1
|
Fok AHK, Lam CHM, Lai CSW. Specific dendritic spine modifications and dendritic transport: From in vitro to in vivo. Neural Regen Res 2026; 21:665-666. [PMID: 39819974 DOI: 10.4103/nrr.nrr-d-24-01159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 12/04/2024] [Indexed: 01/19/2025] Open
Affiliation(s)
- Albert H K Fok
- Center for Research in Neuroscience, Department of Neurology & Neurosurgery, Brain Repair and Integrative Neuroscience Program, The Research Institute of the McGill University Health Centre, Montreal, QC, Canada (Fok AHK)
| | - Charlotte H M Lam
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China (Lam CHM, Lai CSW)
| | - Cora S W Lai
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China (Lam CHM, Lai CSW)
- Advanced Biomedical Instrumentation Center, Hong Kong Science Park, Shatin, New Territories, Hong Kong Special Administrative Region, China (Lai CSW)
- The State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong Special Administrative Region, China (Lai CSW)
| |
Collapse
|
2
|
Li J, Wu D, Mu Y, Wang Y, Zhai L, Jia L. The combined damage of bisphenol A and high fat diet to learning and memory in young male mice: the regulatory effect of BDNF/TrkB/PI3K/AKT pathway on autophagy. ENVIRONMENTAL RESEARCH 2025; 276:121538. [PMID: 40187395 DOI: 10.1016/j.envres.2025.121538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 03/12/2025] [Accepted: 04/02/2025] [Indexed: 04/07/2025]
Abstract
With the popularity of takeaway and processed food, combined exposure to Bisphenol A (BPA) and high fat diet (HFD) is becoming increasingly common. BPA or HFD intake in children could impair learning and memory ability, but the combined effect and mechanisms remain unclear. In this study, we fed young male mice with 0.1 μg/mL BPA (L-BPA), 0.2 μg/mL BPA (H-BPA), 60 %HFD (HFD), 0.1 BPA + HFD (L-BPA + HFD) and 0.2 BPA + HFD (H-BPA + HFD) for 8 weeks. The results showed that recognition memory and free exploration of mice were impaired in the BPA or HFD group, and the damage of exploration was more severe in the combined group. All treated groups showed morphological changes in hippocampal neurons. The levels of synaptic structural protein PSD-95 and SYN were reduced in BPA and HFD alone or in combination groups. BPA or HFD led to changes in autophagy levels in the hippocampus, manifested by decreased protein levels of mTOR and P62, increased level of LC3B, and more significant changes in the combined group. The BDNF/TrkB/PI3K/AKT pathway was inhibited in BPA or HFD groups, especially in the combined group. Our results suggested that combined BPA with HFD exposure could impair learning and memory ability, and the combined effect might be related to the BDNF/TrkB/PI3K/AKT pathway, which regulated mTOR mediated autophagy and finally caused hippocampal synaptic damage.
Collapse
Affiliation(s)
- Jinshi Li
- Department of Child and Adolescent Health, School of Public Health, China Medical University, Shenyang, 110122, Liaoning, China.
| | - Dan Wu
- Department of Child and Adolescent Health, School of Public Health, Chongqing Medical University, Chongqing, 400016, China.
| | - Yuyang Mu
- Department of Child and Adolescent Health, School of Public Health, China Medical University, Shenyang, 110122, Liaoning, China.
| | - Yunzhu Wang
- Department of Child and Adolescent Health, School of Public Health, China Medical University, Shenyang, 110122, Liaoning, China.
| | - Lingling Zhai
- Department of Child and Adolescent Health, School of Public Health, China Medical University, Shenyang, 110122, Liaoning, China; Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, Shenyang, 110122, Liaoning, China.
| | - Lihong Jia
- Department of Child and Adolescent Health, School of Public Health, China Medical University, Shenyang, 110122, Liaoning, China; Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, Shenyang, 110122, Liaoning, China.
| |
Collapse
|
3
|
Sabatier P, Lechner M, Guzmán UH, Beusch CM, Zeng X, Wang L, Izaguirre F, Seth A, Gritsenko O, Rodin S, Grinnemo KH, Ye Z, Olsen JV. Global analysis of protein turnover dynamics in single cells. Cell 2025; 188:2433-2450.e21. [PMID: 40168994 DOI: 10.1016/j.cell.2025.03.002] [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: 07/04/2024] [Revised: 12/20/2024] [Accepted: 03/03/2025] [Indexed: 04/03/2025]
Abstract
Single-cell proteomics (SCPs) has advanced significantly, yet it remains largely unidimensional, focusing primarily on protein abundances. In this study, we employed a pulsed stable isotope labeling by amino acids in cell culture (pSILAC) approach to simultaneously analyze protein abundance and turnover in single cells (SC-pSILAC). Using a state-of-the-art SCP workflow, we demonstrated that two SILAC labels are detectable from ∼4,000 proteins in single HeLa cells recapitulating known biology. We performed a large-scale time-series SC-pSILAC analysis of undirected differentiation of human induced pluripotent stem cells (iPSCs) encompassing 6 sampling times over 2 months and analyzed >1,000 cells. Protein turnover dynamics highlighted differentiation-specific co-regulation of protein complexes with core histone turnover, discriminating dividing and non-dividing cells. Lastly, correlating cell diameter with the abundance of individual proteins showed that histones and some cell-cycle proteins do not scale with cell size. The SC-pSILAC method provides a multidimensional view of protein dynamics in single-cell biology.
Collapse
Affiliation(s)
- Pierre Sabatier
- Novo Nordisk Foundation Center for Protein Research, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark; Cardio-Thoracic Translational Medicine (CTTM) Lab, Department of Surgical Sciences, Uppsala University, 752 37 Uppsala, Sweden.
| | - Maico Lechner
- Novo Nordisk Foundation Center for Protein Research, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Ulises H Guzmán
- Novo Nordisk Foundation Center for Protein Research, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Christian M Beusch
- Cardio-Thoracic Translational Medicine (CTTM) Lab, Department of Surgical Sciences, Uppsala University, 752 37 Uppsala, Sweden; Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA 30322, USA
| | - Xinlei Zeng
- State Key Laboratory of Common Mechanism Research for Major Diseases, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, China
| | - Longteng Wang
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | | | - Anjali Seth
- Cellenion SASU, 60F Avenue Rockefeller, 69008 Lyon, France
| | - Olga Gritsenko
- Cardio-Thoracic Translational Medicine (CTTM) Lab, Department of Surgical Sciences, Uppsala University, 752 37 Uppsala, Sweden
| | - Sergey Rodin
- Cardio-Thoracic Translational Medicine (CTTM) Lab, Department of Surgical Sciences, Uppsala University, 752 37 Uppsala, Sweden
| | - Karl-Henrik Grinnemo
- Cardio-Thoracic Translational Medicine (CTTM) Lab, Department of Surgical Sciences, Uppsala University, 752 37 Uppsala, Sweden
| | - Zilu Ye
- Novo Nordisk Foundation Center for Protein Research, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark; State Key Laboratory of Common Mechanism Research for Major Diseases, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, China.
| | - Jesper V Olsen
- Novo Nordisk Foundation Center for Protein Research, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark.
| |
Collapse
|
4
|
Mohar B, Michel G, Wang YZ, Hernandez V, Grimm JB, Park JY, Patel R, Clarke M, Brown TA, Bergmann C, Gebis KK, Wilen AP, Liu B, Johnson R, Graves A, Tchumatchenko T, Savas JN, Fornasiero EF, Huganir RL, Tillberg PW, Lavis LD, Svoboda K, Spruston N. DELTA: a method for brain-wide measurement of synaptic protein turnover reveals localized plasticity during learning. Nat Neurosci 2025; 28:1089-1098. [PMID: 40164741 DOI: 10.1038/s41593-025-01923-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 02/10/2025] [Indexed: 04/02/2025]
Abstract
Synaptic plasticity alters neuronal connections in response to experience, which is thought to underlie learning and memory. However, the loci of learning-related synaptic plasticity, and the degree to which plasticity is localized or distributed, remain largely unknown. Here we describe a new method, DELTA, for mapping brain-wide changes in synaptic protein turnover with single-synapse resolution, based on Janelia Fluor dyes and HaloTag knock-in mice. During associative learning, the turnover of the ionotropic glutamate receptor subunit GluA2, an indicator of synaptic plasticity, was enhanced in several brain regions, most markedly hippocampal area CA1. More broadly distributed increases in the turnover of synaptic proteins were observed in response to environmental enrichment. In CA1, GluA2 stability was regulated in an input-specific manner, with more turnover in layers containing input from CA3 compared to entorhinal cortex. DELTA will facilitate exploration of the molecular and circuit basis of learning and memory and other forms of plasticity at scales ranging from single synapses to the entire brain.
Collapse
Affiliation(s)
- Boaz Mohar
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA.
| | - Gabriela Michel
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Yi-Zhi Wang
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Veronica Hernandez
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Jonathan B Grimm
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Jin-Yong Park
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Ronak Patel
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Morgan Clarke
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Timothy A Brown
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Cornelius Bergmann
- Institute for Experimental Epileptology and Cognition Research, Medical Faculty, University of Bonn, Bonn, Germany
| | - Kamil K Gebis
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Anika P Wilen
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Bian Liu
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Richard Johnson
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Austin Graves
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Tatjana Tchumatchenko
- Institute for Experimental Epileptology and Cognition Research, Medical Faculty, University of Bonn, Bonn, Germany
| | - Jeffrey N Savas
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Eugenio F Fornasiero
- Department Institute of Neuro- and Sensory Physiology, University Medical Center Göttingen (UMG), Göttingen, Germany
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Richard L Huganir
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Paul W Tillberg
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Luke D Lavis
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Karel Svoboda
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
- Allen Institute for Neural Dynamics, Seattle, WA, USA
| | - Nelson Spruston
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| |
Collapse
|
5
|
Kim D, Park P, Li X, Wong-Campos JD, Tian H, Moult EM, Grimm JB, Lavis LD, Cohen AE. EPSILON: a method for pulse-chase labeling to probe synaptic AMPAR exocytosis during memory formation. Nat Neurosci 2025; 28:1099-1107. [PMID: 40164742 DOI: 10.1038/s41593-025-01922-5] [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: 09/13/2023] [Accepted: 02/10/2025] [Indexed: 04/02/2025]
Abstract
A tool to map changes in synaptic strength during a defined time window could provide powerful insights into the mechanisms of learning and memory. Here we developed a technique, Extracellular Protein Surface Labeling in Neurons (EPSILON), to map α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) exocytosis in vivo by sequential pulse-chase labeling of surface AMPARs with membrane-impermeable dyes. This approach yields synaptic-resolution maps of AMPAR exocytosis, a proxy for synaptic potentiation, in genetically targeted neurons during memory formation. In mice undergoing contextual fear conditioning, we investigated the relationship between synapse-level AMPAR exocytosis in CA1 pyramidal neurons and cell-level expression of the immediate early gene product cFos, a frequently used marker of engram neurons. We observed a strong correlation between AMPAR exocytosis and cFos expression, suggesting a synaptic mechanism for the association of cFos expression with memory engrams. The EPSILON technique is a useful tool for mapping synaptic plasticity and may be extended to investigate trafficking of other transmembrane proteins.
Collapse
Affiliation(s)
- Doyeon Kim
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Pojeong Park
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Department of Brain Sciences, DGIST, Daegu, Republic of Korea
| | - Xiuyuan Li
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Department of Applied Biology and Chemical Technology, Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - J David Wong-Campos
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - He Tian
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Eric M Moult
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Jonathan B Grimm
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Luke D Lavis
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Adam E Cohen
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA.
- Department of Physics, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
6
|
Wagner M, Berecki G, Fazeli W, Nussbaum C, Flemmer AW, Frizzo S, Heer F, Heinen F, Horton R, Jacotin H, Motel W, Spar B, Klein C, Siegel C, Hübener C, Stöcklein S, Paolini M, Staudt M, Tacke M, Wolff M, Petrou S, Souza M, Borggraefe I. Antisense oligonucleotide treatment in a preterm infant with early-onset SCN2A developmental and epileptic encephalopathy. Nat Med 2025:10.1038/s41591-025-03656-0. [PMID: 40263630 DOI: 10.1038/s41591-025-03656-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 03/14/2025] [Indexed: 04/24/2025]
Abstract
Early-onset SCN2A developmental and epileptic encephalopathy is caused by SCN2A gain-of-function variants. Here we describe the clinical experience with intrathecally administered elsunersen, a gapmer antisense oligonucleotide targeting SCN2A, in a female preterm infant with early-onset SCN2A developmental and epileptic encephalopathy, in an expanded access program. Before elsunersen treatement, the patient was in status epilepticus for 7 weeks with a seizure frequency of 20-25 per hour. Voltage-clamp experiments confirmed impaired channel inactivation and increased persistent current consistent with a gain-of-function mechanism. Elsunersen treatment demonstrated a favorable safety profile with no severe or serious adverse events reported after 19 intrathecal administrations over 20 months. After administration in combination with sodium channel blockers, status epilepticus was interrupted intermittently and ultimately ceased after continued dosing. A >60% reduction in seizure frequency corresponding to five to seven seizures per hour was observed, which has been sustained during follow-up until the age of 22 months. These data provide preliminary insights on the safety and efficacy of elsunersen in a preterm infant. Additional investigation on the benefits of elsunersen in clinical trials is warranted.
