologyUniversity of UlmUlmGermany
- German Center for Neurodegenerative Diseases (DZNE)UlmGermany
| | | | - Stefan Britsch
- Institute of Molecular and Cellular AnatomyUniversity of UlmUlmGermany
| | - Tobias Boeckers
- Institute of Anatomy and Cell BiologyUniversity of UlmUlmGermany
- German Center for Neurodegenerative Diseases (DZNE)UlmGermany
| | | | - Carlo Sala
- CNR Neuroscience Institute, MilanoVedano al LambroItaly
| |
Collapse
|
7
|
Chimura T, Manabe T. Ca2+-PP2B-PSD-95 axis: A novel regulatory mechanism of the phosphorylation state of Serine 295 of PSD-95. PLoS One 2024; 19:e0313441. [PMID: 39509447 PMCID: PMC11542788 DOI: 10.1371/journal.pone.0313441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 10/23/2024] [Indexed: 11/15/2024] Open
Abstract
The phosphorylation state of PSD-95 at Serine 295 (Ser295) is important for the regulation of synaptic plasticity. Although the activation of NMDA receptors (NMDARs), which initiates an intracellular calcium signaling cascade, decreases phosphorylated Ser295 (pS295) of PSD-95, the molecular mechanisms are not fully understood. We found that the calcium-activated protein phosphatase PP2B dephosphorylated pS295 not only in basal conditions but also in NMDAR-activated conditions in cultured neurons. The biochemical assay also revealed the dephosphorylation of pS295 by PP2B, consistently supporting the results obtained using neurons. The newly identified calcium signaling cascade "Ca2+-PP2B-PSD-95 axis" would play an important role in the molecular mechanism for NMDA receptor-dependent plasticity.
Collapse
Affiliation(s)
- Takahiko Chimura
- Department of Basic Medical Sciences, Institute of Medical Science, Division of Neuronal Network, University of Tokyo, Tokyo, Japan
| | - Toshiya Manabe
- Department of Basic Medical Sciences, Institute of Medical Science, Division of Neuronal Network, University of Tokyo, Tokyo, Japan
| |
Collapse
|
8
|
Brauer B, Ancatén-González C, Ahumada-Marchant C, Meza RC, Merino-Veliz N, Nardocci G, Varela-Nallar L, Arriagada G, Chávez AE, Bustos FJ. Impact of KDM6B mosaic brain knockout on synaptic function and behavior. Sci Rep 2024; 14:20416. [PMID: 39223259 PMCID: PMC11369245 DOI: 10.1038/s41598-024-70728-5] [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: 04/03/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024] Open
Abstract
Autism spectrum disorders (ASD) are complex neurodevelopmental conditions characterized by impairments in social communication, repetitive behaviors, and restricted interests. Epigenetic modifications serve as critical regulators of gene expression playing a crucial role in controlling brain function and behavior. Lysine (K)-specific demethylase 6B (KDM6B), a stress-inducible H3K27me3 demethylase, has emerged as one of the highest ASD risk genes, but the precise effects of KDM6B mutations on neuronal activity and behavioral function remain elusive. Here we show the impact of KDM6B mosaic brain knockout on the manifestation of different autistic-like phenotypes including repetitive behaviors, social interaction, and significant cognitive deficits. Moreover, KDM6B mosaic knockout display abnormalities in hippocampal excitatory synaptic transmission decreasing NMDA receptor mediated synaptic transmission and plasticity. Understanding the intricate interplay between epigenetic modifications and neuronal function may provide novel insights into the pathophysiology of ASD and potentially inform the development of targeted therapeutic interventions.
Collapse
Affiliation(s)
- Bastian Brauer
- Constantine-Paton Research Laboratory, Instituto de Ciencias Biomédicas, Facultad de Medicina y Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
| | - Carlos Ancatén-González
- Programa de Doctorado en Ciencias, Mención Neurociencia, Universidad de Valparaíso, Valparaíso, Chile
- Instituto de Neurociencias, Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Facultad de Ciencias, Universidad de Valparaíso, 2340000, Valparaiso, Chile
| | - Constanza Ahumada-Marchant
- Constantine-Paton Research Laboratory, Instituto de Ciencias Biomédicas, Facultad de Medicina y Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
| | - Rodrigo C Meza
- Instituto de Neurociencias, Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Facultad de Ciencias, Universidad de Valparaíso, 2340000, Valparaiso, Chile
| | - Nicolas Merino-Veliz
- Constantine-Paton Research Laboratory, Instituto de Ciencias Biomédicas, Facultad de Medicina y Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
| | - Gino Nardocci
- School of Medicine, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
- Molecular Biology and Bioinformatics Lab, Program in Molecular Biology and Bioinformatics, Center for Biomedical Research and Innovation (CIIB), Universidad de Los Andes, Santiago, Chile
| | - Lorena Varela-Nallar
- Constantine-Paton Research Laboratory, Instituto de Ciencias Biomédicas, Facultad de Medicina y Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
- Millennium Nucleus of Neuroepigenetics and Plasticity (EpiNeuro), Santiago, Chile
| | - Gloria Arriagada
- Constantine-Paton Research Laboratory, Instituto de Ciencias Biomédicas, Facultad de Medicina y Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
| | - Andrés E Chávez
- Instituto de Neurociencias, Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Facultad de Ciencias, Universidad de Valparaíso, 2340000, Valparaiso, Chile.
| | - Fernando J Bustos
- Constantine-Paton Research Laboratory, Instituto de Ciencias Biomédicas, Facultad de Medicina y Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile.
- Millennium Nucleus of Neuroepigenetics and Plasticity (EpiNeuro), Santiago, Chile.
| |
Collapse
|
9
|
Fernandes EFA, Palner M, Raval NR, Jeppesen TE, Danková D, Bærentzen SL, Werner C, Eilts J, Maric HM, Doose S, Aripaka SS, Kaalund SS, Aznar S, Kjaer A, Schlosser A, Haugaard-Kedström LM, Knudsen GM, Herth MM, Stro Mgaard K. Development of Peptide-Based Probes for Molecular Imaging of the Postsynaptic Density in the Brain. J Med Chem 2024; 67:11975-11988. [PMID: 38981131 DOI: 10.1021/acs.jmedchem.4c00615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/11/2024]
Abstract
The postsynaptic density (PSD) comprises numerous scaffolding proteins, receptors, and signaling molecules that coordinate synaptic transmission in the brain. Postsynaptic density protein 95 (PSD-95) is a master scaffold protein within the PSD and one of its most abundant proteins and therefore constitutes a very attractive biomarker of PSD function and its pathological changes. Here, we exploit a high-affinity inhibitor of PSD-95, AVLX-144, as a template for developing probes for molecular imaging of the PSD. AVLX-144-based probes were labeled with the radioisotopes fluorine-18 and tritium, as well as a fluorescent tag. Tracer binding showed saturable, displaceable, and uneven distribution in rat brain slices, proving effective in quantitative autoradiography and cell imaging studies. Notably, we observed diminished tracer binding in human post-mortem Parkinson's disease (PD) brain slices, suggesting postsynaptic impairment in PD. We thus offer a suite of translational probes for visualizing and understanding PSD-related pathologies.
Collapse
Affiliation(s)
- Eduardo F A Fernandes
- Center for Biopharmaceuticals, Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, Copenhagen DK-2100, Denmark
| | - Mikael Palner
- Neurobiology Research Unit, Rigshospitalet, Blegdamsvej 9, Copenhagen DK-2100, Denmark
| | - Nakul Ravi Raval
- Neurobiology Research Unit, Rigshospitalet, Blegdamsvej 9, Copenhagen DK-2100, Denmark
- Department of Clinical Medicine, University of Copenhagen, Blegdamsvej 3, Copenhagen DK-2200, Denmark
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut 06520, United States
| | - Troels E Jeppesen
- Department of Clinical Physiology and Nuclear Medicine & Cluster for Molecular Imaging, Copenhagen University Hospital - Rigshospitalet & Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, Copenhagen DK-2200, Denmark
| | - Daniela Danková
- Center for Biopharmaceuticals, Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, Copenhagen DK-2100, Denmark
| | - Simone L Bærentzen
- Neurobiology Research Unit, Rigshospitalet, Blegdamsvej 9, Copenhagen DK-2100, Denmark
| | - Christian Werner
- Department of Biotechnology and Biophysics, Biocenter, Julius-Maximilians-University, Am Hubland, Würzburg D-97074, Germany
| | - Janna Eilts
- Department of Biotechnology and Biophysics, Biocenter, Julius-Maximilians-University, Am Hubland, Würzburg D-97074, Germany
| | - Hans M Maric
- Center for Biopharmaceuticals, Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, Copenhagen DK-2100, Denmark
- Rudolf Virchow Center for Integrative and Translational Bioimaging, Julius-Maximilians-University, Josef-Schneider-Str. 2, Würzburg 97080, Germany
| | - Sören Doose
- Department of Biotechnology and Biophysics, Biocenter, Julius-Maximilians-University, Am Hubland, Würzburg D-97074, Germany
| | - Sanjay Sagar Aripaka
- Neurobiology Research Unit, Rigshospitalet, Blegdamsvej 9, Copenhagen DK-2100, Denmark
| | - Sanne Simone Kaalund
- Center for Neuroscience and Stereology, Bispebjerg University Hospital, Nielsine Nielsens Vej 6B, Copenhagen DK-2400, Denmark
| | - Susana Aznar
- Center for Neuroscience and Stereology, Bispebjerg University Hospital, Nielsine Nielsens Vej 6B, Copenhagen DK-2400, Denmark
- Center for Translational Research, Bispebjerg University Hospital, Nielsine Nielsens Vej 4B, Copenhagen DK-2400, Denmark
| | - Andreas Kjaer
- Department of Clinical Medicine, University of Copenhagen, Blegdamsvej 3, Copenhagen DK-2200, Denmark
- Department of Clinical Physiology and Nuclear Medicine & Cluster for Molecular Imaging, Copenhagen University Hospital - Rigshospitalet & Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, Copenhagen DK-2200, Denmark
| | - Andreas Schlosser
- Department of Biotechnology and Biophysics, Biocenter, Julius-Maximilians-University, Am Hubland, Würzburg D-97074, Germany
| | - Linda M Haugaard-Kedström
- Center for Biopharmaceuticals, Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, Copenhagen DK-2100, Denmark
| | - Gitte M Knudsen
- Neurobiology Research Unit, Rigshospitalet, Blegdamsvej 9, Copenhagen DK-2100, Denmark
- Department of Clinical Medicine, University of Copenhagen, Blegdamsvej 3, Copenhagen DK-2200, Denmark
| | - Matthias M Herth
- Center for Biopharmaceuticals, Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, Copenhagen DK-2100, Denmark
- Neurobiology Research Unit, Rigshospitalet, Blegdamsvej 9, Copenhagen DK-2100, Denmark
- Department of Clinical Physiology and Nuclear Medicine & Cluster for Molecular Imaging, Copenhagen University Hospital - Rigshospitalet & Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, Copenhagen DK-2200, Denmark
| | - Kristian Stro Mgaard
- Center for Biopharmaceuticals, Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, Copenhagen DK-2100, Denmark
| |
Collapse
|
10
|
Metzbower SR, Levy AD, Dharmasri PA, Anderson MC, Blanpied TA. Distinct SAP102 and PSD-95 Nano-organization Defines Multiple Types of Synaptic Scaffold Protein Domains at Single Synapses. J Neurosci 2024; 44:e1715232024. [PMID: 38777601 PMCID: PMC11211720 DOI: 10.1523/jneurosci.1715-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 04/30/2024] [Accepted: 05/05/2024] [Indexed: 05/25/2024] Open
Abstract
MAGUK scaffold proteins play a central role in maintaining and modulating synaptic signaling, providing a framework to retain and position receptors, signaling molecules, and other synaptic components. In particular, the MAGUKs SAP102 and PSD-95 are essential for synaptic function at distinct developmental timepoints and perform both overlapping and unique roles. While their similar structures allow for common binding partners, SAP102 is expressed earlier in synapse development and is required for synaptogenesis, whereas PSD-95 expression peaks later and is associated with synapse maturation. PSD-95 and other key synaptic proteins organize into subsynaptic nanodomains that have a significant impact on synaptic transmission, but the nanoscale organization of SAP102 is unknown. How SAP102 is organized within the synapse, and how it relates spatially to PSD-95 on a nanometer scale, could underlie its unique functions and impact how SAP102 scaffolds synaptic proteins. Here we used DNA-PAINT super-resolution microscopy to measure SAP102 nano-organization and its spatial relationship to PSD-95 at individual synapses in mixed-sex rat cultured neurons. We found that like PSD-95, SAP102 accumulates in high-density subsynaptic nanoclusters (NCs). However, SAP102 NCs were smaller and denser than PSD-95 NCs across development. Additionally, only a subset of SAP102 NCs co-organized with PSD-95, revealing MAGUK nanodomains within individual synapses containing either one or both proteins. These MAGUK nanodomain types had distinct NC properties and were differentially enriched with the presynaptic release protein Munc13-1. This organization into both shared and distinct subsynaptic nanodomains may underlie the ability of SAP102 and PSD-95 to perform both common and unique synaptic functions.
