1
|
Xu N, Chen SY, Tang AH. Tuning synapse strength by nanocolumn plasticity. Trends Neurosci 2025; 48:200-212. [PMID: 39848836 DOI: 10.1016/j.tins.2024.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 12/12/2024] [Accepted: 12/31/2024] [Indexed: 01/25/2025]
Abstract
The precise organization of the complex set of synaptic proteins at the nanometer scale is crucial for synaptic transmission. At the heart of this nanoscale architecture lies the nanocolumn. This aligns presynaptic neurotransmitter release with a high local density of postsynaptic receptor channels, thereby optimizing synaptic strength. Although synapses exhibit diverse protein compositions and nanoscale organizations, the role of structural diversity in the notable differences observed in synaptic physiology remains poorly understood. In this review we examine the current literature on the molecular mechanisms underlying the formation and maintenance of nanocolumns, as well as their role in modulating various aspects of synaptic transmission. We also discuss how the reorganization of nanocolumns contributes to functional dynamics in both synaptic plasticity and pathology.
Collapse
Affiliation(s)
- Na Xu
- Hefei National Laboratory for Physical Sciences at the Microscale, Department of Neurology in the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230088, China; School of Medicine, Anhui University of Science and Technology, Huainan 232001, China.
| | - Si-Yu Chen
- Hefei National Laboratory for Physical Sciences at the Microscale, Department of Neurology in the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230088, China
| | - Ai-Hui Tang
- Hefei National Laboratory for Physical Sciences at the Microscale, Department of Neurology in the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230088, China.
| |
Collapse
|
2
|
Gao W, Cai Q, Ying X, Zhao B. Establishing a Mouse Model of NL3R617W-Associated Autism Spectrum Disorder for a Functional Study. ACTAS ESPANOLAS DE PSIQUIATRIA 2025; 53:253-266. [PMID: 40071366 PMCID: PMC11898267 DOI: 10.62641/aep.v53i2.1780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 11/22/2024] [Accepted: 11/25/2024] [Indexed: 03/15/2025]
Abstract
BACKGROUND Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by deficits in social communication and limited behavior. Despite the association of numerous synaptic gene mutations with ASD, the presence of behavioral abnormalities in mice expressing autism-associated R617W mutation in synaptic adhesion protein neuroligin-3 (NL3) has not been established. This work focuses on establishing a mouse model of ASD caused by NL3 R617W missense mutation (NL3R617W) and characterizing and profiling the molecular as well as behavioral features of the animal model. METHODS The expression and distribution of NL3R617W mutant protein in the 293T cell membrane and intracellular NL3 was detected by using immunofluorescence approach. Meanwhile, synaptic markers (Synapsin I, vesicular glutamate transporter (VGluT) I and vesicular γ-aminobutyric acid transporter (VGAT)) and synapse number were detected with a confocal fluorescence microscope. Thereafter, the effect on NL3R617W was verified. The expression of synaptic proteins, postsynaptic density protein-95 (PSD95) and Src homology domain and multiple ankyrin repeat domains protein 3 (SHANK3), was verified by Western blot. The interaction between NL3 and neurexin 1 (NRXN1) was studied by means of co-immunoprecipitation. The behavior of autistic mice induced by NL3R617W mutation was examined using the Morris water maze and the Y maze. NL3R617W mutant mice were assessed in the open field, and three-chamber test was conducted to assess and observe the presence of hyperactivity, repetitive behavior, friendliness, and social novelty. RESULTS The results indicated that the NL3 mutation could influence the interaction between NL3 and NRXN1, and inhibit the expression of VGluT I. Nevertheless, NL3 mutation would not influence the expression of NL3 on cell membrane, the intracellular distribution of NL3, or the endoplasmic reticulum retention. The outcomes of animal studies demonstrated that the ASD mice with NL3R617W exhibited a significant decrease in the capacity for spatial memory and exploration, as well as the expression levels of the postsynaptic scaffolding proteins, PSD95 and SHANK3 (p < 0.05). The number of excitatory synapses in hippocampal cornu ammonis (CA)1 and CA3 and the sensory cortex was also significantly reduced (p < 0.01). Compared to the control mice, the NL3R617W mutant mice were less active in the open field (p < 0.001), a finding consistent with the three-chamber test result showing reduced degree of activity. Furthermore, compared to the control mice, the NL3R617W mutant animals spent less time with stranger mice (p < 0.05). CONCLUSIONS NL3R617W mutation may inhibit the expression of postsynaptic scaffolding proteins by influencing the interaction with NRXN1, thus inhibiting synapse formation and reducing the number of excitatory synapses.