Collapse
Affiliation(s)
- Matias Wagner
- Division of Pediatric Neurology and Developmental Medicine, Department of Pediatrics and Comprehensive Epilepsy Center, Munich University Center for Children with Medical and Developmental Complexity - MUC iSPZ Hauner, Dr. v. Hauner Children's Hospital, Ludwig Maximilians University Hospital, Munich, Germany
- Institute of Human Genetics, School of Medicine and Health, Technical University of Munich, Munich, Germany
- Institute for Neurogenomics, Helmholtz Centre Munich, German Research Center for Health and Environment, Munich, Germany
| | - Géza Berecki
- Ion Channels and Human Diseases Group, The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Walid Fazeli
- Department of Pediatric Neurology, Children's Hospital, University Hospital Bonn, Bonn, Germany
| | - Claudia Nussbaum
- Division of Neonatology, Dr. v. Hauner Children's Hospital, Ludwig Maximilians University Hospital, Munich, Germany
| | - Andreas W Flemmer
- Division of Neonatology, Dr. v. Hauner Children's Hospital, Ludwig Maximilians University Hospital, Munich, Germany
| | | | - Farina Heer
- Division of Pediatric Neurology and Developmental Medicine, Department of Pediatrics and Comprehensive Epilepsy Center, Munich University Center for Children with Medical and Developmental Complexity - MUC iSPZ Hauner, Dr. v. Hauner Children's Hospital, Ludwig Maximilians University Hospital, Munich, Germany
| | - Florian Heinen
- Division of Pediatric Neurology and Developmental Medicine, Department of Pediatrics and Comprehensive Epilepsy Center, Munich University Center for Children with Medical and Developmental Complexity - MUC iSPZ Hauner, Dr. v. Hauner Children's Hospital, Ludwig Maximilians University Hospital, Munich, Germany
| | | | | | | | - Brian Spar
- Praxis Precision Medicines, Boston, MA, USA
| | - Christoph Klein
- Department of Pediatrics, Dr. v. Hauner Children's Hospital, Ludwig Maximilians University Hospital, Munich, Germany
| | | | - Christoph Hübener
- Department of Obstetrics and Gynecology, Ludwig Maximilians University Hospital, Munich, Germany
| | - Sophia Stöcklein
- Department of Radiology, Ludwig Maximilians University Hospital, Munich, Germany
| | - Marco Paolini
- Department of Radiology, Ludwig Maximilians University Hospital, Munich, Germany
| | - Martin Staudt
- Department of Pediatric Palliative Care, Ludwig Maximilians University Hospital, Munich, Germany
| | - Moritz Tacke
- Division of Pediatric Neurology and Developmental Medicine, Department of Pediatrics and Comprehensive Epilepsy Center, Munich University Center for Children with Medical and Developmental Complexity - MUC iSPZ Hauner, Dr. v. Hauner Children's Hospital, Ludwig Maximilians University Hospital, Munich, Germany
| | - Markus Wolff
- Swiss Epilepsy Center, Klinik Lengg AG, Zurich, Switzerland
| | - Steven Petrou
- Ion Channels and Human Diseases Group, The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
- Praxis Precision Medicines, Boston, MA, USA
| | | | - Ingo Borggraefe
- Division of Pediatric Neurology and Developmental Medicine, Department of Pediatrics and Comprehensive Epilepsy Center, Munich University Center for Children with Medical and Developmental Complexity - MUC iSPZ Hauner, Dr. v. Hauner Children's Hospital, Ludwig Maximilians University Hospital, Munich, Germany.
| |
Collapse
|
7
|
Akter Y, Jones G, Daskivich GJ, Shifflett V, Vargas KJ, Hruska M. Combining nanobody labeling with STED microscopy reveals input-specific and layer-specific organization of neocortical synapses. PLoS Biol 2025; 23:e3002649. [PMID: 40184426 PMCID: PMC12002638 DOI: 10.1371/journal.pbio.3002649] [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: 04/21/2024] [Revised: 04/16/2025] [Accepted: 02/25/2025] [Indexed: 04/06/2025] Open
Abstract
The discovery of synaptic nanostructures revealed key insights into the molecular logic of synaptic function and plasticity. Yet, our understanding of how diverse synapses in the brain organize their nano-architecture remains elusive, largely due to the limitations of super-resolution imaging in complex brain tissue. Here, we characterized single-domain camelid nanobodies for the 3D quantitative multiplex imaging of synaptic nano-organization using tau-STED nanoscopy in cryosections from the mouse primary somatosensory cortex. We focused on thalamocortical (TC) and corticocortical (CC) synapses along the apical-basal axis of layer five pyramidal neurons as models of functionally diverse glutamatergic synapses in the brain. Spines receiving TC input were larger than those receiving CC input in all layers examined. However, the nano-architecture of TC synapses varied with dendritic location. TC afferents on apical dendrites frequently contacted spines with multiple aligned PSD-95/Bassoon nanomodules of constant size. In contrast, TC spines on basal dendrites predominantly contained a single aligned nanomodule, with PSD-95 nanocluster sizes scaling proportionally with spine volume. The nano-organization of CC synapses did not change across cortical layers and resembled modular architecture defined in vitro. These findings highlight the nanoscale diversity of synaptic architecture in the brain, that is, shaped by both the source of afferent input and the subcellular localization of individual synaptic contacts.
Collapse
Affiliation(s)
- Yeasmin Akter
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, West Virginia, United States of America
| | - Grace Jones
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, West Virginia, United States of America
| | - Grant J. Daskivich
- Department of Cell Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Victoria Shifflett
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, West Virginia, United States of America
| | - Karina J. Vargas
- Department of Cell Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Martin Hruska
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, West Virginia, United States of America
| |
Collapse
|
8
|
Xiong L, He R, Du R, Niu R, Xue L, Chen L, Huangfu L, Xiao Q, Li J, Li Y, Zhang S, Yu C, Tian X, Wang T. A Single-Nucleus Transcriptomic Atlas Reveals Cellular and Genetic Characteristics of Alzheimer's-Like Pathology in Aging Tree Shrews. MedComm (Beijing) 2025; 6:e70114. [PMID: 40093585 PMCID: PMC11910145 DOI: 10.1002/mco2.70114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 01/10/2025] [Accepted: 01/25/2025] [Indexed: 03/19/2025] Open
Abstract
The lack of natural aging-inducing Alzheimer's disease (AD) model presents a significant gap in the current preclinical research. Here, we identified a unique cohort of 10 naturally aging tree shrews (TSs) displaying distinct Alzheimer's-like pathology (ALP) from a population of 324, thereby establishing a novel model that closely mirrors human AD progression. Using single-nucleus RNA sequencing, we generated a comprehensive transcriptome atlas, revealing the cellular diversity and gene expression changes underlying AD pathology in aged TSs. Particularly, distinct differentiation trajectories of neural progenitor cells were highly associated with AD pathology. Intriguingly, cross-species comparisons among humans, TSs, monkeys, and mice highlighted a greater cellular homogeneity of TSs to primates and humans than to mice. Our extended cross-species analysis by including a direct comparison between human and TS hippocampal tissue under AD conditions uncovered conserved cell types, enriched synaptic biological processes, and elevated excitatory/inhibitory imbalance across species. Cell-cell communication analysis unveiled parallel patterns between AD human and ALP TSs, with both showing reduced interaction strength and quantity across most cell types. Overall, our study provides rich, high-resolution resources on the cellular and molecular landscape of the ALP TS hippocampus, reinforcing the utility of TSs as a robust model for AD research.
Collapse
Affiliation(s)
- Liu‐Lin Xiong
- Department of AnesthesiologyThe First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University)ZunyiGuizhouChina
| | - Rong He
- Institute of NeuroscienceKunming Medical UniversityKunmingYunnanChina
| | - Ruo‐Lan Du
- Department of AnesthesiologyThe First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University)ZunyiGuizhouChina
| | - Rui‐Ze Niu
- Mental Health Center of Kunming Medical UniversityKunmingYunnanChina
| | - Lu‐Lu Xue
- Institute of Neurological Disease, National‐Local Joint Engineering Research Center of Translational Medicine, State Key Lab of BiotherapyWest China HospitalSichuan UniversityChengduSichuanChina
| | - Li Chen
- Institute of Neurological Disease, National‐Local Joint Engineering Research Center of Translational Medicine, State Key Lab of BiotherapyWest China HospitalSichuan UniversityChengduSichuanChina
| | - Li‐Ren Huangfu
- Institute of NeuroscienceKunming Medical UniversityKunmingYunnanChina
| | - Qiu‐Xia Xiao
- Department of AnesthesiologyThe First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University)ZunyiGuizhouChina
| | - Jing Li
- Institute of NeuroscienceKunming Medical UniversityKunmingYunnanChina
| | - Yong‐Ping Li
- Institute of NeuroscienceKunming Medical UniversityKunmingYunnanChina
| | - Si‐Min Zhang
- Yunnan Key Laboratory of Primate Biomedical Research, Institute of Primate Translational MedicineKunming University of Science and TechnologyKunmingChina
| | - Chang‐Yin Yu
- Department of NeurologyAffiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
| | - Xiao‐He Tian
- Department of AnesthesiologyThe First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University)ZunyiGuizhouChina
| | - Ting‐Hua Wang
- Department of AnesthesiologyThe First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University)ZunyiGuizhouChina
| |
Collapse
|
9
|
Cai Y, Zeng X, Wu M, Chen H, Sun M, Yang H. TET1 mitigates prenatal fluoride-induced cognition impairment by modulating Bcl2 DNA hydroxymethylation level. Mol Med 2025; 31:117. [PMID: 40133886 PMCID: PMC11938627 DOI: 10.1186/s10020-025-01174-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 03/18/2025] [Indexed: 03/27/2025] Open
Abstract
Fluoride exposure during pregnancy commonly compromises fetal neurodevelopment and largely results in a broad spectrum of cognitive deficiencies in the adult offspring. However, the precise mechanisms underlying these effects remain to be fully elucidated. Herein, we investigate the impacts of fluoride on neural excitability and apoptosis, synaptic plasticity, and cognitive function, as well as possible underlying mechanisms. Our results indicated that exposure to a high sodium fluoride (100 mg/L) during pregnancy in the mouse can cause the cognitive deficits of their offspring, accompanied by a decrease in the expression of Tet-eleven translocation protein 1 (TET1), an enzyme responsible for DNA hydroxymethylation. Additionally, there is a reduction in the dendritic spine density and the expression of postsynaptic density protein-95 (PSD95) in the hippocampal regions of male offspring. Furthermore, in vitro fluoride treatment significantly exacerbates neuronal apoptosis and reduces the frequency of spikes in spontaneous action potential. More significantly, we also found that TET1 could directly bind to the promotor region of Bcl2, altering its DNA hydroxymethylation and Bcl2 expression. Intriguingly, Tet1 knock-out mice exhibited cognitive deficits similar to those observed in male animals exposed to high levels of fluoride. Furthermore, the down-regulation of TET1 protein, along with the consequent alteration in Bcl2 hydroxymethylation and increased neuronal apoptosis, are likely mechanisms underlying the impact of prenatal fluoride exposure on the neurodevelopment of male offspring. These findings provide novel insights into the molecular mechanisms by which fluoride exposure induces neurodevelopmental impairment of the male offspring.
Collapse
Affiliation(s)
- Yongle Cai
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Xingdong Zeng
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Mengyan Wu
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Haonan Chen
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Miao Sun
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China.
| | - Hao Yang
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China.