Collapse
Affiliation(s)
- Sarah R Metzbower
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Aaron D Levy
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Poorna A Dharmasri
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland 21201
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Michael C Anderson
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland 21201
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Thomas A Blanpied
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland 21201
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland 21201
- University of Maryland Medicine Institute for Neuroscience Discovery, University of Maryland School of Medicine, Baltimore, Maryland 21201
| |
Collapse
|
11
|
Chen Y, Liu S, Jacobi AA, Jeng G, Ulrich JD, Stein IS, Patriarchi T, Hell JW. Rapid sequential clustering of NMDARs, CaMKII, and AMPARs upon activation of NMDARs at developing synapses. Front Synaptic Neurosci 2024; 16:1291262. [PMID: 38660466 PMCID: PMC11039796 DOI: 10.3389/fnsyn.2024.1291262] [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: 09/08/2023] [Accepted: 03/22/2024] [Indexed: 04/26/2024] Open
Abstract
Rapid, synapse-specific neurotransmission requires the precise alignment of presynaptic neurotransmitter release and postsynaptic receptors. How postsynaptic glutamate receptor accumulation is induced during maturation is not well understood. We find that in cultures of dissociated hippocampal neurons at 11 days in vitro (DIV) numerous synaptic contacts already exhibit pronounced accumulations of the pre- and postsynaptic markers synaptotagmin, synaptophysin, synapsin, bassoon, VGluT1, PSD-95, and Shank. The presence of an initial set of AMPARs and NMDARs is indicated by miniature excitatory postsynaptic currents (mEPSCs). However, AMPAR and NMDAR immunostainings reveal rather smooth distributions throughout dendrites and synaptic enrichment is not obvious. We found that brief periods of Ca2+ influx through NMDARs induced a surprisingly rapid accumulation of NMDARs within 1 min, followed by accumulation of CaMKII and then AMPARs within 2-5 min. Postsynaptic clustering of NMDARs and AMPARs was paralleled by an increase in their mEPSC amplitudes. A peptide that blocked the interaction of NMDAR subunits with PSD-95 prevented the NMDAR clustering. NMDAR clustering persisted for 3 days indicating that brief periods of elevated glutamate fosters permanent accumulation of NMDARs at postsynaptic sites in maturing synapses. These data support the model that strong glutamatergic stimulation of immature glutamatergic synapses results in a fast and substantial increase in postsynaptic NMDAR content that required NMDAR binding to PSD-95 or its homologues and is followed by recruitment of CaMKII and subsequently AMPARs.
Collapse
Affiliation(s)
- Yucui Chen
- Department of Pharmacology, University of Iowa, Iowa City, IA, United States
| | - Shangming Liu
- Department of Pharmacology, University of California, Davis, Davis, CA, United States
| | - Ariel A. Jacobi
- Department of Pharmacology, University of California, Davis, Davis, CA, United States
| | - Grace Jeng
- Department of Pharmacology, University of California, Davis, Davis, CA, United States
| | - Jason D. Ulrich
- Department of Pharmacology, University of Iowa, Iowa City, IA, United States
| | - Ivar S. Stein
- Department of Pharmacology, University of Iowa, Iowa City, IA, United States
- Department of Pharmacology, University of California, Davis, Davis, CA, United States
| | - Tommaso Patriarchi
- Department of Pharmacology, University of California, Davis, Davis, CA, United States
| | - Johannes W. Hell
- Department of Pharmacology, University of Iowa, Iowa City, IA, United States
- Department of Pharmacology, University of California, Davis, Davis, CA, United States
| |
Collapse
|
12
|
Peng X, Chen P, Zhang Y, Wu K, Ji N, Gao J, Wang H, Zhang Y, Xu T, Hua R. MPP2 interacts with SK2 to rescue the excitability of glutamatergic neurons in the BLA and facilitate the extinction of conditioned fear in mice. CNS Neurosci Ther 2024; 30:e14362. [PMID: 37469037 PMCID: PMC10805397 DOI: 10.1111/cns.14362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 05/29/2023] [Accepted: 07/03/2023] [Indexed: 07/21/2023] Open
Abstract
AIMS The basolateral amygdala (BLA) plays an integral role in anxiety disorders (such as post traumatic stress disorder) stem from dysregulated fear memory. The excitability of glutamatergic neurons in the BLA correlates with fear memory, and the afterhyperpolarization current (IAHP ) mediated by small-conductance calcium-activated potassium channel subtype 2 (SK2) dominates the excitability of glutamatergicneurons. This study aimed to explore the effect of MPP2 interacts with SK2 in the excitability of glutamatergic neurons in the BLA and the extinction of conditioned fear in mice. METHODS Fear memory was analyzed via freezing percentage. Western blotting and fluorescence quantitative PCR were used to determine the expression of protein and mRNA respectively. Electrophysiology was employed to measure the excitability of glutamatergic neurons and IAHP . RESULTS Fear conditioning decreased the levels of synaptic SK2 channels in the BLA, which were restored following fear extinction. Notably, reduced expression of synaptic SK2 channels in the BLA during fear conditioning was caused by the increased activity of protein kinase A (PKA), while increased levels of synaptic SK2 channels in the BLA during fear extinction were mediated by interactions with membrane-palmitoylated protein 2 (MPP2). CONCLUSIONS Our results revealed that MPP2 interacts with the SK2 channels and rescues the excitability of glutamatergic neurons by increasing the expression of synaptic SK2 channels in the BLA to promote the normalization of anxiety disorders and provide a new direction for the treatment.
Collapse
Affiliation(s)
- Xiaohan Peng
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic DrugsXuzhou Medical UniversityXuzhouChina
- Jiangsu Province Key Laboratory of AnesthesiologyXuzhou Medical UniversityXuzhouChina
| | - Panpan Chen
- Jiangsu Province Key Laboratory of AnesthesiologyXuzhou Medical UniversityXuzhouChina
- Anesthesiology DepartmentJiangsu Province HospitalNanjingChina
| | - Yang Zhang
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic DrugsXuzhou Medical UniversityXuzhouChina
- Jiangsu Province Key Laboratory of AnesthesiologyXuzhou Medical UniversityXuzhouChina
| | - Ke Wu
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic DrugsXuzhou Medical UniversityXuzhouChina
- Jiangsu Province Key Laboratory of AnesthesiologyXuzhou Medical UniversityXuzhouChina
| | - Ningning Ji
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic DrugsXuzhou Medical UniversityXuzhouChina
- Jiangsu Province Key Laboratory of AnesthesiologyXuzhou Medical UniversityXuzhouChina
| | - Jinghua Gao
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic DrugsXuzhou Medical UniversityXuzhouChina
- Jiangsu Province Key Laboratory of AnesthesiologyXuzhou Medical UniversityXuzhouChina
| | - Hui Wang
- Jiangsu Province Key Laboratory of AnesthesiologyXuzhou Medical UniversityXuzhouChina
| | - Yong‐mei Zhang
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic DrugsXuzhou Medical UniversityXuzhouChina
- Jiangsu Province Key Laboratory of AnesthesiologyXuzhou Medical UniversityXuzhouChina
| | - Tie Xu
- Emergency Medicine DepartmentThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouChina
| | - Rong Hua
- Emergency Medicine DepartmentThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouChina
| |
Collapse
|
13
|
Griesius S, Waldron S, Kamenish KA, Cherbanich N, Wilkinson LS, Thomas KL, Hall J, Mellor JR, Dwyer DM, Robinson ESJ. A mild impairment in reversal learning in a bowl-digging substrate deterministic task but not other cognitive tests in the Dlg2+/- rat model of genetic risk for psychiatric disorder. GENES, BRAIN, AND BEHAVIOR 2023; 22:e12865. [PMID: 37705179 PMCID: PMC10733576 DOI: 10.1111/gbb.12865] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 08/22/2023] [Accepted: 08/25/2023] [Indexed: 09/15/2023]
Abstract
Variations in the Dlg2 gene have been linked to increased risk for psychiatric disorders, including schizophrenia, autism spectrum disorders, intellectual disability, bipolar disorder, attention deficit hyperactivity disorder, and pubertal disorders. Recent studies have reported disrupted brain circuit function and behaviour in models of Dlg2 knockout and haploinsufficiency. Specifically, deficits in hippocampal synaptic plasticity were found in heterozygous Dlg2+/- rats suggesting impacts on hippocampal dependent learning and cognitive flexibility. Here, we tested these predicted effects with a behavioural characterisation of the heterozygous Dlg2+/- rat model. Dlg2+/- rats exhibited a specific, mild impairment in reversal learning in a substrate deterministic bowl-digging reversal learning task. The performance of Dlg2+/- rats in other bowl digging task, visual discrimination and reversal, novel object preference, novel location preference, spontaneous alternation, modified progressive ratio, and novelty-suppressed feeding test were not impaired. These findings suggest that despite altered brain circuit function, behaviour across different domains is relatively intact in Dlg2+/- rats, with the deficits being specific to only one test of cognitive flexibility. The specific behavioural phenotype seen in this Dlg2+/- model may capture features of the clinical presentation associated with variation in the Dlg2 gene.
Collapse
Affiliation(s)
- Simonas Griesius
- Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, University WalkBristolUK
| | - Sophie Waldron
- Neuroscience and Mental Health Research Institute, PsychologyCardiffUK
- Department of PsychologyCardiffUK
| | - Katie A. Kamenish
- Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, University WalkBristolUK
| | - Nick Cherbanich
- Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, University WalkBristolUK
| | - Lawrence S. Wilkinson
- Neuroscience and Mental Health Research Institute, PsychologyCardiffUK
- Department of PsychologyCardiffUK
- MRC Centre for Neuropsychiatric Genetics and Genomics, Schools of Medicine and Genetics and Genomics, Schools of Medicine and PsychologyCardiffUK
| | - Kerrie L. Thomas
- Neuroscience and Mental Health Research Institute, PsychologyCardiffUK
- Department of Medicine and PsychologyCardiffUK
| | - Jeremy Hall
- Neuroscience and Mental Health Research Institute, PsychologyCardiffUK
- MRC Centre for Neuropsychiatric Genetics and Genomics, Schools of Medicine and Genetics and Genomics, Schools of Medicine and PsychologyCardiffUK
- Department of Medicine and PsychologyCardiffUK
| | - Jack R. Mellor
- Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, University WalkBristolUK
| | - Dominic M. Dwyer
- Neuroscience and Mental Health Research Institute, PsychologyCardiffUK
- Department of PsychologyCardiffUK
| | - Emma S. J. Robinson
- Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, University WalkBristolUK
| |
Collapse
|
14
|
Chuang KF, Wang CH, Chen HK, Lin YY, Lin CH, Lin YC, Shih CP, Kuo CY, Chen YC, Chen HC. GRAIL gene knockout mice protect against aging-related and noise-induced hearing loss. J Chin Med Assoc 2023; 86:1101-1108. [PMID: 37820291 DOI: 10.1097/jcma.0000000000001005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/13/2023] Open
Abstract
BACKGROUND Hearing loss is a global health issue and its etiopathologies involve complex molecular pathways. The ubiquitin-proteasome system has been reported to be associated with cochlear development and hearing loss. The gene related to anergy in lymphocytes ( GRAIL ), as an E3 ubiquitin ligase, has not, as yet, been examined in aging-related and noise-induced hearing loss mice models. METHODS This study used wild-type (WT) and GRAIL knockout (KO) mice to examine cochlear hair cells and synaptic ribbons using immunofluorescence staining. The hearing in WT and KO mice was detected using auditory brainstem response. Gene expression patterns were compared using RNA-sequencing to identify potential targets during the pathogenesis of noise-induced hearing loss in WT and KO mice. RESULTS At the 12-month follow-up, GRAIL KO mice had significantly less elevation in threshold level and immunofluorescence staining showed less loss of outer hair cells and synaptic ribbons in the hook region compared with GRAIL WT mice. At days 1, 14, and 28 after noise exposure, GRAIL KO mice had significantly less elevation in threshold level than WT mice. After noise exposure, GRAIL KO mice showed less loss of outer hair cells in the cochlear hook and basal regions compared with WT mice. Moreover, immunofluorescence staining showed less loss of synaptic ribbons in the hook regions of GRAIL KO mice than of WT mice. RNA-seq analysis results showed significant differences in C-C motif chemokine ligand 19 ( CCL19 ), C-C motif chemokine ligand 21 ( CCL21 ), interleukin 25 ( IL25 ), glutathione peroxidase 6 ( GPX6 ), and nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 1 ( NOX1 ) genes after noise exposure. CONCLUSION The present data demonstrated that GRAIL deficiency protects against aging-related and noise-induced hearing loss. The mechanism involved needs to be further clarified from the potential association with synaptic modulation, inflammation, and oxidative stress.