Collapse
Affiliation(s)
- Wei Gao
- Children’s Health Department, Hongkou District Maternal and Child Health Institution, 200000 Shanghai, China
- Department of Pediatric, The First People’s Hospital of Taizhou, 318020 Taizhou, Zhejiang, China
| | - Qiao Cai
- Department of Pediatric, The First People’s Hospital of Taizhou, 318020 Taizhou, Zhejiang, China
| | - Xiaoming Ying
- Department of Pediatric, The First People’s Hospital of Taizhou, 318020 Taizhou, Zhejiang, China
| | - Bei Zhao
- Department of Pediatric, The First People’s Hospital of Taizhou, 318020 Taizhou, Zhejiang, China
| |
Collapse
|
3
|
Wang B, He T, Qiu G, Li C, Xue S, Zheng Y, Wang T, Xia Y, Yao L, Yan J, Chen Y. Altered synaptic homeostasis: a key factor in the pathophysiology of depression. Cell Biosci 2025; 15:29. [PMID: 40001206 PMCID: PMC11863845 DOI: 10.1186/s13578-025-01369-y] [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: 12/14/2024] [Accepted: 02/14/2025] [Indexed: 02/27/2025] Open
Abstract
Depression, a widespread psychiatric disorder, is characterized by a diverse array of symptoms such as melancholic mood and anhedonia, imposing a significant burden on both society and individuals. Despite extensive research into the neurobiological foundations of depression, a complete understanding of its complex mechanisms is yet to be attained, and targeted therapeutic interventions remain under development. Synaptic homeostasis, a compensatory feedback mechanism, involves neurons adjusting synaptic strength by regulating pre- or postsynaptic processes. Recent advancements in depression research reveal a crucial association between the disorder and disruptions in synaptic homeostasis within neural regions and circuits pivotal for emotional and cognitive functions. This paper explores the mechanisms governing synaptic homeostasis in depression, focusing on the role of ion channels, the regulation of presynaptic neurotransmitter release, synaptic scaling processes, and essential signaling molecules. By mapping new pathways in the study of synaptic homeostasis as it pertains to depression, this research aims to provide valuable insights for identifying novel therapeutic targets for more effective antidepressant treatments.
Collapse
Affiliation(s)
- Bokai Wang
- Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, 4655 University Road, Jinan, 250355, China
- Chinese Medicine Innovation Research Institute, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Teng He
- Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, 4655 University Road, Jinan, 250355, China
- Chinese Medicine Innovation Research Institute, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Guofan Qiu
- Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, 4655 University Road, Jinan, 250355, China
- Chinese Medicine Innovation Research Institute, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Chong Li
- Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, 4655 University Road, Jinan, 250355, China
- Chinese Medicine Innovation Research Institute, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Song Xue
- Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, 4655 University Road, Jinan, 250355, China
- Chinese Medicine Innovation Research Institute, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Yuanjia Zheng
- Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, 4655 University Road, Jinan, 250355, China
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China
- Shandong Key Laboratory of Innovation and Application Research in Basic Theory of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Taiyi Wang
- Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, 4655 University Road, Jinan, 250355, China
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China
- Shandong Key Laboratory of Innovation and Application Research in Basic Theory of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Yucen Xia
- Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, 4655 University Road, Jinan, 250355, China
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China
- Shandong Key Laboratory of Innovation and Application Research in Basic Theory of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Lin Yao
- Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, 4655 University Road, Jinan, 250355, China
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China
- Shandong Key Laboratory of Innovation and Application Research in Basic Theory of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Jinglan Yan
- Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, 4655 University Road, Jinan, 250355, China
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China
- Shandong Key Laboratory of Innovation and Application Research in Basic Theory of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Yongjun Chen
- Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, 4655 University Road, Jinan, 250355, China.
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China.
- Shandong Key Laboratory of Innovation and Application Research in Basic Theory of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| |
Collapse
|
4
|
Bay S, Rodina A, Haut F, Roychowdhury T, Argyrousi EK, Staniszewski A, Han K, Sharma S, Chakrabarty S, Digwal CS, Stanisavljevic A, Labuza A, Alldred MJ, Panchal P, SanthaSeela A, Tuffery L, Li Z, Hashmi A, Rosiek E, Chan E, Monetti M, Sasaguri H, Saido TC, Schneider JA, Bennett DA, Fraser PE, Erdjument-Bromage H, Neubert TA, Ginsberg SD, Arancio O, Chiosis G. Systems-Level Interactome Mapping Reveals Actionable Protein Network Dysregulation Across the Alzheimer's Disease Spectrum. RESEARCH SQUARE 2025:rs.3.rs-5930673. [PMID: 39989971 PMCID: PMC11844643 DOI: 10.21203/rs.3.rs-5930673/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Alzheimer's disease (AD) progresses as a continuum, from preclinical stages to late-stage cognitive decline, yet the molecular mechanisms driving this progression remain poorly understood. Here, we provide a systems-level map of protein-protein interaction (PPI) network dysfunction across the AD spectrum and uncover epichaperomes-stable scaffolding platforms formed by chaperones and co-factors-as central drivers of this process. Using over 100 human brain specimens, mouse models, and human neurons, we show that epichaperomes emerge early, even in preclinical AD, and progressively disrupt multiple PPI networks critical for synaptic function and neuroplasticity. Glutamatergic neurons, essential for learning and memory, exhibit heightened vulnerability, with their dysfunction driven by protein sequestration into epichaperome scaffolds, independent of changes in protein expression. Notably, pharmacological disruption of epichaperomes with PU-AD restores PPI network integrity and reverses synaptic and cognitive deficits, directly linking epichaperome-driven network dysfunction to AD pathology. These findings establish epichaperomes as key mediators of molecular collapse in AD and identify network-centric intervention strategies as a promising avenue for disease-modifying therapies.