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia, 750004, China.
| |
Collapse
|
10
|
Chen L, Cui W, Qin J, Zhu M, Zhang H, Yang J, Xu Z, Huang H. FKBP51 is Involved in Epileptic Seizure by Regulating PSD95 in a PTZ-Induced Epileptic Mouse Model. J Integr Neurosci 2025; 24:25710. [PMID: 40152573 DOI: 10.31083/jin25710] [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: 07/15/2024] [Revised: 12/10/2024] [Accepted: 12/20/2024] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Epilepsy, the world's third most prevalent chronic brain disorder, significantly affects patients' quality of life and increases the economic burden on families and society. Previous studies have demonstrated that FK506-binding protein 51 (FKBP51) plays a crucial role in synaptic plasticity. However, FKBP51 exhibits different functions under various physiological and pathological conditions. Our study explored the relationship between FKBP51 and epilepsy and its possible mechanism of action. We also analyzed the expression levels of postsynaptic density-95 (PSD95) and synaptophysin (SYP) in the hippocampus to examine the pathophysiology of epilepsy. METHODS A chronic epileptic kindling model was established by injecting pentylenetetrazole (PTZ) intraperitoneally, and a spontaneous seizure model was created by injecting kainic acid (KA) into the dentate gyrus using a stereotaxic apparatus. Endogenous FKBP51 expression was inhibited using adeno-associated virus (AAV)-FKBP51-Small hairpin RNAs (shRNA). The expression of FKBP51, PSD95, and SYP in the hippocampus and synaptosomes was measured through western blotting. Golgi staining and electron microscopy were used to examine spines and synaptic structures. RESULTS The results showed a significant increase in FKBP51 expression in the hippocampal tissue of the PTZ- and KA-induced epilepsy model groups. Inhibition of FKBP51 expression through AAV-FKBP51-shRNA resulted in a shorter latency and an elevated seizure grade score in mice. Moreover, the suppression of FKBP51 expression enhanced the expression of synaptic plasticity-related proteins, increased the density of dendritic spines, and elevated the quantity of spherical synaptic vesicles in the presynaptic membrane in the hippocampus. CONCLUSIONS FKBP51 may serve as an endogenous protective factor in epilepsy by regulating the expression of the synaptic plasticity-related protein PSD95, the density of dendritic spines, and the number of synaptic vesicles in the hippocampal CA1.
Collapse
Affiliation(s)
- Ling Chen
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, 563000 Zunyi, Guizhou, China
| | - Wenxiu Cui
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, 563000 Zunyi, Guizhou, China
- Department of Neurology, Ziyang Central Hospital, 641300 Ziyang, Sichuan, China
| | - Jiyao Qin
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, 563000 Zunyi, Guizhou, China
| | - Manmin Zhu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, 563000 Zunyi, Guizhou, China
| | - Haiqing Zhang
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, 563000 Zunyi, Guizhou, China
| | - Juan Yang
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, 563000 Zunyi, Guizhou, China
| | - Zucai Xu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, 563000 Zunyi, Guizhou, China
| | - Hao Huang
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, 563000 Zunyi, Guizhou, China
| |
Collapse
|
11
|
Borchers C, Osburn K, Roh HC, Aoki ST. In vivo pulse-chase in C. elegans reveals intestinal histone turnover changes upon starvation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.13.638128. [PMID: 39990428 PMCID: PMC11844474 DOI: 10.1101/2025.02.13.638128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
The ability to study protein dynamics and function in the authentic context of a multicellular organism is paramount to better understand biological phenomena in animal health and disease. Pulse-chase of self-labeling fusion protein tags provide the opportunity to label proteins of interest and track those proteins over time. There are currently several challenges associated with performing in vivo protein pulse-chase in animals, such as cost, reproducibility, and accurate detection methods. The C. elegans model organism has attributes that alleviate many of these challenges. This work tests the feasibility of applying the Halo modified enzyme (HaloTag) for in vivo protein pulse-chase in C. elegans. HaloTag intestinal histone reporters were created in the worm and used to demonstrate that reporter protein could be efficiently pulse-labeled by soaking animals in ligand. Labeled protein stability could be monitored over time by fluorescent confocal microscopy. Further investigation revealed reporter protein stability was dependent on the animal's nutritional state. Chromatin Immunoprecipitation and sequencing (ChIP-seq) of the reporters showed incorporation in chromatin with little change hours into starvation, implying a lack of chromatin regulation at the time point tested. Collectively, this work presents a straightforward method to label and track proteins of interest in C. elegans that can address a multitude of biological questions surrounding protein stability and dynamics in this animal model.
Collapse
Affiliation(s)
- Christopher Borchers
- Department of Biochemistry and Molecular Biology; School of Medicine; Indiana University Indianapolis; Indianapolis, IN, 46202
- Indiana BioMedical Gateway (IBMG) Program; School of Medicine; Indiana University Indianapolis; Indianapolis, IN, 46202
| | - Kara Osburn
- Department of Biochemistry and Molecular Biology; School of Medicine; Indiana University Indianapolis; Indianapolis, IN, 46202
| | - Hyun Cheol Roh
- Department of Biochemistry and Molecular Biology; School of Medicine; Indiana University Indianapolis; Indianapolis, IN, 46202
| | - Scott T. Aoki
- Department of Biochemistry and Molecular Biology; School of Medicine; Indiana University Indianapolis; Indianapolis, IN, 46202
| |
Collapse
|
12
|
Ma R, Han K. Concerns regarding the interpretation of Shank3 protein isoforms expressed in Shank3B -/- mice: potential off-target effects by a neomycin resistance cassette. Mol Psychiatry 2025; 30:791-792. [PMID: 39191866 DOI: 10.1038/s41380-024-02720-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 08/20/2024] [Accepted: 08/22/2024] [Indexed: 08/29/2024]
Affiliation(s)
- Ruiying Ma
- Department of Neuroscience, Korea University College of Medicine, Seoul, Republic of Korea
- Department of Biomedical Sciences, BK21 Graduate Program, Korea University College of Medicine, Seoul, Republic of Korea
| | - Kihoon Han
- Department of Neuroscience, Korea University College of Medicine, Seoul, Republic of Korea.
- Department of Biomedical Sciences, BK21 Graduate Program, Korea University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
13
|
Lovatt C, O'Sullivan TJ, Luis CODS, Ryan TJ, Frank RAW. Memory engram synapse 3D molecular architecture visualized by cryoCLEM-guided cryoET. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.09.632151. [PMID: 39829918 PMCID: PMC11741270 DOI: 10.1101/2025.01.09.632151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Memory is incorporated into the brain as physicochemical changes to engram cells. These are neuronal populations that form complex neuroanatomical circuits, are modified by experiences to store information, and allow for memory recall. At the molecular level, learning modifies synaptic communication to rewire engram circuits, a mechanism known as synaptic plasticity. However, despite its functional role on memory formation, the 3D molecular architecture of synapses within engram circuits is unknown. Here, we demonstrate the use of engram labelling technology and cryogenic correlated light and electron microscopy (cryoCLEM)-guided cryogenic electron tomography (cryoET) to visualize the in-tissue 3D molecular architecture of engram synapses of a contextual fear memory within the CA1 region of the mouse hippocampus. Engram cells exhibited structural diversity of macromolecular constituents and organelles in both pre- and postsynaptic compartments and within the synaptic cleft, including in clusters of membrane proteins, synaptic vesicle occupancy, and F-actin copy number. This 'engram to tomogram' approach, harnessing in vivo functional neuroscience and structural biology, provides a methodological framework for testing fundamental molecular plasticity mechanisms within engram circuits during memory encoding, storage and recall.
Collapse
Affiliation(s)
- Charlie Lovatt
- Astbury Centre for Structural Biology, School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Thomas J O'Sullivan
- Astbury Centre for Structural Biology, School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Clara Ortega-de San Luis
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
- Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
- Department of Health Sciences, University of Jaén, Jaén, Spain
| | - Tomás J Ryan
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
- Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Melbourne, Victoria, Australia
- Child & Brain Development Program, Canadian Institute for Advanced Research (CIFAR), Toronto, Ontario, Canada
| | - René A W Frank
- Astbury Centre for Structural Biology, School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
14
|
Daly S, Bulovaite E, Handa A, Morris K, Muresan L, Adams C, Kaizuka T, Kitching A, Spark A, Chant G, O′Holleran K, Grant SGN, Horrocks MH, Lee SF. 3D Super-Resolution Imaging of PSD95 Reveals an Abundance of Diffuse Protein Supercomplexes in the Mouse Brain. ACS Chem Neurosci 2025; 16:40-51. [PMID: 39702971 PMCID: PMC11697326 DOI: 10.1021/acschemneuro.4c00684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 12/09/2024] [Accepted: 12/10/2024] [Indexed: 12/21/2024] Open
Abstract
PSD95 is an abundant scaffolding protein that assembles multiprotein complexes controlling synaptic physiology and behavior. Confocal microscopy has previously shown that PSD95 is enriched in the postsynaptic terminals of excitatory synapses and two-dimensional (2D) super-resolution microscopy further revealed that it forms nanoclusters. In this study, we utilized three-dimensional (3D) super-resolution microscopy to examine the nanoarchitecture of PSD95 in the mouse brain, characterizing the spatial arrangement of over 8 million molecules. While we were able to identify molecular arrangements that have been previously reported, imaging in 3D allowed us to classify these with higher accuracy. Furthermore, 3D super-resolution microscopy enabled the quantification of protein levels, revealing that an abundance of PSD95 molecules existed outside of synapses as a diffuse population of supercomplexes, containing multiple copies of PSD95. Further analysis of the supercomplexes containing two units identified two populations: one that had PSD95 molecules separated by 39 ± 2 nm, and a second with a separation of 94 ± 27 nm. The finding that there exists supercomplexes containing two PSD95 units outside of the synapse suggests that supercomplexes containing multiple protein copies assemble outside the synapse and then integrate into the synapse to form a supramolecular nanocluster architecture.
Collapse
Affiliation(s)
- Sam Daly
- Yusuf
Hamied Department of Chemistry, University
of Cambridge, Cambridge CB2 1EW, U.K.
| | - Edita Bulovaite
- Genes
to Cognition Program, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, U.K.
| | - Anoushka Handa
- Yusuf
Hamied Department of Chemistry, University
of Cambridge, Cambridge CB2 1EW, U.K.
| | - Katie Morris
- RR Chemistry
Hub, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh EH16 4UU, U.K.
- EaStCHEM
School of Chemistry, University of Edinburgh, Edinburgh EH9 3FJ, U.K.
| | - Leila Muresan
- Cambridge
Advanced Imaging Centre, University of Cambridge, Cambridge CB2 3DY, U.K.
| | - Candace Adams
- RR Chemistry
Hub, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh EH16 4UU, U.K.
- EaStCHEM
School of Chemistry, University of Edinburgh, Edinburgh EH9 3FJ, U.K.
| | - Takeshi Kaizuka
- RR Chemistry
Hub, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh EH16 4UU, U.K.
- EaStCHEM
School of Chemistry, University of Edinburgh, Edinburgh EH9 3FJ, U.K.
| | | | | | - Gregory Chant
- Yusuf
Hamied Department of Chemistry, University
of Cambridge, Cambridge CB2 1EW, U.K.
| | - Kevin O′Holleran
- Cambridge
Advanced Imaging Centre, University of Cambridge, Cambridge CB2 3DY, U.K.
- ZOMP, Maxwell
Centre, JJ Thomson Avenue, Cambridge CB3 0HE, U.K.
| | - Seth G. N. Grant
- Genes
to Cognition Program, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, U.K.
| | - Mathew H. Horrocks
- RR Chemistry
Hub, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh EH16 4UU, U.K.
- EaStCHEM
School of Chemistry, University of Edinburgh, Edinburgh EH9 3FJ, U.K.
| | - Steven F. Lee
- Yusuf
Hamied Department of Chemistry, University
of Cambridge, Cambridge CB2 1EW, U.K.
| |
Collapse
|
15
|
Uchigashima M, Mikuni T. Single-cell synaptome mapping: its technical basis and applications in critical period plasticity research. Front Neural Circuits 2024; 18:1523614. [PMID: 39726910 PMCID: PMC11670323 DOI: 10.3389/fncir.2024.1523614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 11/28/2024] [Indexed: 12/28/2024] Open
Abstract
Our brain adapts to the environment by optimizing its function through experience-dependent cortical plasticity. This plasticity is transiently enhanced during a developmental stage, known as the "critical period," and subsequently maintained at lower levels throughout adulthood. Thus, understanding the mechanism underlying critical period plasticity is crucial for improving brain adaptability across the lifespan. Critical period plasticity relies on activity-dependent circuit remodeling through anatomical and functional changes at individual synapses. However, it remains challenging to identify the molecular signatures of synapses responsible for critical period plasticity and to understand how these plasticity-related synapses are spatiotemporally organized within a neuron. Recent advances in genetic tools and genome editing methodologies have enabled single-cell endogenous protein labeling in the brain, allowing for comprehensive molecular profiling of individual synapses within a neuron, namely "single-cell synaptome mapping." This promising approach can facilitate insights into the spatiotemporal organization of synapses that are sparse yet functionally important within single neurons. In this review, we introduce the basics of single-cell synaptome mapping and discuss its methodologies and applications to investigate the synaptic and cellular mechanisms underlying circuit remodeling during the critical period.