Collapse
Affiliation(s)
- Kai-Fen Chuang
- Department of Otolaryngology-Head and Neck Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Chih-Hung Wang
- Department of Otolaryngology-Head and Neck Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan, ROC
- Division of Otolaryngology, Taipei Veterans General Hospital, Taoyuan Branch, Taoyuan, Taiwan
| | - Hang-Kang Chen
- Department of Otolaryngology-Head and Neck Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Yuan-Yung Lin
- Department of Otolaryngology-Head and Neck Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Chia-Hsin Lin
- Department of Otolaryngology-Head and Neck Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Yi-Chun Lin
- Department of Otolaryngology-Head and Neck Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Cheng-Ping Shih
- Department of Otolaryngology-Head and Neck Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Chao-Yin Kuo
- Department of Otolaryngology-Head and Neck Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Ying-Chuan Chen
- Department of Physiology & Biophysics, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Hsin-Chien Chen
- Department of Otolaryngology-Head and Neck Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
| |
Collapse
|
15
|
Zhu ML, Zhang J, Guo LJ, Yue RZ, Li SS, Cui BY, Guo S, Niu QQ, Yu YN, Wang HH, Yang L, Yin YL, Wang SX, Zhan HQ, Gao ZT, Li P. Amorphous selenium inhibits oxidative stress injury of neurons in vascular dementia rats by activating NMDAR pathway. Eur J Pharmacol 2023; 955:175874. [PMID: 37394029 DOI: 10.1016/j.ejphar.2023.175874] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 06/11/2023] [Accepted: 06/20/2023] [Indexed: 07/04/2023]
Abstract
Vascular dementia (VD) is one of the most common causes of dementia, taking account for about 20% of all cases. Although studies have found that selenium supplementation can improve the cognitive ability of Alzheimer's patients, there is currently no research on the cognitive impairment caused by VD. This study aimed to investigate the role and mechanism of Amorphous selenium nanodots (A SeNDs) in the prevention of VD. The bilateral common carotid artery occlusion (BCCAO) method was used to establish a VD model. The neuroprotective effect of A SeNDs was evaluated by Morris water maze, Transcranial Doppler TCD, hematoxylin-eosin (HE) staining, Neuron-specific nuclear protein (Neu N) staining and Golgi staining. Detect the expression levels of oxidative stress and Calcium-calmodulin dependent protein kinase II (CaMK II), N-methyl-D-aspartate receptor subunit NR2A, and postsynaptic dense protein 95 (PSD95). Finally, measure the concentration of calcium ions in neuronal cells. The results showed that A SeNDs could significantly improve the learning and memory ability of VD rats, restore the posterior arterial blood flow of the brain, improve the neuronal morphology and dendritic remodeling of pyramidal cells in hippocampal CA1 area, reduce the level of oxidative stress in VD rats, increase the expression of NR2A, PSD95, CaMK II proteins and reduce intracellular calcium ion concentration, but the addition of selective NR2A antagonist NVP-AAMO77 eliminated these benefits. It suggests that A SeNDs may improve cognitive dysfunction in vascular dementia rats by regulating the NMDAR pathway.
Collapse
Affiliation(s)
- Mo-Li Zhu
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, College of Pharmacy, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
| | - Jie Zhang
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, College of Pharmacy, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
| | - Li-Juan Guo
- Department of Oncology, First Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453119, China
| | - Rui-Zhu Yue
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, College of Pharmacy, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
| | - Shan-Shan Li
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, College of Pharmacy, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
| | - Bao-Yue Cui
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, College of Pharmacy, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
| | - Shuang Guo
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, Xianning, 437100, China
| | - Qian-Qian Niu
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, College of Pharmacy, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
| | - Ya-Nan Yu
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, College of Pharmacy, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
| | - Huan-Huan Wang
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, College of Pharmacy, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
| | - Lin Yang
- Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals, Key Laboratory of Green Chemical Media and Reactions, Ministry of Education, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, 453007, China
| | - Ya-Ling Yin
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, College of Pharmacy, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China.
| | - Shuang-Xi Wang
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, College of Pharmacy, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China.
| | - He-Qin Zhan
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, College of Pharmacy, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China.
| | - Zhi-Tao Gao
- School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003, China.
| | - Peng Li
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, College of Pharmacy, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China; Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, Xianning, 437100, China.
| |
Collapse
|
16
|
Park J, Berthoux C, Hoyos-Ramirez E, Shan L, Morimoto-Tomita M, Wang Y, Castillo PE, Tomita S. Chemogenetic regulation of the TARP-lipid interaction mimics LTP and reversibly modifies behavior. Cell Rep 2023; 42:112826. [PMID: 37471228 PMCID: PMC10528344 DOI: 10.1016/j.celrep.2023.112826] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 04/10/2023] [Accepted: 07/02/2023] [Indexed: 07/22/2023] Open
Abstract
Long-term potentiation (LTP), a well-characterized form of synaptic plasticity, is believed to underlie memory formation. Hebbian, postsynaptically expressed LTP requires TARPγ-8 phosphorylation for synaptic insertion of AMPA receptors (AMPARs). However, it is unknown whether TARP-mediated AMPAR insertion alone is sufficient to modify behavior. Here, we report the development of a chemogenetic tool, ExSYTE (Excitatory SYnaptic Transmission modulator by Engineered TARPγ-8), to mimic the cytoplasmic interaction of TARP with the plasma membrane in a doxycycline-dependent manner. We use this tool to examine the specific role of synaptic AMPAR potentiation in amygdala neurons that are activated by fear conditioning. Selective expression of active ExSYTE in these neurons potentiates AMPAR-mediated synaptic transmission in a doxycycline-dependent manner, occludes synaptically induced LTP, and mimics freezing triggered by cued fear conditioning. Thus, chemogenetic controlling of the TARP-membrane interaction is sufficient for LTP-like synaptic AMPAR insertion, which mimics fear conditioning.
Collapse
Affiliation(s)
- Joongkyu Park
- Department of Cellular and Molecular Physiology, Department of Neuroscience, Program in Cellular Neuroscience, Neurodegeneration, and Repair, Kavli Institute for Neuroscience, Yale University School of Medicine, New Haven, CT 06520, USA; Department of Pharmacology, Department of Neurology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Coralie Berthoux
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Erika Hoyos-Ramirez
- Department of Cellular and Molecular Physiology, Department of Neuroscience, Program in Cellular Neuroscience, Neurodegeneration, and Repair, Kavli Institute for Neuroscience, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Lili Shan
- Department of Cellular and Molecular Physiology, Department of Neuroscience, Program in Cellular Neuroscience, Neurodegeneration, and Repair, Kavli Institute for Neuroscience, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Megumi Morimoto-Tomita
- Department of Cellular and Molecular Physiology, Department of Neuroscience, Program in Cellular Neuroscience, Neurodegeneration, and Repair, Kavli Institute for Neuroscience, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Yixiang Wang
- Department of Cellular and Molecular Physiology, Department of Neuroscience, Program in Cellular Neuroscience, Neurodegeneration, and Repair, Kavli Institute for Neuroscience, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Pablo E Castillo
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Susumu Tomita
- Department of Cellular and Molecular Physiology, Department of Neuroscience, Program in Cellular Neuroscience, Neurodegeneration, and Repair, Kavli Institute for Neuroscience, Yale University School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
17
|
Faissner A. Low-density lipoprotein receptor-related protein-1 (LRP1) in the glial lineage modulates neuronal excitability. FRONTIERS IN NETWORK PHYSIOLOGY 2023; 3:1190240. [PMID: 37383546 PMCID: PMC10293750 DOI: 10.3389/fnetp.2023.1190240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 05/25/2023] [Indexed: 06/30/2023]
Abstract
The low-density lipoprotein related protein receptor 1 (LRP1), also known as CD91 or α-Macroglobulin-receptor, is a transmembrane receptor that interacts with more than 40 known ligands. It plays an important biological role as receptor of morphogens, extracellular matrix molecules, cytokines, proteases, protease inhibitors and pathogens. In the CNS, it has primarily been studied as a receptor and clearance agent of pathogenic factors such as Aβ-peptide and, lately, Tau protein that is relevant for tissue homeostasis and protection against neurodegenerative processes. Recently, it was found that LRP1 expresses the Lewis-X (Lex) carbohydrate motif and is expressed in the neural stem cell compartment. The removal of Lrp1 from the cortical radial glia compartment generates a strong phenotype with severe motor deficits, seizures and a reduced life span. The present review discusses approaches that have been taken to address the neurodevelopmental significance of LRP1 by creating novel, lineage-specific constitutive or conditional knockout mouse lines. Deficits in the stem cell compartment may be at the root of severe CNS pathologies.
Collapse
|
18
|
Hendricks EL, Liebl FL. A closer look at the synapse: dlg1[4K] enables cell-specific visualization of PSD-95/DLG1. CELL REPORTS METHODS 2023; 3:100487. [PMID: 37323573 PMCID: PMC10261923 DOI: 10.1016/j.crmeth.2023.100487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
The PSD-95 homolog, DLG1, is important for excitatory synapse structure and function throughout the Drosophila nervous system. In this issue of Cell Reports Methods, Parisi et al. present a tool, dlg1[4K], that enables cell-specific DLG1 visualization without altering basal synaptic physiology. This tool will potentially enhance our understanding of neuronal development and function in both circuits and individual synapses.
Collapse
Affiliation(s)
- Emily L. Hendricks
- Department of Biological Sciences, Southern Illinois University Edwardsville, Edwardsville, IL 62025, USA
| | - Faith L.W. Liebl
- Department of Biological Sciences, Southern Illinois University Edwardsville, Edwardsville, IL 62025, USA
| |
Collapse
|
19
|
Scotton E, Casa PL, de Abreu FP, de Avila E Silva S, Wilges RLB, Rossetto MV, Géa LP, Rosa AR, Colombo R. Differentially regulated targets in the fast-acting antidepressant effect of (R)-ketamine: A systems biology approach. Pharmacol Biochem Behav 2023; 223:173523. [PMID: 36731751 DOI: 10.1016/j.pbb.2023.173523] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/24/2023] [Accepted: 01/25/2023] [Indexed: 02/03/2023]
Abstract
Approximately two-thirds of patients with major depressive disorder (MDD) fail to respond to conventional antidepressants, suggesting that additional mechanisms are involved in the MDD pathophysiology. In this scenario, the glutamatergic system represents a promising therapeutic target for treatment-resistant depression. To our knowledge, this is the first study using semantic approach with systems biology to identify potential targets involved in the fast-acting antidepressant effects of ketamine and its enantiomers as well as identifying specific targets of (R)-ketamine. We performed a systematic review, followed by a semantic analysis and functional gene enrichment to identify the main biological processes involved in the therapeutic effects of these agents. Protein-protein interaction networks were constructed, and the genes exclusively regulated by (R)-ketamine were explored. We found that the regulation of α-Amino-3-Hydroxy-5-Methyl-4-Isoxazolepropionic Acid (AMPA) receptor and N-methyl-d-aspartate (NMDA) receptor subunits-Postsynaptic Protein 95 (PSD-95), Brain Derived Neurotrophic Factor (BDNF), and Tyrosine Receptor Kinase B (TrkB) are shared by the three-antidepressant agents, reinforcing the central role of the glutamatergic system and neurogenesis on its therapeutic effects. Differential regulation of Transforming Growth Factor Beta 1 (TGF-β1) receptors-Mitogen-Activated Protein Kinases (MAPK's), Receptor Activator of Nuclear Factor-Kappa Beta Ligand (RANKL), and Serotonin Transporter (SERT) seems to be particularly involved in (R)-ketamine antidepressant effects. Our data helps further studies investigating the relationship between these targets and the mechanisms of (R)-ketamine and searching for other therapeutic compounds that share the regulation of these specific biomolecules. Ultimately, this study could contribute to improve the fast management of depressive-like symptoms with less detrimental side effects than ketamine and (S)-ketamine.
Collapse
Affiliation(s)
- Ellen Scotton
- Laboratory of Molecular Psychiatry, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Pharmacology Department and Graduate Program in Biological Sciences: Pharmacology and Therapeutics, Institute of Health Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil.
| | - Pedro Lenz Casa
- Institute of Biotechnology, Universidade de Caxias do Sul (UCS), Caxias do Sul, RS, Brazil.
| | | | | | - Renata Luiza Boff Wilges
- Laboratory of Molecular Psychiatry, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | | | - Luiza Paul Géa
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada
| | - Adriane R Rosa
- Laboratory of Molecular Psychiatry, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Pharmacology Department and Graduate Program in Biological Sciences: Pharmacology and Therapeutics, Institute of Health Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil; Department of Psychiatry and Graduate Program in Psychiatry and Behavioral Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil.
| | - Rafael Colombo
- Institute of Biotechnology, Universidade de Caxias do Sul (UCS), Caxias do Sul, RS, Brazil.
| |
Collapse
|
20
|
Nalberczak-Skóra M, Beroun A, Skonieczna E, Cały A, Ziółkowska M, Pagano R, Taheri P, Kalita K, Salamian A, Radwanska K. Impaired synaptic transmission in dorsal dentate gyrus increases impulsive alcohol seeking. Neuropsychopharmacology 2023; 48:436-447. [PMID: 36182989 PMCID: PMC9852589 DOI: 10.1038/s41386-022-01464-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 09/13/2022] [Accepted: 09/15/2022] [Indexed: 02/02/2023]
Abstract
Both human and animal studies indicate that the dentate gyrus (DG) of the hippocampus is highly exploited by drug and alcohol abuse. Yet, it is poorly understood how DG dysfunction affects addiction-related behaviors. Here, we used an animal model of alcohol use disorder (AUD) in automated IntelliCages and performed local genetic manipulation to investigate how synaptic transmission in the dorsal DG (dDG) affects alcohol-related behaviors. We show that a cue light induces potentiation-like plasticity of dDG synapses in alcohol-naive mice. This process is impaired in mice trained to drink alcohol. Acamprosate (ACA), a drug that reduces alcohol relapse, rescues the impairment of dDG synaptic transmission in alcohol mice. A molecular manipulation that reduces dDG synaptic AMPAR and NMDAR levels increases impulsive alcohol seeking during cue relapse (CR) in alcohol mice but does not affect alcohol reward, motivation or craving. These findings suggest that hindered dDG synaptic transmission specifically underlies impulsive alcohol seeking induced by alcohol cues, a core symptom of AUD.