Collapse
Affiliation(s)
- Sadik Bay
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Anna Rodina
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Florence Haut
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, New York, NY 10032, USA
| | - Tanaya Roychowdhury
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Elentina K Argyrousi
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, New York, NY 10032, USA
| | - Agnieszka Staniszewski
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, New York, NY 10032, USA
| | - Kyung Han
- Tanz Centre for Research in Neurodegenerative Diseases and Department of Medical Biophysics, University of Toronto, Toronto, ON M5R 0A3, Canada
| | - Sahil Sharma
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Souparna Chakrabarty
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Chander S Digwal
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | | | - Amanda Labuza
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, 10962, USA
| | - Melissa J Alldred
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, 10962, USA
- Department of Psychiatry, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Palak Panchal
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Anand SanthaSeela
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Laura Tuffery
- Proteomics Core, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Zhuoning Li
- Proteomics Core, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Arsalan Hashmi
- Proteomics Core, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Eric Rosiek
- Molecular Cytology Core, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Eric Chan
- Molecular Cytology Core, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Mara Monetti
- Proteomics Core, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Hiroki Sasaguri
- Laboratory for Proteolytic Neuroscience, RIKEN Brain Science Institute, Wako, Saitama 351-0198, Japan
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Brain Science Institute, Wako, Saitama 351-0198, Japan
| | - Julie A Schneider
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois 60612
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois 60612
| | - Paul E Fraser
- Tanz Centre for Research in Neurodegenerative Diseases and Department of Medical Biophysics, University of Toronto, Toronto, ON M5R 0A3, Canada
| | - Hediye Erdjument-Bromage
- Department of Neuroscience and Physiology, NYU Grossman School of Medicine, New York, NY, 10016, USA
| | - Thomas A Neubert
- Department of Neuroscience and Physiology, NYU Grossman School of Medicine, New York, NY, 10016, USA
- NYU Neuroscience Institute, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Stephen D Ginsberg
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, 10962, USA
- Department of Psychiatry, NYU Grossman School of Medicine, New York, NY 10016, USA
- NYU Neuroscience Institute, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Ottavio Arancio
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, New York, NY 10032, USA
- Department of Medicine, Columbia University, New York, NY 10032, USA
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Gabriela Chiosis
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Department of Medicine, Division of Solid Tumors, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
5
|
P A H, Basavaraju N, Gupta A, Kommaddi RP. Actin Cytoskeleton at the Synapse: An Alzheimer's Disease Perspective. Cytoskeleton (Hoboken) 2025. [PMID: 39840749 DOI: 10.1002/cm.21993] [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: 02/28/2024] [Revised: 12/18/2024] [Accepted: 01/05/2025] [Indexed: 01/23/2025]
Abstract
Actin, a ubiquitous and highly conserved cytoskeletal protein, plays a pivotal role in various cellular functions such as structural support, facilitating cell motility, and contributing to the dynamic processes of synaptic function. Apart from its established role in inducing morphological changes, recent developments in the field indicate an active involvement of actin in modulating both the structure and function of pre- and postsynaptic terminals. Within the presynapse, it is involved in the organization and trafficking of synaptic vesicles, contributing to neurotransmitter release. In the postsynapse, actin dynamically modulates dendritic spines, influencing the postsynaptic density organization and anchoring of neurotransmitter receptors. In addition, the dynamic interplay of actin at the synapse underscores its essential role in regulating neural communication. This review strives to offer a comprehensive overview of the recent advancements in understanding the multifaceted role of the actin cytoskeleton in synaptic functions. By emphasizing its aberrant regulation, we aim to provide valuable insights into the underlying mechanisms of Alzheimer's disease pathophysiology.
Collapse
Affiliation(s)
- Haseena P A
- Centre for Brain Research, Indian Institute of Science, Bangalore, India
- Manipal Academy of Higher Education, Manipal, India
| | - Nimisha Basavaraju
- Centre for Brain Research, Indian Institute of Science, Bangalore, India
- Manipal Academy of Higher Education, Manipal, India
| | - Anant Gupta
- Centre for Brain Research, Indian Institute of Science, Bangalore, India
- Manipal Academy of Higher Education, Manipal, India
| | | |
Collapse
|
6
|
Brill J, Clarke B, Hong I, Huganir RL. Dissociation of SYNGAP1 Enzymatic and Structural Roles: Intrinsic Excitability and Seizure Susceptibility. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.14.633019. [PMID: 39868300 PMCID: PMC11761602 DOI: 10.1101/2025.01.14.633019] [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: 01/28/2025]
Abstract
SYNGAP1 is a key Ras-GAP protein enriched at excitatory synapses, with mutations causing intellectual disability and epilepsy in humans. Recent studies have revealed that in addition to its role as a negative regulator of G-protein signaling through its GAP enzymatic activity, SYNGAP1 plays an important structural role through its interaction with post-synaptic density proteins. Here, we reveal that intrinsic excitability deficits and seizure phenotypes in heterozygous Syngap1 knockout (KO) mice are differentially dependent on Syngap1 GAP activity. Cortical excitatory neurons in heterozygous KO mice displayed reduced intrinsic excitability, including lower input resistance, and increased rheobase, a phenotype recapitulated in GAP-deficient Syngap1 mutants. However, seizure severity and susceptibility to pentylenetetrazol (PTZ)-induced seizures were significantly elevated in heterozygous KO mice but unaffected in GAP-deficient mutants, implicating the structural rather than enzymatic role of Syngap1 in seizure regulation. These findings highlight the complex interplay between SYNGAP1 structural and catalytic functions in neuronal physiology and disease.