Collapse
Affiliation(s)
- Motokazu Uchigashima
- Department of Cellular Neuropathology, Brain Research Institute, Niigata University, Niigata, Japan
- International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo, Tokyo, Japan
| | - Takayasu Mikuni
- Department of Cellular Neuropathology, Brain Research Institute, Niigata University, Niigata, Japan
| |
Collapse
|
16
|
Morris K, Bulovaite E, Kaizuka T, Schnorrenberg S, Adams CT, Komiyama N, Mendive-Tapia L, Grant SGN, Horrocks MH. Sequential replacement of PSD95 subunits in postsynaptic supercomplexes is slowest in the cortex. eLife 2024; 13:RP99303. [PMID: 39570289 PMCID: PMC11581426 DOI: 10.7554/elife.99303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2024] Open
Abstract
The concept that dimeric protein complexes in synapses can sequentially replace their subunits has been a cornerstone of Francis Crick's 1984 hypothesis, explaining how long-term memories could be maintained in the face of short protein lifetimes. However, it is unknown whether the subunits of protein complexes that mediate memory are sequentially replaced in the brain and if this process is linked to protein lifetime. We address these issues by focusing on supercomplexes assembled by the abundant postsynaptic scaffolding protein PSD95, which plays a crucial role in memory. We used single-molecule detection, super-resolution microscopy and MINFLUX to probe the molecular composition of PSD95 supercomplexes in mice carrying genetically encoded HaloTags, eGFP, and mEoS2. We found a population of PSD95-containing supercomplexes comprised of two copies of PSD95, with a dominant 12.7 nm separation. Time-stamping of PSD95 subunits in vivo revealed that each PSD95 subunit was sequentially replaced over days and weeks. Comparison of brain regions showed subunit replacement was slowest in the cortex, where PSD95 protein lifetime is longest. Our findings reveal that protein supercomplexes within the postsynaptic density can be maintained by gradual replacement of individual subunits providing a mechanism for stable maintenance of their organization. Moreover, we extend Crick's model by suggesting that synapses with slow subunit replacement of protein supercomplexes and long-protein lifetimes are specialized for long-term memory storage and that these synapses are highly enriched in superficial layers of the cortex where long-term memories are stored.
Collapse
Affiliation(s)
- Katie Morris
- EaStCHEM School of Chemistry, University of EdinburghEdinburghUnited Kingdom
| | - Edita Bulovaite
- Genes to Cognition Program, Centre for Clinical Brain Sciences, University of EdinburghEdinburghUnited Kingdom
| | - Takeshi Kaizuka
- Genes to Cognition Program, Centre for Clinical Brain Sciences, University of EdinburghEdinburghUnited Kingdom
| | | | - Candace T Adams
- EaStCHEM School of Chemistry, University of EdinburghEdinburghUnited Kingdom
- IRR Chemistry Hub, Institute for Regeneration and Repair, University of EdinburghEdinburghUnited Kingdom
| | - Noboru Komiyama
- Genes to Cognition Program, Centre for Clinical Brain Sciences, University of EdinburghEdinburghUnited Kingdom
- Simons Initiative for the Developing Brain (SIDB), Centre for Discovery Brain Sciences, University of EdinburghEdinburghUnited Kingdom
- The Patrick Wild Centre for Research into Autism, Fragile X Syndrome & Intellectual Disabilities, Centre for Discovery Brain Sciences, University of EdinburghEdinburghUnited Kingdom
| | - Lorena Mendive-Tapia
- IRR Chemistry Hub, Institute for Regeneration and Repair, University of EdinburghEdinburghUnited Kingdom
- Centre for Inflammation Research, University of EdinburghEdinburghUnited Kingdom
| | - Seth GN Grant
- Genes to Cognition Program, Centre for Clinical Brain Sciences, University of EdinburghEdinburghUnited Kingdom
- Simons Initiative for the Developing Brain (SIDB), Centre for Discovery Brain Sciences, University of EdinburghEdinburghUnited Kingdom
| | - Mathew H Horrocks
- EaStCHEM School of Chemistry, University of EdinburghEdinburghUnited Kingdom
- IRR Chemistry Hub, Institute for Regeneration and Repair, University of EdinburghEdinburghUnited Kingdom
| |
Collapse
|
17
|
Gay SM, Chartampila E, Lord JS, Grizzard S, Maisashvili T, Ye M, Barker NK, Mordant AL, Mills CA, Herring LE, Diering GH. Developing forebrain synapses are uniquely vulnerable to sleep loss. Proc Natl Acad Sci U S A 2024; 121:e2407533121. [PMID: 39441640 PMCID: PMC11536182 DOI: 10.1073/pnas.2407533121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 09/19/2024] [Indexed: 10/25/2024] Open
Abstract
Sleep is an essential behavior that supports lifelong brain health and cognition. Neuronal synapses are a major target for restorative sleep function and a locus of dysfunction in response to sleep deprivation (SD). Synapse density is highly dynamic during development, becoming stabilized with maturation to adulthood, suggesting sleep exerts distinct synaptic functions between development and adulthood. Importantly, problems with sleep are common in neurodevelopmental disorders including autism spectrum disorder (ASD). Moreover, early life sleep disruption in animal models causes long-lasting changes in adult behavior. Divergent plasticity engaged during sleep necessarily implies that developing and adult synapses will show differential vulnerability to SD. To investigate distinct sleep functions and mechanisms of vulnerability to SD across development, we systematically examined the behavioral and molecular responses to acute SD between juvenile (P21 to P28), adolescent (P42 to P49), and adult (P70 to P100) mice of both sexes. Compared to adults, juveniles lack robust adaptations to SD, precipitating cognitive deficits in the novel object recognition task. Subcellular fractionation, combined with proteome and phosphoproteome analysis revealed the developing synapse is profoundly vulnerable to SD, whereas adults exhibit comparative resilience. SD in juveniles, and not older mice, aberrantly drives induction of synapse potentiation, synaptogenesis, and expression of perineuronal nets. Our analysis further reveals the developing synapse as a putative node of convergence between vulnerability to SD and ASD genetic risk. Together, our systematic analysis supports a distinct developmental function of sleep and reveals how sleep disruption impacts key aspects of brain development, providing insights for ASD susceptibility.
Collapse
Affiliation(s)
- Sean M. Gay
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Elissavet Chartampila
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Julia S. Lord
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Sawyer Grizzard
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Tekla Maisashvili
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Michael Ye
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Natalie K. Barker
- University of North Carolina Proteomics Core Facility, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Angie L. Mordant
- University of North Carolina Proteomics Core Facility, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - C. Allie Mills
- University of North Carolina Proteomics Core Facility, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Laura E. Herring
- University of North Carolina Proteomics Core Facility, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Graham H. Diering
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
- Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
- Carolina Institute for Developmental Disabilities, Carrboro, NC27510
| |
Collapse
|
18
|
Sterin I, Niazi A, Kim J, Park J, Park S. Dynamic Organization of Neuronal Extracellular Matrix Revealed by HaloTag-HAPLN1. J Neurosci 2024; 44:e0666242024. [PMID: 39251350 PMCID: PMC11502233 DOI: 10.1523/jneurosci.0666-24.2024] [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: 04/09/2024] [Revised: 08/04/2024] [Accepted: 08/30/2024] [Indexed: 09/11/2024] Open
Abstract
The brain's extracellular matrix (ECM) regulates neuronal plasticity and animal behavior. ECM staining shows a net-like structure around a subset of neurons, a ring-like structure at the nodes of Ranvier, and diffuse staining in the interstitial matrix. However, understanding the structural features of ECM deposition across various neuronal types and subcellular compartments remains limited. To visualize the organization pattern and assembly process of the hyaluronan-scaffolded ECM in the brain, we fused a HaloTag to hyaluronan proteoglycan link protein 1, which links hyaluronan and proteoglycans. Expression or application of the probe in primary rat neuronal cultures enables us to identify spatial and temporal regulation of ECM deposition and heterogeneity in ECM aggregation among neuronal populations. Dual-color birthdating shows the ECM assembly process in culture and in vivo. Sparse expression in mouse brains of either sex reveals detailed ECM architectures around excitatory neurons and developmentally regulated dendritic ECM. Our study uncovers extensive structural features of the brain's ECM, suggesting diverse roles in regulating neuronal plasticity.
Collapse
Affiliation(s)
- Igal Sterin
- Department of Neurobiology, University of Utah, Salt Lake City, Utah 84112
| | - Ava Niazi
- Department of Neurobiology, University of Utah, Salt Lake City, Utah 84112
- Neuroscience Program, University of Utah, Salt Lake City, Utah 84112
| | - Jennifer Kim
- Department of Neurobiology, University of Utah, Salt Lake City, Utah 84112
| | - Joosang Park
- Department of Neurobiology, University of Utah, Salt Lake City, Utah 84112
| | - Sungjin Park
- Department of Neurobiology, University of Utah, Salt Lake City, Utah 84112
| |
Collapse
|
19
|
Cavarischia-Rega C, Sharma K, Fitzgerald JC, Macek B. Proteome Dynamics in iPSC-Derived Human Dopaminergic Neurons. Mol Cell Proteomics 2024; 23:100838. [PMID: 39251023 PMCID: PMC11474371 DOI: 10.1016/j.mcpro.2024.100838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 08/18/2024] [Accepted: 08/25/2024] [Indexed: 09/11/2024] Open
Abstract
Dopaminergic neurons participate in fundamental physiological processes and are the cell type primarily affected in Parkinson's disease. Their analysis is challenging due to the intricate nature of their function, involvement in diverse neurological processes, and heterogeneity and localization in deep brain regions. Consequently, most of the research on the protein dynamics of dopaminergic neurons has been performed in animal cells ex vivo. Here we use iPSC-derived human mid-brain-specific dopaminergic neurons to study general features of their proteome biology and provide datasets for protein turnover and dynamics, including a human axonal translatome. We cover the proteome to a depth of 9409 proteins and use dynamic SILAC to measure the half-life of more than 4300 proteins. We report uniform turnover rates of conserved cytosolic protein complexes such as the proteasome and map the variable rates of turnover of the respiratory chain complexes in these cells. We use differential dynamic SILAC labeling in combination with microfluidic devices to analyze local protein synthesis and transport between axons and soma. We report 105 potentially novel axonal markers and detect translocation of 269 proteins between axons and the soma in the time frame of our analysis (120 h). Importantly, we provide evidence for local synthesis of 154 proteins in the axon and their retrograde transport to the soma, among them several proteins involved in RNA editing such as ADAR1 and the RNA helicase DHX30, involved in the assembly of mitochondrial ribosomes. Our study provides a workflow and resource for the future applications of quantitative proteomics in iPSC-derived human neurons.
Collapse
Affiliation(s)
- Claudia Cavarischia-Rega
- Quantitative Proteomics, Department of Biology, Interfaculty Institute of Cell Biology, University of Tübingen, Tübingen, Germany
| | - Karan Sharma
- Department of Neurodegeneration, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Julia C Fitzgerald
- Department of Neurodegeneration, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany.
| | - Boris Macek
- Quantitative Proteomics, Department of Biology, Interfaculty Institute of Cell Biology, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
20
|
Zaninello M, Baptista P, Duarte FV. Mitochondrial Dynamics and mRNA Translation: A Local Synaptic Tale. BIOLOGY 2024; 13:746. [PMID: 39336173 PMCID: PMC11428642 DOI: 10.3390/biology13090746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 09/17/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024]
Abstract
Mitochondria are dynamic organelles that can adjust and respond to different stimuli within a cell. This plastic ability allows them to effectively coordinate several cellular functions in cells and becomes particularly relevant in highly complex cells such as neurons. An imbalance in mitochondrial dynamics can disrupt mitochondrial function, leading to abnormal cellular function and ultimately to a range of diseases, including neurodegenerative disorders. Regulation of mRNA transport and local translation inside neurons is crucial for maintaining the proteome of distal mitochondria, which is vital for energy production and synaptic function. A significant portion of the axonal transcriptome is dedicated to mRNAs for mitochondrial proteins, emphasizing the importance of local translation in sustaining mitochondrial function in areas far from the cell body. In neurons, local translation and the regulation of mRNAs encoding mitochondrial-shaping proteins could be essential for synaptic plasticity and neuronal health. The dynamics of these mRNAs, including their transport and local translation, may influence the morphology and function of mitochondria, thereby affecting the overall energy status and responsiveness of synapses. Comprehending the mitochondria-related mRNA regulation and local translation, as well as its influence on mitochondrial morphology near the synapses will help to better understand neuronal physiology and neurological diseases where mitochondrial dysfunction and impaired synaptic plasticity play a central role.
Collapse
Affiliation(s)
- Marta Zaninello
- Institute for Genetics, University of Cologne, 50931 Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), 50931 Cologne, Germany
| | - Pedro Baptista
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
- Department of Life Sciences, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Filipe V Duarte
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
- Department of Life Sciences, University of Coimbra, 3004-504 Coimbra, Portugal
| |
Collapse
|
21
|
Koukaroudi D, Qiu Z, Fransén E, Gokhale R, Bulovaite E, Komiyama NH, Seibt J, Grant SGN. Sleep maintains excitatory synapse diversity in the cortex and hippocampus. Curr Biol 2024; 34:3836-3843.e5. [PMID: 39096907 PMCID: PMC11359089 DOI: 10.1016/j.cub.2024.07.032] [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: 12/11/2023] [Revised: 05/17/2024] [Accepted: 07/05/2024] [Indexed: 08/05/2024]
Abstract
Insufficient sleep is a global problem with serious consequences for cognition and mental health.1 Synapses play a central role in many aspects of cognition, including the crucial function of memory consolidation during sleep.2 Interference with the normal expression or function of synapse proteins is a cause of cognitive, mood, and other behavioral problems in over 130 brain disorders.3 Sleep deprivation (SD) has also been reported to alter synapse protein composition and synapse number, although with conflicting results.4,5,6,7 In our study, we conducted synaptome mapping of excitatory synapses in 125 regions of the mouse brain and found that sleep deprivation selectively reduces synapse diversity in the cortex and in the CA1 region of the hippocampus. Sleep deprivation targeted specific types and subtypes of excitatory synapses while maintaining total synapse density (synapse number/area). Synapse subtypes with longer protein lifetimes exhibited resilience to sleep deprivation, similar to observations in aging and genetic perturbations. Moreover, the altered synaptome architecture affected the responses to neural oscillations, suggesting that sleep plays a vital role in preserving cognitive function by maintaining the brain's synaptome architecture.