Collapse
Affiliation(s)
- Maria Nalberczak-Skóra
- grid.419305.a0000 0001 1943 2944Laboratory of Molecular Basis of Behavior, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland ,grid.460447.50000 0001 2161 9572Experimental Psychopathology Lab, Institute of Psychology of Polish Academy of Sciences, Warsaw, Poland
| | - Anna Beroun
- grid.419305.a0000 0001 1943 2944BRAINCITY, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Edyta Skonieczna
- grid.419305.a0000 0001 1943 2944Laboratory of Molecular Basis of Behavior, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Anna Cały
- grid.419305.a0000 0001 1943 2944Laboratory of Molecular Basis of Behavior, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Magdalena Ziółkowska
- grid.419305.a0000 0001 1943 2944Laboratory of Molecular Basis of Behavior, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Roberto Pagano
- grid.419305.a0000 0001 1943 2944Laboratory of Molecular Basis of Behavior, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Pegah Taheri
- grid.419305.a0000 0001 1943 2944Laboratory of Molecular Basis of Behavior, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Katarzyna Kalita
- grid.419305.a0000 0001 1943 2944BRAINCITY, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Ahmad Salamian
- grid.419305.a0000 0001 1943 2944Laboratory of Molecular Basis of Behavior, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Kasia Radwanska
- Laboratory of Molecular Basis of Behavior, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland.
| |
Collapse
|
21
|
Zhang X, An H, Chen Y, Shu N. Neurobiological Mechanisms of Cognitive Decline Correlated with Brain Aging. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1419:127-146. [PMID: 37418211 DOI: 10.1007/978-981-99-1627-6_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 07/08/2023]
Abstract
Cognitive decline has emerged as one of the greatest health threats of old age. Meanwhile, aging is the primary risk factor for Alzheimer's disease (AD) and other prevalent neurodegenerative disorders. Developing therapeutic interventions for such conditions demands a greater understanding of the processes underlying normal and pathological brain aging. Despite playing an important role in the pathogenesis and incidence of disease, brain aging has not been well understood at a molecular level. Recent advances in the biology of aging in model organisms, together with molecular- and systems-level studies of the brain, are beginning to shed light on these mechanisms and their potential roles in cognitive decline. This chapter seeks to integrate the knowledge about the neurological mechanisms of age-related cognitive changes that underlie aging.
Collapse
Affiliation(s)
- Xiaxia Zhang
- State Key Laboratory of Cognitive Neuroscience and Learning, Faculty of Psychology, Beijing Normal University, Beijing, China
- Beijing Aging Brain Rejuvenation Initiative (BABRI) Centre, Beijing Normal University, Beijing, China
| | - Haiting An
- Beijing Aging Brain Rejuvenation Initiative (BABRI) Centre, Beijing Normal University, Beijing, China
- Beijing Neurosurgical Institute, Beijing Tian Tan Hospital, Capital Medical University, Beijing, China
| | - Yuan Chen
- State Key Laboratory of Cognitive Neuroscience and Learning, Faculty of Psychology, Beijing Normal University, Beijing, China
- Beijing Aging Brain Rejuvenation Initiative (BABRI) Centre, Beijing Normal University, Beijing, China
| | - Ni Shu
- State Key Laboratory of Cognitive Neuroscience and Learning, Faculty of Psychology, Beijing Normal University, Beijing, China.
| |
Collapse
|
22
|
Rho-Kinase/ROCK Phosphorylates PSD-93 Downstream of NMDARs to Orchestrate Synaptic Plasticity. Int J Mol Sci 2022; 24:ijms24010404. [PMID: 36613848 PMCID: PMC9820267 DOI: 10.3390/ijms24010404] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/21/2022] [Accepted: 12/23/2022] [Indexed: 12/28/2022] Open
Abstract
The N-methyl-D-aspartate receptor (NMDAR)-mediated structural plasticity of dendritic spines plays an important role in synaptic transmission in the brain during learning and memory formation. The Rho family of small GTPase RhoA and its downstream effector Rho-kinase/ROCK are considered as one of the major regulators of synaptic plasticity and dendritic spine formation, including long-term potentiation (LTP). However, the mechanism by which Rho-kinase regulates synaptic plasticity is not yet fully understood. Here, we found that Rho-kinase directly phosphorylated discs large MAGUK scaffold protein 2 (DLG2/PSD-93), a major postsynaptic scaffold protein that connects postsynaptic proteins with NMDARs; an ionotropic glutamate receptor, which plays a critical role in synaptic plasticity. Stimulation of striatal slices with an NMDAR agonist induced Rho-kinase-mediated phosphorylation of PSD-93 at Thr612. We also identified PSD-93-interacting proteins, including DLG4 (PSD-95), NMDARs, synaptic Ras GTPase-activating protein 1 (SynGAP1), ADAM metallopeptidase domain 22 (ADAM22), and leucine-rich glioma-inactivated 1 (LGI1), by liquid chromatography-tandem mass spectrometry (LC-MS/MS). Among them, Rho-kinase increased the binding of PSD-93 to PSD-95 and NMDARs. Furthermore, we found that chemical-LTP induced by glycine, which activates NMDARs, increased PSD-93 phosphorylation at Thr612, spine size, and PSD-93 colocalization with PSD-95, while these events were blocked by pretreatment with a Rho-kinase inhibitor. These results indicate that Rho-kinase phosphorylates PSD-93 downstream of NMDARs, and suggest that Rho-kinase mediated phosphorylation of PSD-93 increases the association with PSD-95 and NMDARs to regulate structural synaptic plasticity.
Collapse
|
23
|
Stepan J, Heinz DE, Dethloff F, Bajaj T, Zellner A, Hafner K, Wiechmann S, Mackert S, Mecdad Y, Rabenstein M, Ebert T, Martinelli S, Häusl AS, Pöhlmann ML, Hermann A, Ma X, Pavenstädt H, Schmidt MV, Philipsen A, Turck CW, Deussing JM, Kuster B, Wehr MC, Stein V, Kremerskothen J, Wotjak CT, Gassen NC. Hippo-released WWC1 facilitates AMPA receptor regulatory complexes for hippocampal learning. Cell Rep 2022; 41:111766. [PMID: 36476872 DOI: 10.1016/j.celrep.2022.111766] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 08/23/2022] [Accepted: 11/10/2022] [Indexed: 12/12/2022] Open
Abstract
Learning and memory rely on changes in postsynaptic glutamergic α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)-type receptor (AMPAR) number, spatial organization, and function. The Hippo pathway component WW and C2 domain-containing protein 1 (WWC1) regulates AMPAR surface expression and impacts on memory performance. However, synaptic binding partners of WWC1 and its hierarchical position in AMPAR complexes are largely unclear. Using cell-surface proteomics in hippocampal tissue of Wwc1-deficient mice and by generating a hippocampus-specific interactome, we show that WWC1 is a major regulatory platform in AMPAR signaling networks. Under basal conditions, the Hippo pathway members WWC1 and large tumor-suppressor kinase (LATS) are associated, which might prevent WWC1 effects on synaptic proteins. Reduction of WWC1/LATS binding through a point mutation at WWC1 elevates the abundance of WWC1 in AMPAR complexes and improves hippocampal-dependent learning and memory. Thus, uncoupling of WWC1 from the Hippo pathway to AMPAR-regulatory complexes provides an innovative strategy to enhance synaptic transmission.
Collapse
Affiliation(s)
- Jens Stepan
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, 80804 Munich, Germany; Research Group Neurohomeostasis, Department of Psychiatry and Psychotherapy, University Hospital Bonn, 53127 Bonn, Germany; Department of Obstetrics and Gynecology, Paracelsus Medical University, 5020 Salzburg, Austria.
| | - Daniel E Heinz
- Research Group Neurohomeostasis, Department of Psychiatry and Psychotherapy, University Hospital Bonn, 53127 Bonn, Germany; Research Group Neuronal Plasticity, Max Planck Institute of Psychiatry, 80804 Munich, Germany; Max Planck School of Cognition, 04103 Leipzig, Germany
| | - Frederik Dethloff
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, 80804 Munich, Germany; Metabolomics Core Facility, Max Planck Institute for Biology of Ageing, 50931 Cologne, Germany
| | - Thomas Bajaj
- Research Group Neurohomeostasis, Department of Psychiatry and Psychotherapy, University Hospital Bonn, 53127 Bonn, Germany
| | - Andreas Zellner
- Research Group Neurohomeostasis, Department of Psychiatry and Psychotherapy, University Hospital Bonn, 53127 Bonn, Germany; Chair of Proteomics and Bioanalytics, Technical University of Munich, 85354 Freising, Germany
| | - Kathrin Hafner
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Svenja Wiechmann
- Chair of Proteomics and Bioanalytics, Technical University of Munich, 85354 Freising, Germany; German Cancer Consortium (DKTK), 80336 Munich, Germany; German Cancer Center (DKFZ), 69120 Heidelberg, Germany
| | - Sarah Mackert
- Research Group Neurohomeostasis, Department of Psychiatry and Psychotherapy, University Hospital Bonn, 53127 Bonn, Germany
| | - Yara Mecdad
- Research Group Neurohomeostasis, Department of Psychiatry and Psychotherapy, University Hospital Bonn, 53127 Bonn, Germany
| | - Michael Rabenstein
- Institute of Physiology II, University Hospital Bonn, 53115 Bonn, Germany
| | - Tim Ebert
- Research Group Neurohomeostasis, Department of Psychiatry and Psychotherapy, University Hospital Bonn, 53127 Bonn, Germany; Research Group Neuronal Plasticity, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Silvia Martinelli
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Alexander S Häusl
- Department Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, 80804 Munich, Germany; Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Maximilian L Pöhlmann
- Department Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, 80804 Munich, Germany; Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Anke Hermann
- Department of Medicine D, Division of General Internal Medicine, Nephrology, and Rheumatology, University Hospital Münster, 48149 Münster, Germany
| | - Xiao Ma
- Research Group Signal Transduction, Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, 80336 Munich, Germany
| | - Hermann Pavenstädt
- Department of Medicine D, Division of General Internal Medicine, Nephrology, and Rheumatology, University Hospital Münster, 48149 Münster, Germany
| | - Mathias V Schmidt
- Department Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, 80804 Munich, Germany; Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Alexandra Philipsen
- Clinic for Psychiatry and Psychotherapy, University Hospital Bonn, 53127 Bonn, Germany
| | - Chris W Turck
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Jan M Deussing
- Research Group Molecular Neurogenetics, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Bernhard Kuster
- Chair of Proteomics and Bioanalytics, Technical University of Munich, 85354 Freising, Germany; German Cancer Consortium (DKTK), 80336 Munich, Germany; German Cancer Center (DKFZ), 69120 Heidelberg, Germany; Bavarian Center for Biomolecular Mass Spectrometry, Technical University of Munich, 85354 Freising, Germany
| | - Michael C Wehr
- Research Group Signal Transduction, Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, 80336 Munich, Germany
| | - Valentin Stein
- Institute of Physiology II, University Hospital Bonn, 53115 Bonn, Germany
| | - Joachim Kremerskothen
- Department of Medicine D, Division of General Internal Medicine, Nephrology, and Rheumatology, University Hospital Münster, 48149 Münster, Germany
| | - Carsten T Wotjak
- Research Group Neuronal Plasticity, Max Planck Institute of Psychiatry, 80804 Munich, Germany; Central Nervous System Diseases Research, Boehringer-Ingelheim Pharma GmbH & Co KG, 88400 Biberach, Germany.
| | - Nils C Gassen
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, 80804 Munich, Germany; Research Group Neurohomeostasis, Department of Psychiatry and Psychotherapy, University Hospital Bonn, 53127 Bonn, Germany.
| |
Collapse
|
24
|
Pouyo R, Chung K, Delacroix L, Malgrange B. The ubiquitin-proteasome system in normal hearing and deafness. Hear Res 2022; 426:108366. [PMID: 34645583 DOI: 10.1016/j.heares.2021.108366] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 09/03/2021] [Accepted: 09/23/2021] [Indexed: 12/16/2022]
Abstract
Post-translational modifications of proteins are essential for the proper development and function of many tissues and organs, including the inner ear. Ubiquitination is a highly selective post-translational modification that involves the covalent conjugation of ubiquitin to a substrate protein. The most common outcome of protein ubiquitination is degradation by the ubiquitin-proteasome system (UPS), preventing the accumulation of misfolded, damaged, and excess proteins. In addition to proteasomal degradation, ubiquitination regulates other cellular processes, such as transcription, translation, endocytosis, receptor activity, and subcellular localization. All of these processes are essential for cochlear development and maintenance, as several studies link impairment of UPS with altered cochlear development and hearing loss. In this review, we provide insight into the well-oiled machinery of UPS with a focus on its confirmed role in normal hearing and deafness and potential therapeutic strategies to prevent and treat UPS-associated hearing loss.