Collapse
Affiliation(s)
- Julia Brill
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Blaise Clarke
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ingie Hong
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Richard L. Huganir
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
7
|
Won S, Sweeney CL, Roche KW. Biochemical Properties of Synaptic Proteins Are Dependent on Tissue Preparation: NMDA Receptor Solubility Is Regulated by the C-Terminal Tail. J Cell Biochem 2025; 126:e30664. [PMID: 39370692 PMCID: PMC11730348 DOI: 10.1002/jcb.30664] [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: 07/23/2024] [Revised: 09/04/2024] [Accepted: 09/17/2024] [Indexed: 10/08/2024]
Abstract
Synaptic proteins are essential for neuronal development, synaptic transmission, and synaptic plasticity. The postsynaptic density (PSD) is a membrane-associated structure at excitatory synapses, which is composed of a huge protein complex. To understand the interactions and functions of PSD proteins, researchers have employed a variety of imaging and biochemical approaches including sophisticated mass spectrometry. However, the field is lacking a systematic comparison of different experimental conditions and how they might influence the study of the PSD interactome isolated from various tissue preparations. To evaluate the efficiency of several common solubilization conditions, we isolated receptors, scaffolding proteins, and adhesion molecules from brain tissue or primary cultured neurons or human forebrain neurons differentiated from induced pluripotent stem cells (iPSCs). We observed some striking differences in solubility. We found that N-methyl-d-aspartate receptors (NMDARs) and PSD-95 are relatively insoluble in brain tissue, cultured neurons, and human forebrain neurons compared to α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic receptors (AMPARs) or SAP102. In general, synaptic proteins were more soluble in primary neuronal cultures and human forebrain neurons compared to brain tissue. Interestingly, NMDARs are relatively insoluble in HEK293T cells suggesting that insolubility does not directly represent the synaptic fraction but rather it is related to a detergent-insoluble fraction such as lipid rafts. Surprisingly, truncation of the intracellular carboxyl-terminal tail (C-tail) of NMDAR subunits increased NMDAR solubility in HEK293T cells. Our findings show that detergent, pH, and temperature are important for protein preparations to study PSD protein complexes, and NMDAR solubility is regulated by its C-tail, thus providing a technical guide to study synaptic interactomes and subcellular localization of synaptic proteins.
Collapse
Affiliation(s)
- Sehoon Won
- Receptor Biology SectionNational Institute of Neurological Disorders and Stroke, National Institutes of HealthBethesdaMarylandUSA
| | - Colin L. Sweeney
- Genetic Immunotherapy SectionNational Institute of Allergy and Infectious Diseases, National Institutes of HealthBethesdaMarylandUSA
| | - Katherine W. Roche
- Receptor Biology SectionNational Institute of Neurological Disorders and Stroke, National Institutes of HealthBethesdaMarylandUSA
| |
Collapse
|
8
|
Wutikeli H, Yu Y, Zhang T, Cao J, Nawy S, Shen Y. Role of Elavl-like RNA-binding protein in retinal development and signal transduction. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167518. [PMID: 39307290 DOI: 10.1016/j.bbadis.2024.167518] [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: 07/06/2024] [Revised: 08/25/2024] [Accepted: 09/16/2024] [Indexed: 09/29/2024]
Abstract
RNA-binding proteins (RBPs) play central roles in post-transcriptional gene regulation. However, the function of RBP in retinal progenitor cell differentiation and synaptic signal transmission are largely unexplored. Previously we have shown that Elavl2 regulates amacrine cell (AC) differentiation during retinogenesis, by directly binding to Nr4a2 and Barhl2. Elavl2 is expressed in early neuronal progenitors to mature neurons, and Elavl4 expression begins slightly later, during cortical neuron development as a paralog. Here, Retinal-specific Elavl2 and Elavl4 double knockout mice were made to further explore the role of Elavl2 and Elavl4 in retinal development and signal transduction. We disclose that Elavl4 binds to Satb1 to regulate Neurod1, then promoting retinal progenitor and amacrine cells differentiation. We were also surprised to find that Elavl2 interacted with GABAB receptors at the RNA and protein levels. In conclusion, Elavl2 and Elavl4 regulate amacrine cells differentiation through different pathways, leading to decreased scotopic vision. Our findings reveal the roles of Elavl2 and Elavl4 in retinal amacrine cells differentiation in modulating visual functions.