Collapse
Affiliation(s)
- Dimitra Koukaroudi
- Genes to Cognition Program, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Zhen Qiu
- Genes to Cognition Program, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK; School of Science and Engineering, University of Dundee, Dundee DD1 4HN, UK
| | - Erik Fransén
- Department of Computational Science and Technology, School of Electrical Engineering and Computer Science, KTH Royal Institute of Technology, 10044 Stockholm, Sweden; Science for Life Laboratory, KTH Royal Institute of Technology, 171 65 Solna, Sweden
| | - Ragini Gokhale
- Genes to Cognition Program, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Edita Bulovaite
- Genes to Cognition Program, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Noboru H Komiyama
- Genes to Cognition Program, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK; Simons Initiative for the Developing Brain (SIDB), Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK; The Patrick Wild Centre for Research into Autism, Fragile X Syndrome & Intellectual Disabilities, Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK; Muir Maxwell Epilepsy Centre, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Julie Seibt
- Surrey Sleep Research Centre, School of Biosciences, University of Surrey, Guildford, Surrey GU2 7XP, UK
| | - Seth G N Grant
- Genes to Cognition Program, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK; Simons Initiative for the Developing Brain (SIDB), Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK.
| |
Collapse
|
22
|
Michalska JM, Lyudchik J, Velicky P, Štefaničková H, Watson JF, Cenameri A, Sommer C, Amberg N, Venturino A, Roessler K, Czech T, Höftberger R, Siegert S, Novarino G, Jonas P, Danzl JG. Imaging brain tissue architecture across millimeter to nanometer scales. Nat Biotechnol 2024; 42:1051-1064. [PMID: 37653226 PMCID: PMC11252008 DOI: 10.1038/s41587-023-01911-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 07/20/2023] [Indexed: 09/02/2023]
Abstract
Mapping the complex and dense arrangement of cells and their connectivity in brain tissue demands nanoscale spatial resolution imaging. Super-resolution optical microscopy excels at visualizing specific molecules and individual cells but fails to provide tissue context. Here we developed Comprehensive Analysis of Tissues across Scales (CATS), a technology to densely map brain tissue architecture from millimeter regional to nanometer synaptic scales in diverse chemically fixed brain preparations, including rodent and human. CATS uses fixation-compatible extracellular labeling and optical imaging, including stimulated emission depletion or expansion microscopy, to comprehensively delineate cellular structures. It enables three-dimensional reconstruction of single synapses and mapping of synaptic connectivity by identification and analysis of putative synaptic cleft regions. Applying CATS to the mouse hippocampal mossy fiber circuitry, we reconstructed and quantified the synaptic input and output structure of identified neurons. We furthermore demonstrate applicability to clinically derived human tissue samples, including formalin-fixed paraffin-embedded routine diagnostic specimens, for visualizing the cellular architecture of brain tissue in health and disease.
Collapse
Affiliation(s)
- Julia M Michalska
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Julia Lyudchik
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Philipp Velicky
- Institute of Science and Technology Austria, Klosterneuburg, Austria
- Core Facility Imaging, Medical University of Vienna, Vienna, Austria
| | - Hana Štefaničková
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Jake F Watson
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Alban Cenameri
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Christoph Sommer
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Nicole Amberg
- Department of Neurology, Division of Neuropathology and Neurochemistry, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | | | - Karl Roessler
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
- Department of Neurosurgery, Medical University of Vienna, Vienna, Austria
| | - Thomas Czech
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
- Department of Neurosurgery, Medical University of Vienna, Vienna, Austria
| | - Romana Höftberger
- Department of Neurology, Division of Neuropathology and Neurochemistry, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - Sandra Siegert
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Gaia Novarino
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Peter Jonas
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Johann G Danzl
- Institute of Science and Technology Austria, Klosterneuburg, Austria.
| |
Collapse
|
23
|
Choi YK, Feng L, Jeong WK, Kim J. Connecto-informatics at the mesoscale: current advances in image processing and analysis for mapping the brain connectivity. Brain Inform 2024; 11:15. [PMID: 38833195 DOI: 10.1186/s40708-024-00228-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 05/08/2024] [Indexed: 06/06/2024] Open
Abstract
Mapping neural connections within the brain has been a fundamental goal in neuroscience to understand better its functions and changes that follow aging and diseases. Developments in imaging technology, such as microscopy and labeling tools, have allowed researchers to visualize this connectivity through high-resolution brain-wide imaging. With this, image processing and analysis have become more crucial. However, despite the wealth of neural images generated, access to an integrated image processing and analysis pipeline to process these data is challenging due to scattered information on available tools and methods. To map the neural connections, registration to atlases and feature extraction through segmentation and signal detection are necessary. In this review, our goal is to provide an updated overview of recent advances in these image-processing methods, with a particular focus on fluorescent images of the mouse brain. Our goal is to outline a pathway toward an integrated image-processing pipeline tailored for connecto-informatics. An integrated workflow of these image processing will facilitate researchers' approach to mapping brain connectivity to better understand complex brain networks and their underlying brain functions. By highlighting the image-processing tools available for fluroscent imaging of the mouse brain, this review will contribute to a deeper grasp of connecto-informatics, paving the way for better comprehension of brain connectivity and its implications.
Collapse
Affiliation(s)
- Yoon Kyoung Choi
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, South Korea
- Department of Computer Science and Engineering, Korea University, Seoul, South Korea
| | | | - Won-Ki Jeong
- Department of Computer Science and Engineering, Korea University, Seoul, South Korea
| | - Jinhyun Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, South Korea.
- Department of Computer Science and Engineering, Korea University, Seoul, South Korea.
- KIST-SKKU Brain Research Center, SKKU Institute for Convergence, Sungkyunkwan University, Suwon, South Korea.
| |
Collapse
|
24
|
Kaizuka T, Takumi T. Alteration of synaptic protein composition during developmental synapse maturation. Eur J Neurosci 2024; 59:2894-2914. [PMID: 38571321 DOI: 10.1111/ejn.16304] [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: 05/01/2023] [Revised: 01/02/2024] [Accepted: 02/07/2024] [Indexed: 04/05/2024]
Abstract
The postsynaptic density (PSD) is a collection of specialized proteins assembled beneath the postsynaptic membrane of dendritic spines. The PSD proteome comprises ~1000 proteins, including neurotransmitter receptors, scaffolding proteins and signalling enzymes. Many of these proteins have essential roles in synaptic function and plasticity. During brain development, changes are observed in synapse density and in the stability and shape of spines, reflecting the underlying molecular maturation of synapses. Synaptic protein composition changes in terms of protein abundance and the assembly of protein complexes, supercomplexes and the physical organization of the PSD. Here, we summarize the developmental alterations of postsynaptic protein composition during synapse maturation. We describe major PSD proteins involved in postsynaptic signalling that regulates synaptic plasticity and discuss the effect of altered expression of these proteins during development. We consider the abnormality of synaptic profiles and synaptic protein composition in the brain in neurodevelopmental disorders such as autism spectrum disorders. We also explain differences in synapse development between rodents and primates in terms of synaptic profiles and protein composition. Finally, we introduce recent findings related to synaptic diversity and nanoarchitecture and discuss their impact on future research. Synaptic protein composition can be considered a major determinant and marker of synapse maturation in normality and disease.
Collapse
Affiliation(s)
- Takeshi Kaizuka
- Department of Physiology and Cell Biology, Kobe University School of Medicine, Kobe, Japan
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Toru Takumi
- Department of Physiology and Cell Biology, Kobe University School of Medicine, Kobe, Japan
- RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| |
Collapse
|
25
|
Wu Z, Zhang H, Chen X, Zhang P, Fang J, Yang S, Chen H, Ji J, Chen L, Zheng Y, Yu D, Zhao Y. miR-145a-5p/SIK1/cAMP-dependent alteration of synaptic structural plasticity drives cognitive impairment induced by coke oven emissions. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 277:116401. [PMID: 38677069 DOI: 10.1016/j.ecoenv.2024.116401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 04/22/2024] [Accepted: 04/24/2024] [Indexed: 04/29/2024]
Abstract
Exposure to fine particulate matter (PM) is associated with the neurodegenerative diseases. Coke oven emissions (COEs) in occupational environment are important sources of PM. However, its neurotoxicity is still unclear. Therefore, evaluating the toxicological effects of COE on the nervous system is necessary. In the present study, we constructed mouse models of COE exposure by tracheal instillation. Mice exposed to COE showed signs of cognitive impairment. This was accompanied by a decrease in miR-145a-5p and an increase in SIK1 expression in the hippocampus, along with synaptic structural damage. Our results demonstrated that COE-induced miR-145a-5p downregulation could increase the expression of SIK1 and phosphorylated SIK1, inhibiting the cAMP/PKA/CREB pathway by activating PDE4D, which was associated with reduced synaptic structural plasticity. Furthermore, restoring of miR-145a-5p expression based on COE exposure in HT22 cells could partially reversed the negative effects of COE exposure through the SIK1/PDE4D/cAMP axis. Collectively, our findings link epigenetic regulation with COE-induced neurotoxicity and imply that miR-145a-5p could be an early diagnostic marker for neurological diseases in patients with COE occupational exposure.
Collapse
Affiliation(s)
- Zhaoxu Wu
- Department of Toxicology, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Heng Zhang
- Department of Toxicology, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Xian Chen
- Department of Toxicology, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Pimei Zhang
- Department of Toxicology, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Jiacheng Fang
- Department of Nutrition, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Shuaishuai Yang
- Department of Toxicology, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Hongguang Chen
- Department of Toxicology, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Jing Ji
- Department of Toxicology, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Lei Chen
- Department of Nutrition, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Yuxin Zheng
- Department of Toxicology, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Dianke Yu
- Department of Toxicology, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Yanjie Zhao
- Department of Toxicology, School of Public Health, Qingdao University, Qingdao 266071, China.
| |
Collapse
|
26
|
Dashkova AS, Kovalev VI, Chaplygina AV, Zhdanova DY, Bobkova NV. Unique Properties of Synaptosomes and Prospects for Their Use for the Treatment of Alzheimer's Disease. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:1031-1044. [PMID: 38981699 DOI: 10.1134/s0006297924060051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 03/21/2024] [Accepted: 03/23/2024] [Indexed: 07/11/2024]
Abstract
Alzheimer's disease (AD) is a severe neurodegenerative condition affecting millions worldwide. Prevalence of AD correlates with increased life expectancy and aging population in the developed countries. Considering that AD is a multifactorial disease involving various pathological processes such as synaptic dysfunction, neuroinflammation, oxidative stress, and improper protein folding, a comprehensive approach targeting multiple pathways may prove effective in slowing the disease progression. Cellular therapy and its further development in the form of cell vesicle and particularly mitochondrial transplantation represent promising approaches for treating neurodegeneration. The use of synaptosomes, due to uniqueness of their contents, could mark a new stage in the development of comprehensive therapies for neurodegenerative diseases, particularly AD. Synaptosomes contain unique memory mitochondria, which differ not only in size but also in functionality compared to the mitochondria in the neuronal soma. These synaptosomal mitochondria actively participate in cellular communication and signal transmission within synapses. Synaptosomes also contain other elements such as their own protein synthesis machinery, synaptic vesicles with neurotransmitters, synaptic adhesion molecules, and microRNAs - all crucial for synaptic transmission and, consequently, cognitive processes. Complex molecular ensemble ensures maintenance of the synaptic autonomy of mitochondria. Additionally, synaptosomes, with their affinity for neurons, can serve as an optimal platform for targeted drug delivery to nerve cells. This review discusses unique composition of synaptosomes, their capabilities and advantages, as well as limitations of their suggested use as therapeutic agents for treating neurodegenerative pathologies, particularly AD.