Collapse
Affiliation(s)
- Ronald Pouyo
- GIGA-Stem Cells, Developmental Neurobiology Unit, University of Liege, Avenue hippocrate 15, B36 1st Floor B, Liege 4000, Belgium
| | - Keshi Chung
- GIGA-Stem Cells, Developmental Neurobiology Unit, University of Liege, Avenue hippocrate 15, B36 1st Floor B, Liege 4000, Belgium
| | - Laurence Delacroix
- GIGA-Stem Cells, Developmental Neurobiology Unit, University of Liege, Avenue hippocrate 15, B36 1st Floor B, Liege 4000, Belgium
| | - Brigitte Malgrange
- GIGA-Stem Cells, Developmental Neurobiology Unit, University of Liege, Avenue hippocrate 15, B36 1st Floor B, Liege 4000, Belgium.
| |
Collapse
|
25
|
Jaric I, Voelkl B, Clerc M, Schmid MW, Novak J, Rosso M, Rufener R, von Kortzfleisch VT, Richter SH, Buettner M, Bleich A, Amrein I, Wolfer DP, Touma C, Sunagawa S, Würbel H. The rearing environment persistently modulates mouse phenotypes from the molecular to the behavioural level. PLoS Biol 2022; 20:e3001837. [PMID: 36269766 PMCID: PMC9629646 DOI: 10.1371/journal.pbio.3001837] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 11/02/2022] [Accepted: 09/20/2022] [Indexed: 11/07/2022] Open
Abstract
The phenotype of an organism results from its genotype and the influence of the environment throughout development. Even when using animals of the same genotype, independent studies may test animals of different phenotypes, resulting in poor replicability due to genotype-by-environment interactions. Thus, genetically defined strains of mice may respond differently to experimental treatments depending on their rearing environment. However, the extent of such phenotypic plasticity and its implications for the replicability of research findings have remained unknown. Here, we examined the extent to which common environmental differences between animal facilities modulate the phenotype of genetically homogeneous (inbred) mice. We conducted a comprehensive multicentre study, whereby inbred C57BL/6J mice from a single breeding cohort were allocated to and reared in 5 different animal facilities throughout early life and adolescence, before being transported to a single test laboratory. We found persistent effects of the rearing facility on the composition and heterogeneity of the gut microbial community. These effects were paralleled by persistent differences in body weight and in the behavioural phenotype of the mice. Furthermore, we show that environmental variation among animal facilities is strong enough to influence epigenetic patterns in neurons at the level of chromatin organisation. We detected changes in chromatin organisation in the regulatory regions of genes involved in nucleosome assembly, neuronal differentiation, synaptic plasticity, and regulation of behaviour. Our findings demonstrate that common environmental differences between animal facilities may produce facility-specific phenotypes, from the molecular to the behavioural level. Furthermore, they highlight an important limitation of inferences from single-laboratory studies and thus argue that study designs should take environmental background into account to increase the robustness and replicability of findings. The phenotype of an organism results not only from its genotype but also the influence of its environment throughout development. This study shows that common environmental differences between animal facilities can induce substantial variation in the phenotype of mice, thereby highlighting an important limitation of inferences from single-laboratory studies in animal research.
Collapse
Affiliation(s)
- Ivana Jaric
- Animal Welfare Division, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- * E-mail: (IJ); (HW)
| | - Bernhard Voelkl
- Animal Welfare Division, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Melanie Clerc
- Department of Biology, Institute of Microbiology and Swiss Institute of Bioinformatics, ETH Zürich, Zürich, Switzerland
| | | | - Janja Novak
- Animal Welfare Division, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Marianna Rosso
- Animal Welfare Division, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Reto Rufener
- Department of Oncology-Pathology, Karolinska Institutet, Solna, Sweden
| | | | - S. Helene Richter
- Department of Behavioural Biology, University of Münster, Münster, Germany
| | - Manuela Buettner
- Institute for Laboratory Animal Science and Central Animal Facility, Hannover Medical School, Hannover, Germany
| | - André Bleich
- Institute for Laboratory Animal Science and Central Animal Facility, Hannover Medical School, Hannover, Germany
| | - Irmgard Amrein
- Institute of Anatomy, Division of Functional Neuroanatomy, University of Zürich, Zürich, Switzerland; Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | - David P. Wolfer
- Institute of Anatomy, Division of Functional Neuroanatomy, University of Zürich, Zürich, Switzerland; Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | - Chadi Touma
- Department of Behavioural Biology, Osnabrück University, Osnabrück, Germany
| | - Shinichi Sunagawa
- Department of Biology, Institute of Microbiology and Swiss Institute of Bioinformatics, ETH Zürich, Zürich, Switzerland
| | - Hanno Würbel
- Animal Welfare Division, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- * E-mail: (IJ); (HW)
| |
Collapse
|
26
|
van Zundert B, Montecino M. Epigenetic Changes and Chromatin Reorganization in Brain Function: Lessons from Fear Memory Ensemble and Alzheimer’s Disease. Int J Mol Sci 2022; 23:ijms232012081. [PMID: 36292933 PMCID: PMC9602769 DOI: 10.3390/ijms232012081] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 10/03/2022] [Accepted: 10/05/2022] [Indexed: 11/16/2022] Open
Abstract
Healthy brain functioning in mammals requires a continuous fine-tuning of gene expression. Accumulating evidence over the last three decades demonstrates that epigenetic mechanisms and dynamic changes in chromatin organization are critical components during the control of gene transcription in neural cells. Recent genome-wide analyses show that the regulation of brain genes requires the contribution of both promoter and long-distance enhancer elements, which must functionally interact with upregulated gene expression in response to physiological cues. Hence, a deep comprehension of the mechanisms mediating these enhancer–promoter interactions (EPIs) is critical if we are to understand the processes associated with learning, memory and recall. Moreover, the onset and progression of several neurodegenerative diseases and neurological alterations are found to be strongly associated with changes in the components that support and/or modulate the dynamics of these EPIs. Here, we overview relevant discoveries in the field supporting the role of the chromatin organization and of specific epigenetic mechanisms during the control of gene transcription in neural cells from healthy mice subjected to the fear conditioning paradigm, a relevant model to study memory ensemble. Additionally, special consideration is dedicated to revising recent results generated by investigators working with animal models and human postmortem brain tissue to address how changes in the epigenome and chromatin architecture contribute to transcriptional dysregulation in Alzheimer’s disease, a widely studied neurodegenerative disease. We also discuss recent developments of potential new therapeutic strategies involving epigenetic editing and small chromatin-modifying molecules (or epidrugs).
Collapse
Affiliation(s)
- Brigitte van Zundert
- Institute of Biomedical Sciences, Faculty of Medicine and Faculty of Life Sciences, Universidad Andres Bello, Santiago 8370186, Chile
- CARE Biomedical Research Center, Santiago 8330005, Chile
- Correspondence: (B.v.Z.); (M.M.)
| | - Martin Montecino
- Institute of Biomedical Sciences, Faculty of Medicine and Faculty of Life Sciences, Universidad Andres Bello, Santiago 8370186, Chile
- Millennium Institute Center for Genome Regulation CRG, Santiago 8370186, Chile
- Correspondence: (B.v.Z.); (M.M.)
| |
Collapse
|
27
|
Yasuda R, Hayashi Y, Hell JW. CaMKII: a central molecular organizer of synaptic plasticity, learning and memory. Nat Rev Neurosci 2022; 23:666-682. [PMID: 36056211 DOI: 10.1038/s41583-022-00624-2] [Citation(s) in RCA: 197] [Impact Index Per Article: 65.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/19/2022] [Indexed: 12/30/2022]
Abstract
Calcium-calmodulin (CaM)-dependent protein kinase II (CaMKII) is the most abundant protein in excitatory synapses and is central to synaptic plasticity, learning and memory. It is activated by intracellular increases in calcium ion levels and triggers molecular processes necessary for synaptic plasticity. CaMKII phosphorylates numerous synaptic proteins, thereby regulating their structure and functions. This leads to molecular events crucial for synaptic plasticity, such as receptor trafficking, localization and activity; actin cytoskeletal dynamics; translation; and even transcription through synapse-nucleus shuttling. Several new tools affording increasingly greater spatiotemporal resolution have revealed the link between CaMKII activity and downstream signalling processes in dendritic spines during synaptic and behavioural plasticity. These technologies have provided insights into the function of CaMKII in learning and memory.
Collapse
Affiliation(s)
- Ryohei Yasuda
- Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA.
| | - Yasunori Hayashi
- Department of Pharmacology, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| | - Johannes W Hell
- Department of Pharmacology, University of California, Davis, Davis, CA, USA.
| |
Collapse
|
28
|
Carvalho C, Correia SC, Seiça R, Moreira PI. WWOX inhibition by Zfra1-31 restores mitochondrial homeostasis and viability of neuronal cells exposed to high glucose. Cell Mol Life Sci 2022; 79:487. [PMID: 35984507 PMCID: PMC11071800 DOI: 10.1007/s00018-022-04508-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 07/26/2022] [Accepted: 07/27/2022] [Indexed: 02/04/2023]
Abstract
Diabetes has been associated with an increased risk of cognitive decline and dementia. However, the mechanisms underlying this association remain unclear and no effective therapeutic interventions exist. Accumulating evidence demonstrates that mitochondrial defects are a key feature of diabetes contributing to neurodegenerative events. It has also been demonstrated that the putative tumor suppressor WW domain-containing oxidoreductase 1 (WWOX) can interact with mitochondria in several pathological conditions. However, its role in diabetes-associated neurodegeneration remains unknown. So, this study aimed to evaluate the role of WWOX activation in high glucose-induced neuronal damage and death. Our experiments were mainly performed in differentiated SH-SY5Y neuroblastoma cells exposed to high glucose and treated (or not) with Zfra1-31, the specific inhibitor of WWOX. Several parameters were analyzed namely cell viability, WWOX activation (tyrosine 33 residue phosphorylation), mitochondrial function, reactive oxygen species (ROS) production, biogenesis, and dynamics, autophagy and oxidative stress/damage. The levels of the neurotoxic proteins amyloid β (Aβ) and phosphorylated Tau (pTau) and of synaptic integrity markers were also evaluated. We observed that high glucose increased the levels of activated WWOX. Interestingly, brain cortical and hippocampal homogenates from young (6-month old) diabetic GK rats showed increased levels of activated WWOX compared to older GK rats (12-month old) suggesting that WWOX plays an early role in the diabetic brain. In neuronal cells, high glucose impaired mitochondrial respiration, dynamics and biogenesis, increased mitochondrial ROS production and decreased mitochondrial membrane potential and ATP production. More, high glucose augmented oxidative stress/damage and the levels of Aβ and pTau proteins and affected autophagy, contributing to the loss of synaptic integrity and cell death. Of note, the activation of WWOX preceded mitochondrial dysfunction and cell death. Importantly, the inhibition of WWOX with Zfra1-31 reversed, totally or partially, the alterations promoted by high glucose. Altogether our observations demonstrate that under high glucose conditions WWOX activation contributes to mitochondrial anomalies and neuronal damage and death, which suggests that WWOX is a potential therapeutic target for early interventions. Our findings also support the efficacy of Zfra1-31 in treating hyperglycemia/diabetes-associated neurodegeneration.
Collapse
Affiliation(s)
- Cristina Carvalho
- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal.
- Center for Innovation in Biomedicine and Biotechnology (CIBB), Coimbra, Portugal.
- Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal.
| | - Sónia C Correia
- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
- Center for Innovation in Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Raquel Seiça
- Institute of Physiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Paula I Moreira
- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal.
- Center for Innovation in Biomedicine and Biotechnology (CIBB), Coimbra, Portugal.
- Institute of Physiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.
- Laboratory of Physiology, Faculty of Medicine, University of Coimbra, 3000-354, Coimbra, Portugal.
| |
Collapse
|
29
|
Griesius S, O'Donnell C, Waldron S, Thomas KL, Dwyer DM, Wilkinson LS, Hall J, Robinson ESJ, Mellor JR. Reduced expression of the psychiatric risk gene DLG2 (PSD93) impairs hippocampal synaptic integration and plasticity. Neuropsychopharmacology 2022; 47:1367-1378. [PMID: 35115661 PMCID: PMC9117295 DOI: 10.1038/s41386-022-01277-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 01/04/2022] [Accepted: 01/12/2022] [Indexed: 11/15/2022]
Abstract
Copy number variants indicating loss of function in the DLG2 gene have been associated with markedly increased risk for schizophrenia, autism spectrum disorder, and intellectual disability. DLG2 encodes the postsynaptic scaffolding protein DLG2 (PSD93) that interacts with NMDA receptors, potassium channels, and cytoskeletal regulators but the net impact of these interactions on synaptic plasticity, likely underpinning cognitive impairments associated with these conditions, remains unclear. Here, hippocampal CA1 neuronal excitability and synaptic function were investigated in a novel clinically relevant heterozygous Dlg2+/- rat model using ex vivo patch-clamp electrophysiology, pharmacology, and computational modelling. Dlg2+/- rats had reduced supra-linear dendritic integration of synaptic inputs resulting in impaired associative long-term potentiation. This impairment was not caused by a change in synaptic input since NMDA receptor-mediated synaptic currents were, conversely, increased and AMPA receptor-mediated currents were unaffected. Instead, the impairment in associative long-term potentiation resulted from an increase in potassium channel function leading to a decrease in input resistance, which reduced supra-linear dendritic integration. Enhancement of dendritic excitability by blockade of potassium channels or activation of muscarinic M1 receptors with selective allosteric agonist 77-LH-28-1 reduced the threshold for dendritic integration and 77-LH-28-1 rescued the associative long-term potentiation impairment in the Dlg2+/- rats. These findings demonstrate a biological phenotype that can be reversed by compound classes used clinically, such as muscarinic M1 receptor agonists, and is therefore a potential target for therapeutic intervention.