Collapse
Affiliation(s)
- Huxitaer Wutikeli
- Eye Center, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, PR China
| | - Yao Yu
- Eye Center, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, PR China
| | - Tianlu Zhang
- Eye Center, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, PR China
| | | | - Scott Nawy
- University of California Berkeley, Department of Molecular and Cell Biology, Berkeley, CA, USA
| | - Yin Shen
- Eye Center, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, PR China; Frontier Science Center for Immunology and Metabolism, Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China.
| |
Collapse
|
9
|
Krueger-Burg D. Understanding GABAergic synapse diversity and its implications for GABAergic pharmacotherapy. Trends Neurosci 2025; 48:47-61. [PMID: 39779392 DOI: 10.1016/j.tins.2024.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 10/17/2024] [Accepted: 11/21/2024] [Indexed: 01/11/2025]
Abstract
Despite the substantial contribution of disruptions in GABAergic inhibitory neurotransmission to the etiology of psychiatric, neurodevelopmental, and neurodegenerative disorders, surprisingly few drugs targeting the GABAergic system are currently available, partly due to insufficient understanding of circuit-specific GABAergic synapse biology. In addition to GABA receptors, GABAergic synapses contain an elaborate organizational protein machinery that regulates the properties of synaptic transmission. Until recently, this machinery remained largely unexplored, but key methodological advances have now led to the identification of a wealth of new GABAergic organizer proteins. Notably, many of these proteins appear to function only at specific subsets of GABAergic synapses, creating a diversity of organizer complexes that may serve as circuit-specific targets for pharmacotherapies. The present review aims to summarize the methodological developments that underlie this newfound knowledge and provide a current overview of synapse-specific GABAergic organizer complexes, as well as outlining future avenues and challenges in translating this knowledge into clinical applications.
Collapse
Affiliation(s)
- Dilja Krueger-Burg
- Laboratory of Cell Biology and Neuroscience, Institute of Anatomy, University Medical Center of the Johannes Gutenberg University Mainz, Duesbergweg 6, 55128 Mainz, Germany.
| |
Collapse
|
10
|
Barão S, Hong I, Müller U, Huganir RL. Syngap1 and the development of murine neocortical progenitor cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.18.629233. [PMID: 39763888 PMCID: PMC11702710 DOI: 10.1101/2024.12.18.629233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Abstract
SYNGAP1 is a major regulator of synaptic plasticity through its interaction with synaptic scaffold proteins and modulation of Ras and Rap GTPase signaling pathways. SYNGAP1 mutations in humans are often associated with intellectual disability, epilepsy, and autism spectrum disorder. Syngap1 heterozygous loss-of-function results in impaired LTP, premature maturation of dendritic spines, learning disabilities and seizures in mice. More recently, SYNGAP1 was shown to influence cortical neurogenesis and the proliferation of progenitors in human organoids. Here, we show that the absence or haploinsufficiency of Syngap1 does not influence the properties of neocortical progenitors and their cellular output in mice. This discrepancy highlights potential species-specific or methodological differences and raises important questions about the broader applicability of SYNGAP1's role in neurogenesis.
Collapse
Affiliation(s)
- Soraia Barão
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ingie Hong
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ulrich Müller
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Richard L. Huganir
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
11
|
Evans EF, Shyr ZA, Traynor BJ, Zheng W. Therapeutic development approaches to treat haploinsufficiency diseases: restoring protein levels. Drug Discov Today 2024; 29:104201. [PMID: 39384033 DOI: 10.1016/j.drudis.2024.104201] [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: 07/23/2024] [Revised: 09/18/2024] [Accepted: 10/02/2024] [Indexed: 10/11/2024]
Abstract
Rare diseases affect one in ten people but only a small fraction of these diseases have an FDA-approved treatment. Haploinsufficiency, caused by a dominant loss-of-function mutation, is a unique rare disease group because patients have one normal allele of the affected gene. This makes rare haploinsufficiency diseases promising candidates for drug development by increasing expression of the normal gene allele, decreasing the target protein degradation and enhancing the target protein function. This review summarizes recent progresses and approaches used in the translational research of therapeutics to treat haploinsufficiency diseases including gene therapy, nucleotide-based therapeutics and small-molecule drug development. We hope that these drug development strategies will accelerate therapeutic development to treat haploinsufficiency diseases.
Collapse
Affiliation(s)
- Elena F Evans
- National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Bethesda, MD 20892-3375, USA
| | - Zeenat A Shyr
- National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Bethesda, MD 20892-3375, USA
| | - Bryan J Traynor
- National Institute on Aging, National Institutes of Health, 35 Convent Drive, Bethesda, MD 20814, USA
| | - Wei Zheng
- National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Bethesda, MD 20892-3375, USA.