Collapse
Affiliation(s)
- Alla S Dashkova
- Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - Vladimir I Kovalev
- Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia.
| | - Alina V Chaplygina
- Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - Daria Yu Zhdanova
- Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - Natalia V Bobkova
- Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| |
Collapse
|
27
|
Jones G, Akter Y, Shifflett V, Hruska M. Nanoscale analysis of functionally diverse glutamatergic synapses in the neocortex reveals input and layer-specific organization. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.01.592008. [PMID: 38746319 PMCID: PMC11092571 DOI: 10.1101/2024.05.01.592008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Discovery of synaptic nanostructures suggests a molecular logic for the flexibility of synaptic function. We still have little understanding of how functionally diverse synapses in the brain organize their nanoarchitecture due to challenges associated with super-resolution imaging in complex brain tissue. Here, we characterized single-domain camelid nanobodies for the 3D quantitative multiplex imaging of synaptic nano-organization in 6 µm brain cryosections using STED nanoscopy. We focused on thalamocortical (TC) and corticocortical (CC) synapses along the apical-basal axis of layer 5 pyramidal neurons as models of functionally diverse glutamatergic synapses in the brain. Spines receiving TC input were larger than CC spines in all layers examined. However, TC synapses on apical and basal dendrites conformed to different organizational principles. TC afferents on apical dendrites frequently contacted spines with multiple aligned PSD-95/Bassoon nanomodules, which are larger. TC spines on basal dendrites contained mostly one aligned PSD-95/Bassoon nanocluster. However, PSD-95 nanoclusters were larger and scaled with spine volume. The nano-organization of CC synapses did not change across cortical layers. These results highlight striking nanoscale diversity of functionally distinct glutamatergic synapses, relying on afferent input and sub-cellular localization of individual synaptic connections.
Collapse
|
28
|
Sterin I, Niazi A, Kim J, Park J, Park S. Novel extracellular matrix architecture on excitatory neurons revealed by HaloTag-HAPLN1. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.29.587384. [PMID: 38585814 PMCID: PMC10996768 DOI: 10.1101/2024.03.29.587384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
The brain's extracellular matrix (ECM) regulates neuronal plasticity and animal behavior. ECM staining shows an aggregated pattern in a net-like structure around a subset of neurons and diffuse staining in the interstitial matrix. However, understanding the structural features of ECM deposition across various neuronal types and subcellular compartments remains limited. To visualize the organization pattern and assembly process of the hyaluronan-scaffolded ECM in the brain, we fused a HaloTag to HAPLN1, which links hyaluronan and proteoglycans. Expression or application of the probe enables us to identify spatial and temporal regulation of ECM deposition and heterogeneity in ECM aggregation among neuronal populations. Dual-color birthdating shows the ECM assembly process in culture and in vivo. Sparse expression in vivo reveals novel forms of ECM architecture around excitatory neurons and developmentally regulated dendritic ECM. Overall, our study uncovers extensive structural features of the brain' ECM, suggesting diverse roles in regulating neuronal plasticity.
Collapse
Affiliation(s)
- Igal Sterin
- Department of Neurobiology, University of Utah, Salt Lake City, Utah, USA
| | - Ava Niazi
- Department of Neurobiology, University of Utah, Salt Lake City, Utah, USA
- Neuroscience Program, University of Utah, Salt Lake City, Utah, USA
| | - Jennifer Kim
- Department of Neurobiology, University of Utah, Salt Lake City, Utah, USA
| | - Joosang Park
- Department of Neurobiology, University of Utah, Salt Lake City, Utah, USA
| | - Sungjin Park
- Department of Neurobiology, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
29
|
López-Otín C, Kroemer G. The missing hallmark of health: psychosocial adaptation. Cell Stress 2024; 8:21-50. [PMID: 38476764 PMCID: PMC10928495 DOI: 10.15698/cst2024.03.294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 02/09/2024] [Accepted: 02/12/2024] [Indexed: 03/14/2024] Open
Abstract
The eight biological hallmarks of health that we initially postulated (Cell. 2021 Jan 7;184(1):33-63) include features of spatial compartmentalization (integrity of barriers, containment of local perturbations), maintenance of homeostasis over time (recycling & turnover, integration of circuitries, rhythmic oscillations) and an array of adequate responses to stress (homeostatic resilience, hormetic regulation, repair & regeneration). These hallmarks affect all eight somatic strata of the human body (molecules, organelles, cells, supracellular units, organs, organ systems, systemic circuitries and meta-organism). Here we postulate that mental and socioeconomic factors must be added to this 8×8 matrix as an additional hallmark of health ("psychosocial adaptation") and as an additional stratum ("psychosocial interactions"), hence building a 9×9 matrix. Potentially, perturbation of each of the somatic hallmarks and strata affects psychosocial factors and vice versa. Finally, we discuss the (patho)physiological bases of these interactions and their implications for mental health improvement.
Collapse
Affiliation(s)
- Carlos López-Otín
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Facultad de Ciencias de la Vida y la Naturaleza, Universidad Nebrija, Madrid, Spain
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| |
Collapse
|
30
|
Jiang G, Liu H, Liu H, Ke G, Ren TB, Xiong B, Zhang XB, Yuan L. Chemical Approaches to Optimize the Properties of Organic Fluorophores for Imaging and Sensing. Angew Chem Int Ed Engl 2024; 63:e202315217. [PMID: 38081782 DOI: 10.1002/anie.202315217] [Citation(s) in RCA: 34] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Indexed: 12/30/2023]
Abstract
Organic fluorophores are indispensable tools in cells, tissue and in vivo imaging, and have enabled much progress in the wide range of biological and biomedical fields. However, many available dyes suffer from insufficient performances, such as short absorption and emission wavelength, low brightness, poor stability, small Stokes shift, and unsuitable permeability, restricting their application in advanced imaging technology and complex imaging. Over the past two decades, many efforts have been made to improve these performances of fluorophores. Starting with the luminescence principle of fluorophores, this review clarifies the mechanisms of the insufficient performance for traditional fluorophores to a certain extent, systematically summarizes the modified approaches of optimizing properties, highlights the typical applications of the improved fluorophores in imaging and sensing, and indicates existing problems and challenges in this area. This progress not only proves the significance of improving fluorophores properties, but also provide a theoretical guidance for the development of high-performance fluorophores.
Collapse
Affiliation(s)
- Gangwei Jiang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, 410082, Changsha, P. R. China
| | - Han Liu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, 410082, Changsha, P. R. China
| | - Hong Liu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, 410082, Changsha, P. R. China
| | - Guoliang Ke
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, 410082, Changsha, P. R. China
| | - Tian-Bing Ren
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, 410082, Changsha, P. R. China
| | - Bin Xiong
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, 410082, Changsha, P. R. China
| | - Xiao-Bing Zhang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, 410082, Changsha, P. R. China
| | - Lin Yuan
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, 410082, Changsha, P. R. China
| |
Collapse
|
31
|
Huppertz MC, Wilhelm J, Grenier V, Schneider MW, Falt T, Porzberg N, Hausmann D, Hoffmann DC, Hai L, Tarnawski M, Pino G, Slanchev K, Kolb I, Acuna C, Fenk LM, Baier H, Hiblot J, Johnsson K. Recording physiological history of cells with chemical labeling. Science 2024; 383:890-897. [PMID: 38386755 DOI: 10.1126/science.adg0812] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 01/22/2024] [Indexed: 02/24/2024]
Abstract
Recordings of the physiological history of cells provide insights into biological processes, yet obtaining such recordings is a challenge. To address this, we introduce a method to record transient cellular events for later analysis. We designed proteins that become labeled in the presence of both a specific cellular activity and a fluorescent substrate. The recording period is set by the presence of the substrate, whereas the cellular activity controls the degree of the labeling. The use of distinguishable substrates enabled the recording of successive periods of activity. We recorded protein-protein interactions, G protein-coupled receptor activation, and increases in intracellular calcium. Recordings of elevated calcium levels allowed selections of cells from heterogeneous populations for transcriptomic analysis and tracking of neuronal activities in flies and zebrafish.
Collapse
Affiliation(s)
- Magnus-Carsten Huppertz
- Department of Chemical Biology, Max Planck Institute for Medical Research, Jahnstrasse 29, 69120 Heidelberg, Germany
| | - Jonas Wilhelm
- Department of Chemical Biology, Max Planck Institute for Medical Research, Jahnstrasse 29, 69120 Heidelberg, Germany
| | - Vincent Grenier
- Department of Chemical Biology, Max Planck Institute for Medical Research, Jahnstrasse 29, 69120 Heidelberg, Germany
| | - Martin W Schneider
- Department Genes - Circuits - Behavior, Max Planck Institute for Biological Intelligence, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Tjalda Falt
- Active Sensing, Max Planck Institute for Biological Intelligence, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Nicola Porzberg
- Department of Chemical Biology, Max Planck Institute for Medical Research, Jahnstrasse 29, 69120 Heidelberg, Germany
| | - David Hausmann
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dirk C Hoffmann
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neurology and Neurooncology Program, National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Ling Hai
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neurology and Neurooncology Program, National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
- Bioinformatics and Omics Data Analytics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Miroslaw Tarnawski
- Protein Expression and Characterization Facility, Max Planck Institute for Medical Research, Jahnstrasse 29, 69120 Heidelberg, Germany
| | - Gabriela Pino
- Chica and Heinz Schaller Foundation, Institute of Anatomy and Cell Biology, Heidelberg University, Im Neuenheimer Feld 307, 69120 Heidelberg, Germany
| | - Krasimir Slanchev
- Department Genes - Circuits - Behavior, Max Planck Institute for Biological Intelligence, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Ilya Kolb
- GENIE Project Team, Janelia Research Campus, Ashburn, VA 20147, USA
| | - Claudio Acuna
- Chica and Heinz Schaller Foundation, Institute of Anatomy and Cell Biology, Heidelberg University, Im Neuenheimer Feld 307, 69120 Heidelberg, Germany
| | - Lisa M Fenk
- Active Sensing, Max Planck Institute for Biological Intelligence, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Herwig Baier
- Department Genes - Circuits - Behavior, Max Planck Institute for Biological Intelligence, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Julien Hiblot
- Department of Chemical Biology, Max Planck Institute for Medical Research, Jahnstrasse 29, 69120 Heidelberg, Germany
| | - Kai Johnsson
- Department of Chemical Biology, Max Planck Institute for Medical Research, Jahnstrasse 29, 69120 Heidelberg, Germany
- Institute of Chemical Sciences and Engineering (ISIC), École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| |
Collapse
|
32
|
Dejanovic B, Sheng M, Hanson JE. Targeting synapse function and loss for treatment of neurodegenerative diseases. Nat Rev Drug Discov 2024; 23:23-42. [PMID: 38012296 DOI: 10.1038/s41573-023-00823-1] [Citation(s) in RCA: 42] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/06/2023] [Indexed: 11/29/2023]
Abstract
Synapse dysfunction and loss are hallmarks of neurodegenerative diseases that correlate with cognitive decline. However, the mechanisms and therapeutic strategies to prevent or reverse synaptic damage remain elusive. In this Review, we discuss recent advances in understanding the molecular and cellular pathways that impair synapses in neurodegenerative diseases, including the effects of protein aggregation and neuroinflammation. We also highlight emerging therapeutic approaches that aim to restore synaptic function and integrity, such as enhancing synaptic plasticity, preventing synaptotoxicity, modulating neuronal network activity and targeting immune signalling. We discuss the preclinical and clinical evidence for each strategy, as well as the challenges and opportunities for developing effective synapse-targeting therapeutics for neurodegenerative diseases.
Collapse
Affiliation(s)
| | - Morgan Sheng
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jesse E Hanson
- Department of Neuroscience, Genentech, South San Francisco, CA, USA.
| |
Collapse
|
33
|
Parkins EV, Burwinkel JM, Ranatunga R, Yaser S, Hu YC, Tiwari D, Gross C. Age-Dependent Regulation of Dendritic Spine Density and Protein Expression in Mir324 KO Mice. J Mol Neurosci 2023; 73:818-830. [PMID: 37773316 PMCID: PMC10793736 DOI: 10.1007/s12031-023-02157-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 09/08/2023] [Indexed: 10/01/2023]
Abstract
Dendritic spines are small, dynamic protrusions along the dendrite that comprise more than 90% of excitatory connections in the brain, making them essential sites for neuronal communication. These synaptic sites change throughout the process of development, reducing in density and shifting morphology as synapses are refined. One important class of dendritic spine regulators is microRNA (miRNA), small-noncoding RNAs that post-transcriptionally regulate gene expression. Several studies suggest that miRNA-324-5p regulates dendritic spine formation. In addition, we have previously shown that miR-324-5p plays a role in seizure and long-term potentiation, both of which involve dendritic spine changes. In this study, we aimed to characterize the role of miRNA-324-5p in developmental spine regulation by assessing the effect of Mir324 knockout (KO) on dendritic spine density and expression of a subset of dendritic proteins at select developmental time points. We show that miR-324-5p expression is developmentally regulated and peaks at 4 weeks of age. We demonstrate that loss of miR-324-5p expression leads to differential changes in both target protein expression and spine density at different time points during development, disrupting the pattern of spine density changes and leading to a premature loss of dendritic spines in KO mice, which is compensated later. Our findings indicate that miR-324-5p plays a role in synaptic refinement across development. Additionally, our data illustrate the importance of context in the study of miRNA, as regulation by and/or of miRNA can vary dramatically across development and in disease.