Collapse
Affiliation(s)
- Simonas Griesius
- Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, University Walk, Bristol, BS8 1TD, UK
| | - Cian O'Donnell
- Computational Neuroscience Unit, School of Computer Science, Electrical and Electronic Engineering, and Engineering Mathematics, University of Bristol, Bristol, BS8 1UB, UK
| | - Sophie Waldron
- Neuroscience and Mental Health Research Institute, Cardiff, CF24 4HQ, UK
- School of Psychology, Cardiff, CF24 4HQ, UK
| | - Kerrie L Thomas
- Neuroscience and Mental Health Research Institute, Cardiff, CF24 4HQ, UK
- School of Medicine, Cardiff, CF24 4HQ, UK
| | - Dominic M Dwyer
- Neuroscience and Mental Health Research Institute, Cardiff, CF24 4HQ, UK
- School of Psychology, Cardiff, CF24 4HQ, UK
| | - Lawrence S Wilkinson
- Neuroscience and Mental Health Research Institute, Cardiff, CF24 4HQ, UK
- School of Psychology, Cardiff, CF24 4HQ, UK
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff, CF24 4HQ, UK
| | - Jeremy Hall
- Neuroscience and Mental Health Research Institute, Cardiff, CF24 4HQ, UK
- School of Medicine, Cardiff, CF24 4HQ, UK
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff, CF24 4HQ, UK
| | - Emma S J Robinson
- Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, University Walk, Bristol, BS8 1TD, UK
| | - Jack R Mellor
- Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, University Walk, Bristol, BS8 1TD, UK.
| |
Collapse
|
30
|
Choquet D, Opazo P. The role of AMPAR lateral diffusion in memory. Semin Cell Dev Biol 2022; 125:76-83. [PMID: 35123863 DOI: 10.1016/j.semcdb.2022.01.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 01/26/2022] [Accepted: 01/27/2022] [Indexed: 12/17/2022]
Abstract
The accumulation of AMPARs to synapses is a fundamental step in Long-term potentiation (LTP) of synaptic transmission, a well-established cellular correlate of learning and memory. The discovery of a sizeable and highly mobile population of extrasynaptic AMPARs - randomly scanning the synaptic surface under basal conditions - provided a conceptual framework for a simplified model: LTP can be induced by the capture, and hence accumulation, of laterally diffusing extrasynaptic AMPARs. Here, we review the evidence supporting a rate-limiting role of AMPAR lateral diffusion in LTP and as consequence, in learning and memory. We propose that there are "multiple solutions" for achieving the diffusional trapping of AMPAR during LTP, mainly mediated by the interaction between interchangeable AMPAR auxiliary subunits and cell-adhesion molecules containing PDZ-binding domains and synaptic scaffolds containing PDZ-domains. We believe that this molecular degeneracy in the diffusional trapping of AMPAR during LTP serve to ensure the robustness of this crucial step in the making of memories. All in all, the role of AMPAR lateral diffusion in LTP is not only a conceptual leap in our understanding of memory, but it might also hold the keys for the development of therapeutics against disorders associated with memory deficits such as Alzheimer's disease.
Collapse
Affiliation(s)
- Daniel Choquet
- Interdisciplinary Institute for Neuroscience, CNRS, Univ. Bordeaux, IINS, UMR 5297, Bordeaux, France; Univ. Bordeaux, CNRS, INSERM, Bordeaux Imaging Center, BIC, UMS 3420, Bordeaux, France.
| | - Patricio Opazo
- UK Dementia Research Institute, Centre for Discovery Brain Sciences, University of Edinburgh, Chancellor's Building, Edinburgh Medical School, Edinburgh EH16 4SB, UK; Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia.
| |
Collapse
|
31
|
Neurodevelopmental Disorders Associated with PSD-95 and Its Interaction Partners. Int J Mol Sci 2022; 23:ijms23084390. [PMID: 35457207 PMCID: PMC9025546 DOI: 10.3390/ijms23084390] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/13/2022] [Accepted: 04/14/2022] [Indexed: 01/17/2023] Open
Abstract
The postsynaptic density (PSD) is a massive protein complex, critical for synaptic strength and plasticity in excitatory neurons. Here, the scaffolding protein PSD-95 plays a crucial role as it organizes key PSD components essential for synaptic signaling, development, and survival. Recently, variants in DLG4 encoding PSD-95 were found to cause a neurodevelopmental disorder with a variety of clinical features including intellectual disability, developmental delay, and epilepsy. Genetic variants in several of the interaction partners of PSD-95 are associated with similar phenotypes, suggesting that deficient PSD-95 may affect the interaction partners, explaining the overlapping symptoms. Here, we review the transmembrane interaction partners of PSD-95 and their association with neurodevelopmental disorders. We assess how the structural changes induced by DLG4 missense variants may disrupt or alter such protein-protein interactions, and we argue that the pathological effect of DLG4 variants is, at least partly, exerted indirectly through interaction partners of PSD-95. This review presents a direction for functional studies to elucidate the pathogenic mechanism of deficient PSD-95, providing clues for therapeutic strategies.
Collapse
|
32
|
Waldron S, Pass R, Griesius S, Mellor JR, Robinson ESJ, Thomas KL, Wilkinson LS, Humby T, Hall J, Dwyer DM. Behavioural and molecular characterisation of the Dlg2 haploinsufficiency rat model of genetic risk for psychiatric disorder. GENES, BRAIN, AND BEHAVIOR 2022; 21:e12797. [PMID: 35075790 PMCID: PMC9393932 DOI: 10.1111/gbb.12797] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 01/13/2022] [Accepted: 01/13/2022] [Indexed: 11/28/2022]
Abstract
Genetic studies implicate disruption to the DLG2 gene in copy number variants as increasing risk for schizophrenia, autism spectrum disorders and intellectual disability. To investigate psychiatric endophenotypes associated with DLG2 haploinsufficiency (and concomitant PSD-93 protein reduction) a novel clinically relevant Dlg2+/- rat was assessed for abnormalities in anxiety, sensorimotor gating, hedonic reactions, social behaviour, and locomotor response to the N-Methyl-D-aspartic acid receptor antagonist phencyclidine. Dlg gene and protein expression were also investigated to assess model validity. Reductions in PSD-93 messenger RNA and protein were observed in the absence of compensation by other related genes or proteins. Behaviourally Dlg2+/- rats show a potentiated locomotor response to phencyclidine, as is typical of psychotic disorder models, in the absence of deficits in the other behavioural phenotypes assessed here. This shows that the behavioural effects of Dlg2 haploinsufficiency may specifically relate to psychosis vulnerability but are subtle, and partially dissimilar to behavioural deficits previously reported in Dlg2+/- mouse models demonstrating issues surrounding the comparison of models with different aetiology and species. Intact performance on many of the behavioural domains assessed here, such as anxiety and reward processing, will remove these as confounds when continuing investigation into this model using more complex cognitive tasks.
Collapse
Affiliation(s)
| | - Rachel Pass
- Neuroscience and Mental Health Research InstituteCardiff UniversityCardiffUK
- Neurobiology Research UnitOkinawa Institute of Science and TechnologyOnna‐sonOkinawaJapan
| | - Simonas Griesius
- Centre for Synaptic Plasticity, School of Physiology, Pharmacology and NeuroscienceUniversity of Bristol, University WalkBristolUK
| | - Jack R. Mellor
- Centre for Synaptic Plasticity, School of Physiology, Pharmacology and NeuroscienceUniversity of Bristol, University WalkBristolUK
| | - Emma S. J. Robinson
- Centre for Synaptic Plasticity, School of Physiology, Pharmacology and NeuroscienceUniversity of Bristol, University WalkBristolUK
| | - Kerrie L. Thomas
- Neuroscience and Mental Health Research InstituteCardiff UniversityCardiffUK
| | - Lawrence S. Wilkinson
- School of PsychologyCardiff UniversityCardiffUK
- Neuroscience and Mental Health Research InstituteCardiff UniversityCardiffUK
| | | | - Jeremy Hall
- Neuroscience and Mental Health Research InstituteCardiff UniversityCardiffUK
- Medical Research Council Centre for Neuropsychiatric Genetics and GenomicsCardiff UniversityCardiffUK
| | | |
Collapse
|
33
|
Pass R, Haan N, Humby T, Wilkinson LS, Hall J, Thomas KL. Selective behavioural impairments in mice heterozygous for the cross disorder psychiatric risk gene DLG2. GENES, BRAIN, AND BEHAVIOR 2022; 21:e12799. [PMID: 35118804 PMCID: PMC9393930 DOI: 10.1111/gbb.12799] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 01/10/2022] [Accepted: 01/19/2022] [Indexed: 12/17/2022]
Abstract
Mutations affecting DLG2 are emerging as a genetic risk factor associated with neurodevelopmental psychiatric disorders including schizophrenia, autism spectrum disorder, and bipolar disorder. Discs large homolog 2 (DLG2) is a member of the membrane-associated guanylate kinase protein superfamily of scaffold proteins, a component of the post-synaptic density in excitatory neurons and regulator of synaptic function and plasticity. It remains an important question whether and how haploinsuffiency of DLG2 contributes to impairments in basic behavioural and cognitive functions that may underlie symptomatic domains in patients that cross diagnostic boundaries. Using a heterozygous Dlg2 mouse model we examined the impact of reduced Dlg2 expression on functions commonly impaired in neurodevelopmental psychiatric disorders including motor co-ordination and learning, pre-pulse inhibition and habituation to novel stimuli. The heterozygous Dlg2 mice exhibited behavioural impairments in long-term motor learning and long-term habituation to a novel context, but not motor co-ordination, initial responses to a novel context, PPI of acoustic startle or anxiety. We additionally showed evidence for the reduced regulation of the synaptic plasticity-associated protein cFos in the motor cortex during motor learning. The sensitivity of selective behavioural and cognitive functions, particularly those dependent on synaptic plasticity, to reduced expression of DLG2 give further credence for DLG2 playing a critical role in specific brain functions but also a mechanistic understanding of symptom expression shared across psychiatric disorders.
Collapse
Affiliation(s)
- Rachel Pass
- Neuroscience and Mental Health Research InstituteCardiff UniversityCardiffUK
- Okinawa Institute of Science and TechnologyOkinawaJapan
| | - Niels Haan
- Neuroscience and Mental Health Research InstituteCardiff UniversityCardiffUK
| | | | - Lawrence S. Wilkinson
- Neuroscience and Mental Health Research InstituteCardiff UniversityCardiffUK
- School of PsychologyCardiff UniversityCardiffUK
| | - Jeremy Hall
- Neuroscience and Mental Health Research InstituteCardiff UniversityCardiffUK
- Medical Research Council Centre for Neuropsychiatric Genetics and GenomicsCardiff UniversityCardiffUK
| | - Kerrie L. Thomas
- Neuroscience and Mental Health Research InstituteCardiff UniversityCardiffUK
- School of BiosciencesCardiff UniversityCardiffUK
| |
Collapse
|
34
|
Chater TE, Goda Y. The Shaping of AMPA Receptor Surface Distribution by Neuronal Activity. Front Synaptic Neurosci 2022; 14:833782. [PMID: 35387308 PMCID: PMC8979068 DOI: 10.3389/fnsyn.2022.833782] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 02/25/2022] [Indexed: 12/29/2022] Open
Abstract
Neurotransmission is critically dependent on the number, position, and composition of receptor proteins on the postsynaptic neuron. Of these, α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptors (AMPARs) are responsible for the majority of postsynaptic depolarization at excitatory mammalian synapses following glutamate release. AMPARs are continually trafficked to and from the cell surface, and once at the surface, AMPARs laterally diffuse in and out of synaptic domains. Moreover, the subcellular distribution of AMPARs is shaped by patterns of activity, as classically demonstrated by the synaptic insertion or removal of AMPARs following the induction of long-term potentiation (LTP) and long-term depression (LTD), respectively. Crucially, there are many subtleties in the regulation of AMPARs, and exactly how local and global synaptic activity drives the trafficking and retention of synaptic AMPARs of different subtypes continues to attract attention. Here we will review how activity can have differential effects on AMPAR distribution and trafficking along with its subunit composition and phosphorylation state, and we highlight some of the controversies and remaining questions. As the AMPAR field is extensive, to say the least, this review will focus primarily on cellular and molecular studies in the hippocampus. We apologise to authors whose work could not be cited directly owing to space limitations.