| |
Collapse
|
12
|
Jasińska M, Jasek-Gajda E, Ziaja M, Litwin JA, Lis GJ, Pyza E. Light-Modulated Circadian Synaptic Plasticity in the Somatosensory Cortex: Link to Locomotor Activity. Int J Mol Sci 2024; 25:12870. [PMID: 39684579 DOI: 10.3390/ijms252312870] [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: 10/29/2024] [Revised: 11/25/2024] [Accepted: 11/27/2024] [Indexed: 12/18/2024] Open
Abstract
The circadian clock controls various physiological processes, including synaptic function and neuronal activity, affecting the functioning of the entire organism. Light is an important external factor regulating the day-night cycle. This study examined the effects of the circadian clock and light on synaptic plasticity, and explored how locomotor activity contributes to these processes. We analyzed synaptic protein expression and excitatory synapse density in the somatosensory cortex of mice from four groups exposed to different lighting conditions (LD 12:12, DD, LD 16:8, and LL). Locomotor activity was assessed through individual wheel-running monitoring. To explore daily and circadian changes in synaptic proteins, we performed double-immunofluorescence labeling and laser scanning confocal microscopy imaging, targeting three pairs of presynaptic and postsynaptic proteins (Synaptophysin 1/PSD95, Piccolo/Homer 1, Neurexins/PICK1). Excitatory synapse density was evaluated by co-labeling presynaptic and postsynaptic markers. Our results demonstrated that all the analyzed synaptic proteins exhibited circadian regulation modulated by light. Under constant light conditions, only Piccolo and Homer 1 showed rhythmicity. Locomotor activity was also associated with the circadian clock's effects on synaptic proteins, showing a stronger connection to changes in postsynaptic protein levels. Excitatory synapse density peaked during the day/subjective day and exhibited an inverse relationship with locomotor activity. Continued light exposure disrupted cyclic changes in synapse density but kept it consistently elevated. These findings underscore the crucial roles of light and locomotor activity in regulating synaptic plasticity.
Collapse
Affiliation(s)
- Małgorzata Jasińska
- Department of Histology, Jagiellonian University Medical College, 31-034 Krakow, Poland
| | - Ewa Jasek-Gajda
- Department of Histology, Jagiellonian University Medical College, 31-034 Krakow, Poland
| | - Marek Ziaja
- Department of Histology, Jagiellonian University Medical College, 31-034 Krakow, Poland
| | - Jan A Litwin
- Department of Histology, Jagiellonian University Medical College, 31-034 Krakow, Poland
| | - Grzegorz J Lis
- Department of Histology, Jagiellonian University Medical College, 31-034 Krakow, Poland
| | - Elżbieta Pyza
- Department of Cell Biology and Imaging, Institute of Zoology and Biomedical Research, Jagiellonian University, 30-387 Krakow, Poland
| |
Collapse
|
13
|
Scorrano G, Di Francesco L, Di Ludovico A, Chiarelli F, Matricardi S. Exploring the Landscape of Pre- and Post-Synaptic Pediatric Disorders with Epilepsy: A Narrative Review on Molecular Mechanisms Involved. Int J Mol Sci 2024; 25:11982. [PMID: 39596051 PMCID: PMC11593774 DOI: 10.3390/ijms252211982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 11/03/2024] [Accepted: 11/06/2024] [Indexed: 11/28/2024] Open
Abstract
Neurodevelopmental disorders (NDDs) are a group of conditions affecting brain development, with variable degrees of severity and heterogeneous clinical features. They include intellectual disability (ID), autism spectrum disorder (ASD), attention-deficit/hyperactivity disorder (ADHD), often coexisting with epilepsy, extra-neurological comorbidities, and multisystemic involvement. In recent years, next-generation sequencing (NGS) technologies allowed the identification of several gene pathogenic variants etiologically related to these disorders in a large cohort of affected children. These genes encode proteins involved in synaptic homeostasis, such as SNARE proteins, implicated in calcium-triggered pre-synaptic release of neurotransmitters, or channel subunit proteins, such as post-synaptic ionotropic glutamate receptors involved in the brain's fast excitatory neurotransmission. In this narrative review, we dissected emerged molecular mechanisms related to NDDs and epilepsy due to defects in pre- and post-synaptic transmission. We focused on the most recently discovered SNAREopathies and AMPA-related synaptopathies.
Collapse
Affiliation(s)
- Giovanna Scorrano
- Department of Pediatrics, University of Chieti-Pescara, Sant’Annunziata Hospital, 66100 Chieti, Italy; (G.S.); (A.D.L.); (F.C.)
| | - Ludovica Di Francesco
- Department of Neonatology, University of L’Aquila, San Salvatore Hospital, 67100 L’Aquila, Italy;
| | - Armando Di Ludovico
- Department of Pediatrics, University of Chieti-Pescara, Sant’Annunziata Hospital, 66100 Chieti, Italy; (G.S.); (A.D.L.); (F.C.)
| | - Francesco Chiarelli
- Department of Pediatrics, University of Chieti-Pescara, Sant’Annunziata Hospital, 66100 Chieti, Italy; (G.S.); (A.D.L.); (F.C.)
| | - Sara Matricardi
- Department of Pediatrics, University of Chieti-Pescara, Sant’Annunziata Hospital, 66100 Chieti, Italy; (G.S.); (A.D.L.); (F.C.)
| |
Collapse
|
14
|
Chimura T, Manabe T. Ca2+-PP2B-PSD-95 axis: A novel regulatory mechanism of the phosphorylation state of Serine 295 of PSD-95. PLoS One 2024; 19:e0313441. [PMID: 39509447 PMCID: PMC11542788 DOI: 10.1371/journal.pone.0313441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 10/23/2024] [Indexed: 11/15/2024] Open
Abstract
The phosphorylation state of PSD-95 at Serine 295 (Ser295) is important for the regulation of synaptic plasticity. Although the activation of NMDA receptors (NMDARs), which initiates an intracellular calcium signaling cascade, decreases phosphorylated Ser295 (pS295) of PSD-95, the molecular mechanisms are not fully understood. We found that the calcium-activated protein phosphatase PP2B dephosphorylated pS295 not only in basal conditions but also in NMDAR-activated conditions in cultured neurons. The biochemical assay also revealed the dephosphorylation of pS295 by PP2B, consistently supporting the results obtained using neurons. The newly identified calcium signaling cascade "Ca2+-PP2B-PSD-95 axis" would play an important role in the molecular mechanism for NMDA receptor-dependent plasticity.