Collapse
Affiliation(s)
- Emma V Parkins
- University of Cincinnati Neuroscience Graduate Program, Cincinnati, OH, 45229, USA
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - John M Burwinkel
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Ruvi Ranatunga
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Sarah Yaser
- University of Cincinnati Neuroscience Graduate Program, Cincinnati, OH, 45229, USA
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Yueh-Chiang Hu
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA
- Transgenic Animal and Genome Editing Core Facility, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Durgesh Tiwari
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA
| | - Christina Gross
- University of Cincinnati Neuroscience Graduate Program, Cincinnati, OH, 45229, USA.
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA.
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA.
| |
Collapse
|
34
|
Berson E, Gajera CR, Phongpreecha T, Perna A, Bukhari SA, Becker M, Chang AL, De Francesco D, Espinosa C, Ravindra NG, Postupna N, Latimer CS, Shively CA, Register TC, Craft S, Montine KS, Fox EJ, Keene CD, Bendall SC, Aghaeepour N, Montine TJ. Cross-species comparative analysis of single presynapses. Sci Rep 2023; 13:13849. [PMID: 37620363 PMCID: PMC10449792 DOI: 10.1038/s41598-023-40683-8] [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: 03/13/2023] [Accepted: 08/16/2023] [Indexed: 08/26/2023] Open
Abstract
Comparing brain structure across species and regions enables key functional insights. Leveraging publicly available data from a novel mass cytometry-based method, synaptometry by time of flight (SynTOF), we applied an unsupervised machine learning approach to conduct a comparative study of presynapse molecular abundance across three species and three brain regions. We used neural networks and their attractive properties to model complex relationships among high dimensional data to develop a unified, unsupervised framework for comparing the profile of more than 4.5 million single presynapses among normal human, macaque, and mouse samples. An extensive validation showed the feasibility of performing cross-species comparison using SynTOF profiling. Integrative analysis of the abundance of 20 presynaptic proteins revealed near-complete separation between primates and mice involving synaptic pruning, cellular energy, lipid metabolism, and neurotransmission. In addition, our analysis revealed a strong overlap between the presynaptic composition of human and macaque in the cerebral cortex and neostriatum. Our unique approach illuminates species- and region-specific variation in presynapse molecular composition.
Collapse
Affiliation(s)
- Eloïse Berson
- Department of Pathology, Stanford University, 300 Pasteur Dr., Stanford, CA, 94304, USA
- Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University, Stanford, CA, USA
| | - Chandresh R Gajera
- Department of Pathology, Stanford University, 300 Pasteur Dr., Stanford, CA, 94304, USA
| | - Thanaphong Phongpreecha
- Department of Pathology, Stanford University, 300 Pasteur Dr., Stanford, CA, 94304, USA
- Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University, Stanford, CA, USA
| | - Amalia Perna
- Department of Pathology, Stanford University, 300 Pasteur Dr., Stanford, CA, 94304, USA
| | - Syed A Bukhari
- Department of Pathology, Stanford University, 300 Pasteur Dr., Stanford, CA, 94304, USA
| | - Martin Becker
- Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University, Stanford, CA, USA
| | - Alan L Chang
- Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University, Stanford, CA, USA
| | - Davide De Francesco
- Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University, Stanford, CA, USA
| | - Camilo Espinosa
- Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University, Stanford, CA, USA
| | - Neal G Ravindra
- Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University, Stanford, CA, USA
| | - Nadia Postupna
- Department of Laboratory Medicine & Pathology, University of Washington, Seattle, WA, USA
| | - Caitlin S Latimer
- Department of Laboratory Medicine & Pathology, University of Washington, Seattle, WA, USA
| | - Carol A Shively
- Department of Pathology/Comparative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Thomas C Register
- Department of Pathology/Comparative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Suzanne Craft
- Department of Internal Medicine-Geriatrics, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Kathleen S Montine
- Department of Pathology, Stanford University, 300 Pasteur Dr., Stanford, CA, 94304, USA
| | - Edward J Fox
- Department of Pathology, Stanford University, 300 Pasteur Dr., Stanford, CA, 94304, USA
| | - C Dirk Keene
- Department of Laboratory Medicine & Pathology, University of Washington, Seattle, WA, USA
| | - Sean C Bendall
- Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University, Stanford, CA, USA
| | - Nima Aghaeepour
- Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University, Stanford, CA, USA
- Department of Pediatrics, Stanford University, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Thomas J Montine
- Department of Pathology, Stanford University, 300 Pasteur Dr., Stanford, CA, 94304, USA.
| |
Collapse
|
35
|
Parkins EV, Burwinkel JM, Ranatunga R, Yaser S, Hu YC, Tiwari D, Gross C. Age-dependent regulation of dendritic spine density and protein expression in Mir324 KO mice. RESEARCH SQUARE 2023:rs.3.rs-3221779. [PMID: 37609225 PMCID: PMC10441466 DOI: 10.21203/rs.3.rs-3221779/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Dendritic spines are small, dynamic protrusions along the dendrite that comprise more than 90% of excitatory connections in the brain, making them essential sites for neuronal communication. These synaptic sites change throughout the process of development, reducing in density and shifting morphology as synapses are refined. One important class of dendritic spine regulators is microRNA (miRNA), small noncoding RNAs that post-transcriptionally regulate gene expression. Several studies suggest that miRNA-324-5p regulates dendritic spine formation. In addition, we have previously shown that miR-324-5p plays a role in seizure and long-term potentiation, both of which involve dendritic spine changes. In this study, we aimed to characterize the role of miRNA-324-5p in developmental spine regulation by assessing the effect of Mir324 knockout (KO) on dendritic spine density and expression of a subset of dendritic proteins at select developmental time points. We show that miR-324-5p expression is developmentally regulated and peaks at four weeks of age. We demonstrate that loss of miR-324-5p expression leads to differential changes in both target protein expression and spine density at different time points during development, disrupting the pattern of spine density changes and leading to a premature loss of dendritic spines in KO mice, which is compensated later. Our findings indicate that miR-324-5p plays a role in synaptic refinement across development. Additionally, our data illustrate the importance of context in the study of miRNA, as regulation by and/or of miRNA can vary dramatically across development and in disease.
Collapse
Affiliation(s)
| | | | | | - Sarah Yaser
- Cincinnati Children's Hospital Medical Center
| | | | | | | |
Collapse
|
36
|
Zhu WH, Yang XX, Gou XZ, Fu SM, Chen JH, Gao F, Shen Y, Bi DL, Tang AH. Nanoscale reorganisation of synaptic proteins in Alzheimer's disease. Neuropathol Appl Neurobiol 2023; 49:e12924. [PMID: 37461203 DOI: 10.1111/nan.12924] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 05/30/2023] [Accepted: 06/24/2023] [Indexed: 08/31/2023]
Abstract
AIMS Synaptic strength depends strongly on the subsynaptic organisation of presynaptic transmitter release and postsynaptic receptor densities, and their alterations are expected to underlie pathologies. Although synaptic dysfunctions are common pathogenic traits of Alzheimer's disease (AD), it remains unknown whether synaptic protein nano-organisation is altered in AD. Here, we systematically characterised the alterations in the subsynaptic organisation in cellular and mouse models of AD. METHODS We used immunostaining and super-resolution stochastic optical reconstruction microscopy imaging to quantitatively examine the synaptic protein nano-organisation in both Aβ1-42-treated neuronal cultures and cortical sections from a mouse model of AD, APP23 mice. RESULTS We found that Aβ1-42-treatment of cultured hippocampal neurons decreased the synaptic retention of postsynaptic scaffolds and receptors and disrupted their nanoscale alignment to presynaptic transmitter release sites. In cortical sections, we found that while GluA1 receptors in wild-type mice were organised in subsynaptic nanoclusters with high local densities, receptors in APP23 mice distributed more homogeneously within synapses. This reorganisation, together with the reduced overall receptor density, led to reduced glutamatergic synaptic transmission. Meanwhile, the transsynaptic alignment between presynaptic release-guiding RIM1/2 and postsynaptic scaffolding protein PSD-95 was reduced in APP23 mice. Importantly, these reorganisations were progressive with age and were more pronounced in synapses in close vicinity of Aβ plaques with dense cores. CONCLUSIONS Our study revealed a spatiotemporal-specific reorganisation of synaptic nanostructures in AD and identifies dense-core amyloid plaques as the major local inductor in APP23 mice.
Collapse
Affiliation(s)
- Wang-Hui Zhu
- Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, Ministry of Education Key Laboratory for Membrane-less Organelles and Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, China
| | - Xiao-Xu Yang
- Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, Ministry of Education Key Laboratory for Membrane-less Organelles and Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Sciences and Technology of China, Hefei, China
- Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei, China
| | - Xu-Zhuo Gou
- Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, Ministry of Education Key Laboratory for Membrane-less Organelles and Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, China
| | - Shu-Mei Fu
- Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, Ministry of Education Key Laboratory for Membrane-less Organelles and Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Sciences and Technology of China, Hefei, China
- Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei, China
| | - Jia-Hui Chen
- Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, Ministry of Education Key Laboratory for Membrane-less Organelles and Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, China
- Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei, China
| | - Feng Gao
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Sciences and Technology of China, Hefei, China
| | - Yong Shen
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Sciences and Technology of China, Hefei, China
- Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei, China
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, China
| | - Dan-Lei Bi
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, China
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Sciences and Technology of China, Hefei, China
- Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei, China
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, China
| | - Ai-Hui Tang
- Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, Ministry of Education Key Laboratory for Membrane-less Organelles and Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, China
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Sciences and Technology of China, Hefei, China
- Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei, China
| |
Collapse
|
37
|
Velicky P, Miguel E, Michalska JM, Lyudchik J, Wei D, Lin Z, Watson JF, Troidl J, Beyer J, Ben-Simon Y, Sommer C, Jahr W, Cenameri A, Broichhagen J, Grant SGN, Jonas P, Novarino G, Pfister H, Bickel B, Danzl JG. Dense 4D nanoscale reconstruction of living brain tissue. Nat Methods 2023; 20:1256-1265. [PMID: 37429995 PMCID: PMC10406607 DOI: 10.1038/s41592-023-01936-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 05/22/2023] [Indexed: 07/12/2023]
Abstract
Three-dimensional (3D) reconstruction of living brain tissue down to an individual synapse level would create opportunities for decoding the dynamics and structure-function relationships of the brain's complex and dense information processing network; however, this has been hindered by insufficient 3D resolution, inadequate signal-to-noise ratio and prohibitive light burden in optical imaging, whereas electron microscopy is inherently static. Here we solved these challenges by developing an integrated optical/machine-learning technology, LIONESS (live information-optimized nanoscopy enabling saturated segmentation). This leverages optical modifications to stimulated emission depletion microscopy in comprehensively, extracellularly labeled tissue and previous information on sample structure via machine learning to simultaneously achieve isotropic super-resolution, high signal-to-noise ratio and compatibility with living tissue. This allows dense deep-learning-based instance segmentation and 3D reconstruction at a synapse level, incorporating molecular, activity and morphodynamic information. LIONESS opens up avenues for studying the dynamic functional (nano-)architecture of living brain tissue.
Collapse
Affiliation(s)
- Philipp Velicky
- Institute of Science and Technology Austria (ISTA), Klosterneuburg, Austria
- Core Facility Imaging, Medical University of Vienna, Vienna, Austria
| | - Eder Miguel
- Institute of Science and Technology Austria (ISTA), Klosterneuburg, Austria
| | - Julia M Michalska
- Institute of Science and Technology Austria (ISTA), Klosterneuburg, Austria
| | - Julia Lyudchik
- Institute of Science and Technology Austria (ISTA), Klosterneuburg, Austria
| | - Donglai Wei
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Department of Computer Science, Boston College, Boston, MA, USA
| | - Zudi Lin
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Jake F Watson
- Institute of Science and Technology Austria (ISTA), Klosterneuburg, Austria
| | - Jakob Troidl
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Johanna Beyer
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Yoav Ben-Simon
- Institute of Science and Technology Austria (ISTA), Klosterneuburg, Austria
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Christoph Sommer
- Institute of Science and Technology Austria (ISTA), Klosterneuburg, Austria
| | - Wiebke Jahr
- Institute of Science and Technology Austria (ISTA), Klosterneuburg, Austria
- In-Vision Technologies, Guntramsdorf, Austria
| | - Alban Cenameri
- Institute of Science and Technology Austria (ISTA), Klosterneuburg, Austria
| | | | - Seth G N Grant
- Genes to Cognition Program, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- Simons Initiative for the Developing Brain (SIDB), Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Peter Jonas
- Institute of Science and Technology Austria (ISTA), Klosterneuburg, Austria
| | - Gaia Novarino
- Institute of Science and Technology Austria (ISTA), Klosterneuburg, Austria
| | - Hanspeter Pfister
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Bernd Bickel
- Institute of Science and Technology Austria (ISTA), Klosterneuburg, Austria
| | - Johann G Danzl
- Institute of Science and Technology Austria (ISTA), Klosterneuburg, Austria.
| |
Collapse
|
38
|
Osanai H, Nair IR, Kitamura T. Dissecting cell-type-specific pathways in medial entorhinal cortical-hippocampal network for episodic memory. J Neurochem 2023; 166:172-188. [PMID: 37248771 PMCID: PMC10538947 DOI: 10.1111/jnc.15850] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/07/2023] [Accepted: 05/10/2023] [Indexed: 05/31/2023]
Abstract
Episodic memory, which refers to our ability to encode and recall past events, is essential to our daily lives. Previous research has established that both the entorhinal cortex (EC) and hippocampus (HPC) play a crucial role in the formation and retrieval of episodic memories. However, to understand neural circuit mechanisms behind these processes, it has become necessary to monitor and manipulate the neural activity in a cell-type-specific manner with high temporal precision during memory formation, consolidation, and retrieval in the EC-HPC networks. Recent studies using cell-type-specific labeling, monitoring, and manipulation have demonstrated that medial EC (MEC) contains multiple excitatory neurons that have differential molecular markers, physiological properties, and anatomical features. In this review, we will comprehensively examine the complementary roles of superficial layers of neurons (II and III) and the roles of deeper layers (V and VI) in episodic memory formation and recall based on these recent findings.