Collapse
|
35
|
The synaptic scaffold protein MPP2 interacts with GABAA receptors at the periphery of the postsynaptic density of glutamatergic synapses. PLoS Biol 2022; 20:e3001503. [PMID: 35312684 PMCID: PMC8970474 DOI: 10.1371/journal.pbio.3001503] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 03/31/2022] [Accepted: 12/02/2021] [Indexed: 01/08/2023] Open
Abstract
Recent advances in imaging technology have highlighted that scaffold proteins and receptors are arranged in subsynaptic nanodomains. The synaptic membrane-associated guanylate kinase (MAGUK) scaffold protein membrane protein palmitoylated 2 (MPP2) is a component of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor–associated protein complexes and also binds to the synaptic cell adhesion molecule SynCAM 1. Using superresolution imaging, we show that—like SynCAM 1—MPP2 is situated at the periphery of the postsynaptic density (PSD). In order to explore MPP2-associated protein complexes, we used a quantitative comparative proteomics approach and identified multiple γ-aminobutyric acid (GABA)A receptor subunits among novel synaptic MPP2 interactors. In line with a scaffold function for MPP2 in the assembly and/or modulation of intact GABAA receptors, manipulating MPP2 expression had effects on inhibitory synaptic transmission. We further show that GABAA receptors are found together with MPP2 in a subset of dendritic spines and thus highlight MPP2 as a scaffold that serves as an adaptor molecule, linking peripheral synaptic elements critical for inhibitory regulation to central structures at the PSD of glutamatergic synapses. This study shows that the MAGUK scaffold protein MPP2 is located at the periphery of postsynaptic densities in excitatory neurons, where it interacts with GABA-A receptors, thereby serving as a functional adaptor that links excitatory and inhibitory components of synaptic transmission at glutamatergic synapses.
Collapse
|
36
|
Kay Y, Tsan L, Davis EA, Tian C, Décarie-Spain L, Sadybekov A, Pushkin AN, Katritch V, Kanoski SE, Herring BE. Schizophrenia-associated SAP97 mutations increase glutamatergic synapse strength in the dentate gyrus and impair contextual episodic memory in rats. Nat Commun 2022; 13:798. [PMID: 35145085 PMCID: PMC8831576 DOI: 10.1038/s41467-022-28430-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 01/05/2022] [Indexed: 12/24/2022] Open
Abstract
Mutations in the putative glutamatergic synapse scaffolding protein SAP97 are associated with the development of schizophrenia in humans. However, the role of SAP97 in synaptic regulation is unclear. Here we show that SAP97 is expressed in the dendrites of granule neurons in the dentate gyrus but not in the dendrites of other hippocampal neurons. Schizophrenia-related perturbations of SAP97 did not affect CA1 pyramidal neuron synapse function. Conversely, these perturbations produce dramatic augmentation of glutamatergic neurotransmission in granule neurons that can be attributed to a release of perisynaptic GluA1-containing AMPA receptors into the postsynaptic densities of perforant pathway synapses. Furthermore, inhibiting SAP97 function in the dentate gyrus was sufficient to impair contextual episodic memory. Together, our results identify a cell-type-specific synaptic regulatory mechanism in the dentate gyrus that, when disrupted, impairs contextual information processing in rats. The effects of SAP97 mutations associated with schizophrenia on synaptic function are unclear. Here, the authors show that schizophrenia-related SAP97 mutations enhance glutamatergic synapse strength in the dentate gyrus, impairing contextual episodic memory in rats.
Collapse
Affiliation(s)
- Yuni Kay
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, 90089, USA
| | - Linda Tsan
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, 90089, USA
| | - Elizabeth A Davis
- Department of Biological Sciences, Human and Evolutionary Biology Section, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, CA, 90089, USA
| | - Chen Tian
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, 90089, USA
| | - Léa Décarie-Spain
- Department of Biological Sciences, Human and Evolutionary Biology Section, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, CA, 90089, USA
| | - Anastasiia Sadybekov
- Department of Chemistry, University of Southern California, Los Angeles, CA, 90089, USA
| | - Anna N Pushkin
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, 90089, USA
| | - Vsevolod Katritch
- Department of Chemistry, University of Southern California, Los Angeles, CA, 90089, USA.,Quantitative and Computational Biology, University of Southern California, Los Angeles, CA, 90089, USA
| | - Scott E Kanoski
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, 90089, USA.,Department of Biological Sciences, Human and Evolutionary Biology Section, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, CA, 90089, USA
| | - Bruce E Herring
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, 90089, USA. .,Department of Biological Sciences, Neurobiology Section, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, CA, 90089, USA.
| |
Collapse
|
37
|
PSD-93 up-regulates the synaptic activity of corticotropin-releasing hormone neurons in the paraventricular nucleus in depression. Acta Neuropathol 2021; 142:1045-1064. [PMID: 34536123 DOI: 10.1007/s00401-021-02371-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 09/07/2021] [Accepted: 09/10/2021] [Indexed: 12/28/2022]
Abstract
Since the discovery of ketamine anti-depressant effects in last decade, it has effectively revitalized interest in investigating excitatory synapses hypothesis in the pathogenesis of depression. In the present study, we aimed to reveal the excitatory synaptic regulation of corticotropin-releasing hormone (CRH) neuron in the hypothalamus, which is the driving force in hypothalamic-pituitary-adrenal (HPA) axis regulation. This study constitutes the first observation of an increased density of PSD-93-CRH co-localized neurons in the hypothalamic paraventricular nucleus (PVN) of patients with major depression. PSD-93 overexpression in CRH neurons in the PVN induced depression-like behaviors in mice, accompanied by increased serum corticosterone level. PSD-93 knockdown relieved the depression-like phenotypes in a lipopolysaccharide (LPS)-induced depression model. Electrophysiological data showed that PSD-93 overexpression increased CRH neurons synaptic activity, while PSD-93 knockdown decreased CRH neurons synaptic activity. Furthermore, we found that LPS induced increased the release of glutamate from microglia to CRH neurons resulted in depression-like behaviors using fiber photometry recordings. Together, these results show that PSD-93 is involved in the pathogenesis of depression via increasing the synaptic activity of CRH neurons in the PVN, leading to the hyperactivity of the HPA axis that underlies depression-like behaviors.
Collapse
|
38
|
Djemil S, Ressel CR, Abdel-Ghani M, Schneeweis AK, Pak DTS. Central Cholinergic Synapse Formation in Optimized Primary Septal-Hippocampal Co-cultures. Cell Mol Neurobiol 2021; 41:1787-1799. [PMID: 32860154 PMCID: PMC7914286 DOI: 10.1007/s10571-020-00948-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 08/14/2020] [Indexed: 11/29/2022]
Abstract
Septal innervation of basal forebrain cholinergic neurons to the hippocampus is critical for normal learning and memory and is severely degenerated in Alzheimer's disease. To understand the molecular events underlying physiological cholinergic synaptogenesis and remodeling, as well as pathological loss, we developed an optimized primary septal-hippocampal co-culture system. Hippocampal and septal tissue were harvested from embryonic Sprague-Dawley rat brain and cultured together at varying densities, cell ratios, and in the presence of different growth factors. We identified conditions that produced robust septal-hippocampal synapse formation. We used confocal microscopy with primary antibodies and fluorescent ligands to validate that this system was capable of generating developmentally mature cholinergic synapses. Such synapses were comprised of physiological synaptic partners and mimicked the molecular composition of in vivo counterparts. This co-culture system will facilitate the study of the formation, plasticity, and dysfunction of central mammalian cholinergic synapses.
Collapse
Affiliation(s)
- Sarra Djemil
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, D.C., USA
| | - Claire R Ressel
- Department of Biology, Georgetown University, Washington, D.C., USA
| | - Mai Abdel-Ghani
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, D.C., USA
| | - Amanda K Schneeweis
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, D.C., USA
| | - Daniel T S Pak
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, D.C., USA.
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, D.C., USA.
| |
Collapse
|
39
|
Díaz-Alonso J, Nicoll RA. AMPA receptor trafficking and LTP: Carboxy-termini, amino-termini and TARPs. Neuropharmacology 2021; 197:108710. [PMID: 34271016 PMCID: PMC9122021 DOI: 10.1016/j.neuropharm.2021.108710] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 05/28/2021] [Accepted: 07/08/2021] [Indexed: 12/11/2022]
Abstract
AMPA receptors (AMPARs) are fundamental elements in excitatory synaptic transmission and synaptic plasticity in the CNS. Long term potentiation (LTP), a form of synaptic plasticity which contributes to learning and memory formation, relies on the accumulation of AMPARs at the postsynapse. This phenomenon requires the coordinated recruitment of different elements in the AMPAR complex. Based on recent research reviewed herein, we propose an updated AMPAR trafficking and LTP model which incorporates both extracellular as well as intracellular mechanisms. This article is part of the special Issue on 'Glutamate Receptors - AMPA receptors'.
Collapse
Affiliation(s)
- Javier Díaz-Alonso
- Department of Anatomy and Neurobiology, USA; Center for the Neurobiology of Learning and Memory, University of California at Irvine, USA.
| | - Roger A Nicoll
- Departments of Cellular and Molecular Pharmacology, USA; Physiology, University of California at San Francisco, USA.
| |
Collapse
|
40
|
Fukata Y, Hirano Y, Miyazaki Y, Yokoi N, Fukata M. Trans-synaptic LGI1–ADAM22–MAGUK in AMPA and NMDA receptor regulation. Neuropharmacology 2021; 194:108628. [DOI: 10.1016/j.neuropharm.2021.108628] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 05/16/2021] [Accepted: 05/18/2021] [Indexed: 02/06/2023]
|
41
|
Matthews PM, Pinggera A, Kampjut D, Greger IH. Biology of AMPA receptor interacting proteins - From biogenesis to synaptic plasticity. Neuropharmacology 2021; 197:108709. [PMID: 34271020 DOI: 10.1016/j.neuropharm.2021.108709] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/19/2021] [Accepted: 07/08/2021] [Indexed: 12/19/2022]
Abstract
AMPA-type glutamate receptors mediate the majority of excitatory synaptic transmission in the central nervous system. Their signaling properties and abundance at synapses are both crucial determinants of synapse efficacy and plasticity, and are therefore under sophisticated control. Unique to this ionotropic glutamate receptor (iGluR) is the abundance of interacting proteins that contribute to its complex regulation. These include transient interactions with the receptor cytoplasmic tail as well as the N-terminal domain locating to the synaptic cleft, both of which are involved in AMPAR trafficking and receptor stabilization at the synapse. Moreover, an array of transmembrane proteins operate as auxiliary subunits that in addition to receptor trafficking and stabilization also substantially impact AMPAR gating and pharmacology. Here, we provide an overview of the catalogue of AMPAR interacting proteins, and how they contribute to the complex biology of this central glutamate receptor.
Collapse
Affiliation(s)
- Peter M Matthews
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Alexandra Pinggera
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Domen Kampjut
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Ingo H Greger
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, UK.
| |
Collapse
|
42
|
Abstract
This study presents evidence that the MAGUK family of synaptic scaffolding proteins plays an essential, but redundant, role in long-term potentiation (LTP). The action of PSD-95, but not that of SAP102, requires the binding to the transsynaptic adhesion protein ADAM22, which is required for nanocolumn stabilization. Based on these and previous results, we propose a two-step process in the recruitment of AMPARs during LTP. First, AMPARs, via TARPs, bind to exposed PSD-95 in the PSD. This alone is not adequate to enhance synaptic transmission. Second, the AMPAR/TARP/PSD-95 complex is stabilized in the nanocolumn by binding to ADAM22. A second, ADAM22-independent pathway is proposed for SAP102.
Collapse
|
43
|
Regulation of the NMDA receptor by its cytoplasmic domains: (How) is the tail wagging the dog? Neuropharmacology 2021; 195:108634. [PMID: 34097949 DOI: 10.1016/j.neuropharm.2021.108634] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 05/20/2021] [Accepted: 05/31/2021] [Indexed: 12/18/2022]
Abstract
Excitatory neurotransmission mediated by N-methyl-d-aspartate receptors (NMDARs) is critical for synapse development, function, and plasticity in the brain. NMDARs are tetra-heteromeric cation-channels that mediate synaptic transmission and plasticity. Extensive human studies show the existence of genetic variants in NMDAR subunits genes (GRIN genes) that are associated with neurodevelopmental and neuropsychiatric disorders, including autism spectrum disorders (ASD), epilepsy (EP), intellectual disability (ID), attention deficit hyperactivity disorder (ADHD), and schizophrenia (SCZ). NMDAR subunits have a unique modular architecture with four semiautonomous domains. Here we focus on the carboxyl terminal domain (CTD), also known as the intracellular C-tail, which varies in length among the glutamate receptor subunits and is the most diverse domain in terms of amino acid sequence. The CTD shows no sequence homology to any known proteins but encodes short docking motifs for intracellular binding proteins and covalent modifications. Our review will discuss the many important functions of the CTD in regulating NMDA membrane and synaptic targeting, stabilization, degradation targeting, allosteric modulation and metabotropic signaling of the receptor. This article is part of the special issue on 'Glutamate Receptors - NMDA Receptors'.