Collapse
Affiliation(s)
- Takahiko Chimura
- Department of Basic Medical Sciences, Institute of Medical Science, Division of Neuronal Network, University of Tokyo, Tokyo, Japan
| | - Toshiya Manabe
- Department of Basic Medical Sciences, Institute of Medical Science, Division of Neuronal Network, University of Tokyo, Tokyo, Japan
| |
Collapse
|
15
|
Li D, Sun N, Guo Y, Huang S, Yin C, Xiao Y, Ma W. Investigating the Effects of Perampanel on Autophagy-mediated Regulation of GluA2 and PSD95 in Epilepsy. Mol Neurobiol 2024; 61:9210-9221. [PMID: 38602656 DOI: 10.1007/s12035-024-04136-1] [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: 01/08/2024] [Accepted: 03/20/2024] [Indexed: 04/12/2024]
Abstract
Epilepsy is a chronic neurological disorder characterized by recurrent seizures. Despite various treatment approaches, a significant number of patients continue to experience uncontrolled seizures, leading to refractory epilepsy. The emergence of novel anti-epileptic drugs, such as perampanel (PER), has provided promising options for effective epilepsy treatment. However, the specific mechanisms underlying the therapeutic effects of PER remain unclear. This study aimed to investigate the intrinsic molecular regulatory mechanisms involved in the downregulation of GluA2, a key subunit of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors, following epileptic seizures. Primary mouse hippocampal neurons were cultured and subjected to an epilepsy cell model. The expression levels of GluA2 and autophagy-related proteins were assessed using Western blotting and real-time fluorescent quantitative PCR. Immunofluorescence and immunohistochemistry techniques were employed to investigate the nuclear translocation of CREB-regulated transcriptional coactivator 1 (CRTC1). Additionally, status epilepticus animal models were established to further validate the findings. The epilepsy cell model exhibited a significant decrease in GluA2 expression, accompanied by elevated levels of autophagy-related proteins. Immunofluorescence analysis revealed the nuclear translocation of CRTC1, which correlated with the expression of autophagy-related genes. Treatment with an autophagy inhibitor reversed the decreased expression of GluA2 in the epilepsy cell model. Furthermore, the calcium/calmodulin-dependent protein phosphatase inhibitor FK506 and CaN overexpression affected the dephosphorylation and nuclear translocation of CRTC1, consequently influencing GluA2 expression. Animal model results further supported the involvement of these molecular mechanisms in epilepsy. Our findings suggest that the downregulation of GluA2 following epileptic seizures involves the activation of autophagy and the regulation of CRTC1 nuclear translocation. These intrinsic molecular regulatory mechanisms provide potential targets for developing novel therapeutic strategies to alleviate refractory epilepsy and preserve cognitive functions in patients.
Collapse
Affiliation(s)
- Dan Li
- Department of Pediatrics, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xiwu Road, Xi'an, Shaanxi, China
| | - Na Sun
- Department of Pediatrics, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xiwu Road, Xi'an, Shaanxi, China
| | - Yingying Guo
- Department of Pediatrics, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xiwu Road, Xi'an, Shaanxi, China
| | - Shaoping Huang
- Department of Pediatrics, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xiwu Road, Xi'an, Shaanxi, China
| | - Chunyan Yin
- Department of Pediatrics, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xiwu Road, Xi'an, Shaanxi, China
| | - Yanfeng Xiao
- Department of Pediatrics, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xiwu Road, Xi'an, Shaanxi, China.
| | - Weijun Ma
- Department of Otolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| |
Collapse
|
16
|
Ng B, Tasaki S, Greathouse KM, Walker CK, Zhang A, Covitz S, Cieslak M, Weber AJ, Adamson AB, Andrade JP, Poovey EH, Curtis KA, Muhammad HM, Seidlitz J, Satterthwaite T, Bennett DA, Seyfried NT, Vogel J, Gaiteri C, Herskowitz JH. Integration across biophysical scales identifies molecular and cellular correlates of person-to-person variability in human brain connectivity. Nat Neurosci 2024; 27:2240-2252. [PMID: 39482360 PMCID: PMC11537986 DOI: 10.1038/s41593-024-01788-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 09/16/2024] [Indexed: 11/03/2024]
Abstract
Brain connectivity arises from interactions across biophysical scales, ranging from molecular to cellular to anatomical to network level. To date, there has been little progress toward integrated analysis across these scales. To bridge this gap, from a unique cohort of 98 individuals, we collected antemortem neuroimaging and genetic data, as well as postmortem dendritic spine morphometric, proteomic and gene expression data from the superior frontal and inferior temporal gyri. Through the integration of the molecular and dendritic spine morphology data, we identified hundreds of proteins that explain interindividual differences in functional connectivity and structural covariation. These proteins are enriched for synaptic structures and functions, energy metabolism and RNA processing. By integrating data at the genetic, molecular, subcellular and tissue levels, we link specific biochemical changes at synapses to connectivity between brain regions. These results demonstrate the feasibility of integrating data from vastly different biophysical scales to provide a more comprehensive understanding of brain connectivity.