Collapse
Affiliation(s)
- Hisayuki Osanai
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Indrajith R Nair
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Takashi Kitamura
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
39
|
Duarte FV, Ciampi D, Duarte CB. Mitochondria as central hubs in synaptic modulation. Cell Mol Life Sci 2023; 80:173. [PMID: 37266732 DOI: 10.1007/s00018-023-04814-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 05/10/2023] [Accepted: 05/19/2023] [Indexed: 06/03/2023]
Abstract
Mitochondria are present in the pre- and post-synaptic regions, providing the energy required for the activity of these very specialized neuronal compartments. Biogenesis of synaptic mitochondria takes place in the cell body, and these organelles are then transported to the synapse by motor proteins that carry their cargo along microtubule tracks. The transport of mitochondria along neurites is a highly regulated process, being modulated by the pattern of neuronal activity and by extracellular cues that interact with surface receptors. These signals act by controlling the distribution of mitochondria and by regulating their activity. Therefore, mitochondria activity at the synapse allows the integration of different signals and the organelles are important players in the response to synaptic stimulation. Herein we review the available evidence regarding the regulation of mitochondrial dynamics by neuronal activity and by neuromodulators, and how these changes in the activity of mitochondria affect synaptic communication.
Collapse
Affiliation(s)
- Filipe V Duarte
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- III - Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Daniele Ciampi
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Carlos B Duarte
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.
- Department of Life Sciences, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
40
|
Kilisch M, Gere-Becker M, Wüstefeld L, Bonnas C, Crauel A, Mechmershausen M, Martens H, Götzke H, Opazo F, Frey S. Simple and Highly Efficient Detection of PSD95 Using a Nanobody and Its Recombinant Heavy-Chain Antibody Derivatives. Int J Mol Sci 2023; 24:ijms24087294. [PMID: 37108454 PMCID: PMC10138605 DOI: 10.3390/ijms24087294] [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: 03/16/2023] [Revised: 04/03/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
The post-synaptic density protein 95 (PSD95) is a crucial scaffolding protein participating in the organization and regulation of synapses. PSD95 interacts with numerous molecules, including neurotransmitter receptors and ion channels. The functional dysregulation of PSD95 as well as its abundance and localization has been implicated with several neurological disorders, making it an attractive target for developing strategies able to monitor PSD95 accurately for diagnostics and therapeutics. This study characterizes a novel camelid single-domain antibody (nanobody) that binds strongly and with high specificity to rat, mouse, and human PSD95. This nanobody allows for more precise detection and quantification of PSD95 in various biological samples. We expect that the flexibility and unique performance of this thoroughly characterized affinity tool will help to further understand the role of PSD95 in normal and diseased neuronal synapses.
Collapse
Affiliation(s)
- Markus Kilisch
- NanoTag Biotechnologies GmbH, Rudolf-Wissell-Straβe 28a, 37079 Göttingen, Germany
| | - Maja Gere-Becker
- NanoTag Biotechnologies GmbH, Rudolf-Wissell-Straβe 28a, 37079 Göttingen, Germany
| | - Liane Wüstefeld
- Synaptic Systems GmbH, Rudolf-Wissell-Straβe 28a, 37079 Göttingen, Germany
| | - Christel Bonnas
- Synaptic Systems GmbH, Rudolf-Wissell-Straβe 28a, 37079 Göttingen, Germany
| | - Alexander Crauel
- NanoTag Biotechnologies GmbH, Rudolf-Wissell-Straβe 28a, 37079 Göttingen, Germany
| | - Maja Mechmershausen
- NanoTag Biotechnologies GmbH, Rudolf-Wissell-Straβe 28a, 37079 Göttingen, Germany
| | - Henrik Martens
- Synaptic Systems GmbH, Rudolf-Wissell-Straβe 28a, 37079 Göttingen, Germany
| | - Hansjörg Götzke
- NanoTag Biotechnologies GmbH, Rudolf-Wissell-Straβe 28a, 37079 Göttingen, Germany
| | - Felipe Opazo
- NanoTag Biotechnologies GmbH, Rudolf-Wissell-Straβe 28a, 37079 Göttingen, Germany
- Institute of Neuro- and Sensory Physiology, University Medical Center Göttingen, 37073 Göttingen, Germany
- Center for Biostructural Imaging of Neurodegeneration (BIN), University of Göttingen Medical Center, 37075 Göttingen, Germany
| | - Steffen Frey
- NanoTag Biotechnologies GmbH, Rudolf-Wissell-Straβe 28a, 37079 Göttingen, Germany
| |
Collapse
|
41
|
Abstract
During aging, animals experience a decline in proteostasis activity, including loss of stress-response activation, culminating in the accumulation of misfolded proteins and toxic aggregates, which are causal in the onset of some chronic diseases. Finding genetic and pharmaceutical treatments that can increase organismal proteostasis and lengthen life is an ongoing goal of current research. The regulation of stress responses by cell non-autonomous mechanisms appears to be a potent way to impact organismal healthspan. In this Review, we cover recent findings in the intersection of proteostasis and aging, with a special focus on articles and preprints published between November 2021 and October 2022. A significant number of papers published during this time increased our understanding of how cells communicate with each other during proteotoxic stress. Finally, we also draw attention to emerging datasets that can be explored to generate new hypotheses that explain age-related proteostasis collapse.
Collapse
Affiliation(s)
- Maximilian A. Thompson
- Neurobiology Division, Medical Research Council Laboratory of Molecular Biology, Cambridge, CB2 0QH, UK
| | - Evandro A. De-Souza
- Neurobiology Division, Medical Research Council Laboratory of Molecular Biology, Cambridge, CB2 0QH, UK
| |
Collapse
|
42
|
O'Reilly CL, Miller BF, Lewis TL. Exercise and mitochondrial remodeling to prevent age-related neurodegeneration. J Appl Physiol (1985) 2023; 134:181-189. [PMID: 36519568 PMCID: PMC9829476 DOI: 10.1152/japplphysiol.00611.2022] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/30/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Healthy brain activity requires precise ion and energy management creating a strong reliance on mitochondrial function. Age-related neurodegeneration leads to a decline in mitochondrial function and increased oxidative stress, with associated declines in mitochondrial mass, respiration capacity, and respiration efficiency. The interdependent processes of mitochondrial protein turnover and mitochondrial dynamics, known together as mitochondrial remodeling, play essential roles in mitochondrial health and therefore brain function. This mini-review describes the role of mitochondria in neurodegeneration and brain health, current practices for assessing both aspects of mitochondrial remodeling, and how exercise mitigates the adverse effects of aging in the brain. Exercise training elicits functional adaptations to improve brain health, and current literature strongly suggests that mitochondrial remodeling plays a vital role in these positive adaptations. Despite substantial implications that the two aspects of mitochondrial remodeling are interdependent, very few investigations have simultaneously measured mitochondrial dynamics and protein synthesis. An improved understanding of the partnership between mitochondrial protein turnover and mitochondrial dynamics will provide a better understanding of their role in both brain health and disease, as well as how they induce protection following exercise.
Collapse
Affiliation(s)
- Colleen L O'Reilly
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
| | - Benjamin F Miller
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
- Oklahoma City Veterans Association, Oklahoma City, Oklahoma
| | - Tommy L Lewis
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
| |
Collapse
|
43
|
Moroz LL, Romanova DY. Alternative neural systems: What is a neuron? (Ctenophores, sponges and placozoans). Front Cell Dev Biol 2022; 10:1071961. [PMID: 36619868 PMCID: PMC9816575 DOI: 10.3389/fcell.2022.1071961] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022] Open
Abstract
How to make a neuron, a synapse, and a neural circuit? Is there only one 'design' for a neural architecture with a universally shared genomic blueprint across species? The brief answer is "No." Four early divergent lineages from the nerveless common ancestor of all animals independently evolved distinct neuroid-type integrative systems. One of these is a subset of neural nets in comb jellies with unique synapses; the second lineage is the well-known Cnidaria + Bilateria; the two others are non-synaptic neuroid systems in sponges and placozoans. By integrating scRNA-seq and microscopy data, we revise the definition of neurons as synaptically-coupled polarized and highly heterogenous secretory cells at the top of behavioral hierarchies with learning capabilities. This physiological (not phylogenetic) definition separates 'true' neurons from non-synaptically and gap junction-coupled integrative systems executing more stereotyped behaviors. Growing evidence supports the hypothesis of multiple origins of neurons and synapses. Thus, many non-bilaterian and bilaterian neuronal classes, circuits or systems are considered functional rather than genetic categories, composed of non-homologous cell types. In summary, little-explored examples of convergent neuronal evolution in representatives of early branching metazoans provide conceptually novel microanatomical and physiological architectures of behavioral controls in animals with prospects of neuro-engineering and synthetic biology.
Collapse
Affiliation(s)
- Leonid L. Moroz
- Departments of Neuroscience and McKnight Brain Institute, University of Florida, Gainesville, FL, United States
- Whitney Laboratory for Marine Bioscience, University of Florida, St. Augustine, FL, United States
| | - Daria Y. Romanova
- Institute of Higher Nervous Activity and Neurophysiology of RAS, 5A Butlerova, Moscow, Russia
| |
Collapse
|
44
|
Tomas-Roca L, Qiu Z, Fransén E, Gokhale R, Bulovaite E, Price DJ, Komiyama NH, Grant SGN. Developmental disruption and restoration of brain synaptome architecture in the murine Pax6 neurodevelopmental disease model. Nat Commun 2022; 13:6836. [PMID: 36369219 PMCID: PMC9652404 DOI: 10.1038/s41467-022-34131-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 10/14/2022] [Indexed: 11/13/2022] Open
Abstract
Neurodevelopmental disorders of genetic origin delay the acquisition of normal abilities and cause disabling phenotypes. Nevertheless, spontaneous attenuation and even complete amelioration of symptoms in early childhood and adolescence can occur in many disorders, suggesting that brain circuits possess an intrinsic capacity to overcome the deficits arising from some germline mutations. We examined the molecular composition of almost a trillion excitatory synapses on a brain-wide scale between birth and adulthood in mice carrying a mutation in the homeobox transcription factor Pax6, a neurodevelopmental disorder model. Pax6 haploinsufficiency had no impact on total synapse number at any age. By contrast, the molecular composition of excitatory synapses, the postnatal expansion of synapse diversity and the acquisition of normal synaptome architecture were delayed in all brain regions, interfering with networks and electrophysiological simulations of cognitive functions. Specific excitatory synapse types and subtypes were affected in two key developmental age-windows. These phenotypes were reversed within 2-3 weeks of onset, restoring synapse diversity and synaptome architecture to the normal developmental trajectory. Synapse subtypes with rapid protein turnover mediated the synaptome remodeling. This brain-wide capacity for remodeling of synapse molecular composition to recover and maintain the developmental trajectory of synaptome architecture may help confer resilience to neurodevelopmental genetic disorders.
Collapse
Affiliation(s)
- Laura Tomas-Roca
- Genes to Cognition Program, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, EH16 4SB, UK
| | - Zhen Qiu
- Genes to Cognition Program, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, EH16 4SB, UK
| | - Erik Fransén
- Science for Life Laboratory, KTH Royal Institute of Technology, SE-171 65, Solna, Sweden
| | - Ragini Gokhale
- Genes to Cognition Program, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, EH16 4SB, UK
| | - Edita Bulovaite
- Genes to Cognition Program, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, EH16 4SB, UK
| | - David J Price
- Simons Initiative for the Developing Brain (SIDB), Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH8 9XD, UK
| | - Noboru H Komiyama
- Genes to Cognition Program, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, EH16 4SB, UK
- Simons Initiative for the Developing Brain (SIDB), Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH8 9XD, UK
| | - Seth G N Grant
- Genes to Cognition Program, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, EH16 4SB, UK.
- Simons Initiative for the Developing Brain (SIDB), Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH8 9XD, UK.
| |
Collapse
|