Collapse
|
44
|
Feng Z, Wu X, Zhang M. Presynaptic bouton compartmentalization and postsynaptic density-mediated glutamate receptor clustering via phase separation. Neuropharmacology 2021; 193:108622. [PMID: 34051266 DOI: 10.1016/j.neuropharm.2021.108622] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 04/25/2021] [Accepted: 05/17/2021] [Indexed: 01/21/2023]
Abstract
Neuronal synapses encompass three compartments: presynaptic axon terminal, synaptic cleft, and postsynaptic dendrite. Each compartment contains densely packed molecular machineries that are involved in synaptic transmission. In recent years, emerging evidence indicates that the assembly of these membraneless substructures or assemblies that are not enclosed by membranes are driven by liquid-liquid phase separation. We review here recent studies that suggest the phase separation-mediated organization of these synaptic compartments. We discuss how synaptic function may be linked to its organization as biomolecular condensates. We conclude with a discussion of areas of future interest in the field for better understanding of the structural architecture of neuronal synapses and its contribution to synaptic functions.
Collapse
Affiliation(s)
- Zhe Feng
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China; Institute for Advanced Study, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Xiandeng Wu
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Mingjie Zhang
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China; School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China.
| |
Collapse
|
45
|
Transcriptomic expression of AMPA receptor subunits and their auxiliary proteins in the human brain. Neurosci Lett 2021; 755:135938. [PMID: 33915226 DOI: 10.1016/j.neulet.2021.135938] [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: 04/02/2021] [Revised: 04/22/2021] [Accepted: 04/23/2021] [Indexed: 11/21/2022]
Abstract
Receptors to glutamate of the AMPA type (AMPARs) serve as the major gates of excitation in the human brain, where they participate in fundamental processes underlying perception, cognition and movement. Due to their central role in brain function, dysregulation of these receptors has been implicated in neuropathological states associated with a large variety of diseases that manifest with abnormal behaviors. The participation of functional abnormalities of AMPARs in brain disorders is strongly supported by genomic, transcriptomic and proteomic studies. Most of these studies have focused on the expression and function of the subunits that make up the channel and define AMPARs (GRIA1-GRIA4), as well of some accessory proteins. However, it is increasingly evident that native AMPARs are composed of a complex array of accessory proteins that regulate their trafficking, localization, kinetics and pharmacology, and a better understanding of the diversity and regional expression of these accessory proteins is largely needed. In this review we will provide an update on the state of current knowledge of AMPA receptors subunits in the context of their accessory proteins at the transcriptome level. We also summarize the regional expression in the human brain and its correlation with the channel forming subunits. Finally, we discuss some of the current limitations of transcriptomic analysis and propose potential ways to overcome them.
Collapse
|
46
|
Gao WJ, Mack NR. From Hyposociability to Hypersociability-The Effects of PSD-95 Deficiency on the Dysfunctional Development of Social Behavior. Front Behav Neurosci 2021; 15:618397. [PMID: 33584217 PMCID: PMC7876227 DOI: 10.3389/fnbeh.2021.618397] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 01/05/2021] [Indexed: 01/11/2023] Open
Abstract
Abnormal social behavior, including both hypo- and hypersociability, is often observed in neurodevelopmental disorders such as autism spectrum disorders. However, the mechanisms associated with these two distinct social behavior abnormalities remain unknown. Postsynaptic density protein-95 (PSD-95) is a highly abundant scaffolding protein in the excitatory synapses and an essential regulator of synaptic maturation by binding to NMDA and AMPA receptors. The DLG4 gene encodes PSD-95, and it is a risk gene for hypersocial behavior. Interestingly, PSD-95 knockout mice exhibit hyposociability during adolescence but hypersociability in adulthood. The adolescent hyposociability is accompanied with an NMDAR hyperfunction in the medial prefrontal cortex (mPFC), an essential part of the social brain for control of sociability. The maturation of mPFC development is delayed until young adults. However, how PSD-95 deficiency affects the functional maturation of mPFC and its connection with other social brain regions remains uncharacterized. It is especially unknown how PSD-95 knockout drives the switch of social behavior from hypo- to hyper-sociability during adolescent-to-adult development. We propose an NMDAR-dependent developmental switch of hypo- to hyper-sociability. PSD-95 deficiency disrupts NMDAR-mediated synaptic connectivity of mPFC and social brain during development in an age- and pathway-specific manner. By utilizing the PSD-95 deficiency mouse, the mechanisms contributing to both hypo- and hyper-sociability can be studied in the same model. This will allow us to assess both local and long-range connectivity of mPFC and examine how they are involved in the distinct impairments in social behavior and how changes in these connections may mature over time.
Collapse
Affiliation(s)
- Wen-Jun Gao
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, United States
| | | |
Collapse
|
47
|
Cieślik M, Gassowska-Dobrowolska M, Zawadzka A, Frontczak-Baniewicz M, Gewartowska M, Dominiak A, Czapski GA, Adamczyk A. The Synaptic Dysregulation in Adolescent Rats Exposed to Maternal Immune Activation. Front Mol Neurosci 2021; 13:555290. [PMID: 33519375 PMCID: PMC7840660 DOI: 10.3389/fnmol.2020.555290] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 11/27/2020] [Indexed: 12/18/2022] Open
Abstract
Maternal immune activation (MIA) is a risk factor for neurodevelopmental disorders in offspring, but the pathomechanism is largely unknown. The aim of our study was to analyse the molecular mechanisms contributing to synaptic alterations in hippocampi of adolescent rats exposed prenatally to MIA. MIA was evoked in pregnant female rats by i.p. administration of lipopolysaccharide at gestation day 9.5. Hippocampi of offspring (52-53-days-old rats) were analysed using transmission electron microscopy (TEM), qPCR and Western blotting. Moreover, mitochondrial membrane potential, activity of respiratory complexes, and changes in glutathione system were measured. It was found that MIA induced changes in hippocampi morphology, especially in the ultrastructure of synapses, including synaptic mitochondria, which were accompanied by impairment of mitochondrial electron transport chain and decreased mitochondrial membrane potential. These phenomena were in agreement with increased generation of reactive oxygen species, which was evidenced by a decreased reduced/oxidised glutathione ratio and an increased level of dichlorofluorescein (DCF) oxidation. Activation of cyclin-dependent kinase 5, and phosphorylation of glycogen synthase kinase 3β on Ser9 occurred, leading to its inhibition and, accordingly, to hypophosphorylation of microtubule associated protein tau (MAPT). Abnormal phosphorylation and dysfunction of MAPT, the manager of the neuronal cytoskeleton, harmonised with changes in synaptic proteins. In conclusion, this is the first study demonstrating widespread synaptic changes in hippocampi of adolescent offspring prenatally exposed to MIA.
Collapse
Affiliation(s)
- Magdalena Cieślik
- Department of Cellular Signalling, Mossakowski Medical Research Centre Polish Academy of Sciences, Warsaw, Poland
| | | | - Aleksandra Zawadzka
- Department of Cellular Signalling, Mossakowski Medical Research Centre Polish Academy of Sciences, Warsaw, Poland
| | | | - Magdalena Gewartowska
- Electron Microscopy Platform, Mossakowski Medical Research Centre Polish Academy of Sciences, Warsaw, Poland
| | - Agnieszka Dominiak
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Grzegorz A Czapski
- Department of Cellular Signalling, Mossakowski Medical Research Centre Polish Academy of Sciences, Warsaw, Poland
| | - Agata Adamczyk
- Department of Cellular Signalling, Mossakowski Medical Research Centre Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
48
|
AMPA and NMDA Receptor Trafficking at Cocaine-Generated Synapses. J Neurosci 2021; 41:1996-2011. [PMID: 33436529 DOI: 10.1523/jneurosci.1918-20.2021] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 12/24/2020] [Accepted: 01/04/2021] [Indexed: 11/21/2022] Open
Abstract
Cocaine experience generates AMPA receptor (AMPAR)-silent synapses in the nucleus accumbens (NAc), which are thought to be new synaptic contacts enriched in GluN2B-containing NMDA receptors (NMDARs). After drug withdrawal, some of these synapses mature by recruiting AMPARs, strengthening the newly established synaptic transmission. Silent synapse generation and maturation are two consecutive cellular steps through which NAc circuits are profoundly remodeled to promote cue-induced cocaine seeking after drug withdrawal. However, the basic cellular processes that mediate these two critical steps remains underexplored. Using a combination of electrophysiology, viral-mediated gene transfer, and confocal imaging in male rats as well as knock-in (KI) mice of both sexes, our current study characterized the dynamic roles played by AMPARs and NMDARs in generation and maturation of silent synapses on NAc medium spiny neurons after cocaine self-administration and withdrawal. We report that cocaine-induced generation of silent synapses not only required synaptic insertion of GluN2B-containing NMDARs, but also, counterintuitively, involved insertion of AMPARs, which subsequently internalized, resulting in the AMPAR-silent state on withdrawal day 1. Furthermore, GluN2B NMDARs functioned to maintain these cocaine-generated synapses in the AMPAR-silent state during drug withdrawal, until they were replaced by nonGluN2B NMDARs, a switch that allowed AMPAR recruitment and maturation of silent synapses. These results reveal dynamic interactions between AMPARs and NMDARs during the generation and maturation of silent synapses after cocaine experience and provide a mechanistic basis through which new synaptic contacts and possibly new neural network patterns created by these synapses can be manipulated for therapeutic benefit.SIGNIFICANCE STATEMENT Studies over the past decade reveal a critical role of AMPA receptor-silent, NMDA receptor-containing synapses in forming cocaine-related memories that drive cocaine relapse. However, it remains incompletely understood how AMPA and NMDA receptors traffic at these synapses during their generation and maturation. The current study characterizes a two-step AMPA receptor trafficking cascade that contributes to the generation of silent synapses in response to cocaine experience, and a two-step NMDA receptor trafficking cascade that contributes to the maturation of these synapses after cocaine withdrawal. These results depict a highly regulated cellular procedure through which nascent glutamatergic synapses are generated in the adult brain after drug experience and provide significant insight into the roles of glutamate receptors in synapse formation and maturation.
Collapse
|
49
|
Pang Y, Shi M. Repetitive Transcranial Magnetic Stimulation Improves Mild Cognitive Impairment Associated with Alzheimer's Disease in Mice by Modulating the miR-567/NEUROD2/PSD95 Axis. Neuropsychiatr Dis Treat 2021; 17:2151-2161. [PMID: 34239303 PMCID: PMC8259939 DOI: 10.2147/ndt.s311183] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 04/17/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Mild cognitive impairment (MCI) is a typical symptom of early Alzheimer's disease (AD) and is driven by the dysfunction of microRNAs (miRs). Repetitive transcranial magnetic stimulation (rTMS) is a non-invasive technique for handling neuropsychiatric disorders and has universally effects on the functions of miRs. In the current study, the improvement effects of rTMS on MCI associated with AD were explored by focusing on miR-567/NEUROD2/PSD95 axis. METHODS MCI was induced in mice using scopolamine and was treated with rTMS of two frequencies (1 Hz and 10 Hz). The changes in cognitive function, brain structure, neurotrophic factor levels, and activity of miR-567/NEUROD2/PSD95 axis were assessed. The interaction between rTMS and miR-567 was further verified by inducing the level of miR-567 in AD mice. RESULTS The administrations of rTMS improved the cognitive function of AD mice and attenuated brain tissue destruction, which were associated with the restored production of BDNF and NGF. Additionally, rTMS administrations suppressed the expression of miR-567 and up-regulated the expressions of NEUROD2 and PSD95, which contributed to the improved condition in central nerve system. With the induced level of miR-567, the effects of rTMS were counteracted: the learning and memorizing abilities of mice were impaired, the brain neuron viability was suppressed, and the production of neurotrophic factors was suppressed even under the administration of rTMS. The changes in brain function and tissues were associated with the inhibited expressions of NEUROD2 and PSD95. CONCLUSION The findings outlined in the current study demonstrated that rTMS treatment could protect brain against AD-induced MCI without significant side effects, and the function depended on the inhibition of miR-567.
Collapse
Affiliation(s)
- Yongfeng Pang
- Department of Rehabilitation, Tiantai People's Hospital of Zhejiang Province, Zhengjiang, 317200, People's Republic of China
| | - Mingfei Shi
- Department of Rehabilitation, Tiantai People's Hospital of Zhejiang Province, Zhengjiang, 317200, People's Republic of China
| |
Collapse
|
50
|
Gobert D, Schohl A, Kutsarova E, Ruthazer ES. TORC1 selectively regulates synaptic maturation and input convergence in the developing visual system. Dev Neurobiol 2020; 80:332-350. [DOI: 10.1002/dneu.22782] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 08/07/2020] [Accepted: 09/16/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Delphine Gobert
- Montreal Neurological Institute‐Hospital McGill University Montreal QC Canada
| | - Anne Schohl
- Montreal Neurological Institute‐Hospital McGill University Montreal QC Canada
| | - Elena Kutsarova
- Montreal Neurological Institute‐Hospital McGill University Montreal QC Canada
| | - Edward S. Ruthazer
- Montreal Neurological Institute‐Hospital McGill University Montreal QC Canada
| |
Collapse
|