Collapse
Affiliation(s)
- Bernard Ng
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Shinya Tasaki
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Kelsey M Greathouse
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Courtney K Walker
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ada Zhang
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Sydney Covitz
- Penn/CHOP Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA, USA
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Matt Cieslak
- Penn/CHOP Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA, USA
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Audrey J Weber
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ashley B Adamson
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Julia P Andrade
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Emily H Poovey
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kendall A Curtis
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hamad M Muhammad
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jakob Seidlitz
- Penn/CHOP Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA, USA
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA, USA
- Department of Child and Adolescent Psychiatry and Behavioral Science, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Ted Satterthwaite
- Penn/CHOP Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA, USA
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Nicholas T Seyfried
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Jacob Vogel
- Penn/CHOP Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA, USA
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA, USA
- Department of Clinical Science, Malmö, SciLifeLab, Lund University, Lund, Sweden
| | - Chris Gaiteri
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA.
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY, USA.
| | - Jeremy H Herskowitz
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
17
|
Cuinat S, Bézieau S, Deb W, Mercier S, Vignard V, Isidor B, Küry S, Ebstein F. Understanding neurodevelopmental proteasomopathies as new rare disease entities: A review of current concepts, molecular biomarkers, and perspectives. Genes Dis 2024; 11:101130. [PMID: 39220754 PMCID: PMC11364055 DOI: 10.1016/j.gendis.2023.101130] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 07/30/2023] [Accepted: 08/19/2023] [Indexed: 09/04/2024] Open
Abstract
The recent advances in high throughput sequencing technology have drastically changed the practice of medical diagnosis, allowing for rapid identification of hundreds of genes causing human diseases. This unprecedented progress has made clear that most forms of intellectual disability that affect more than 3% of individuals worldwide are monogenic diseases. Strikingly, a substantial fraction of the mendelian forms of intellectual disability is associated with genes related to the ubiquitin-proteasome system, a highly conserved pathway made up of approximately 1200 genes involved in the regulation of protein homeostasis. Within this group is currently emerging a new class of neurodevelopmental disorders specifically caused by proteasome pathogenic variants which we propose to designate "neurodevelopmental proteasomopathies". Besides cognitive impairment, these diseases are typically associated with a series of syndromic clinical manifestations, among which facial dysmorphism, motor delay, and failure to thrive are the most prominent ones. While recent efforts have been made to uncover the effects exerted by proteasome variants on cell and tissue landscapes, the molecular pathogenesis of neurodevelopmental proteasomopathies remains ill-defined. In this review, we discuss the cellular changes typically induced by genomic alterations in proteasome genes and explore their relevance as biomarkers for the diagnosis, management, and potential treatment of these new rare disease entities.
Collapse
Affiliation(s)
- Silvestre Cuinat
- Nantes Université, CHU Nantes, Service de Génétique Médicale, Nantes F-44000, France
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes F-44000, France
| | - Stéphane Bézieau
- Nantes Université, CHU Nantes, Service de Génétique Médicale, Nantes F-44000, France
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes F-44000, France
| | - Wallid Deb
- Nantes Université, CHU Nantes, Service de Génétique Médicale, Nantes F-44000, France
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes F-44000, France
| | - Sandra Mercier
- Nantes Université, CHU Nantes, Service de Génétique Médicale, Nantes F-44000, France
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes F-44000, France
| | - Virginie Vignard
- Nantes Université, CHU Nantes, Service de Génétique Médicale, Nantes F-44000, France
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes F-44000, France
| | - Bertrand Isidor
- Nantes Université, CHU Nantes, Service de Génétique Médicale, Nantes F-44000, France
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes F-44000, France
| | - Sébastien Küry
- Nantes Université, CHU Nantes, Service de Génétique Médicale, Nantes F-44000, France
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes F-44000, France
| | - Frédéric Ebstein
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes F-44000, France
| |
Collapse
|
18
|
Bhembre N, Paolino A, Das SS, Guntupalli S, Fenlon LR, Anggono V. Learning-induced remodelling of inhibitory synapses in the motor cortex. Open Biol 2024; 14:240109. [PMID: 39532150 PMCID: PMC11557243 DOI: 10.1098/rsob.240109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 08/09/2024] [Accepted: 09/26/2024] [Indexed: 11/16/2024] Open
Abstract
Robust structural and functional plasticity occurs at excitatory synapses in the motor cortex in response to learning. It is well established that local spinogenesis and the subsequent maintenance of newly formed spines are crucial for motor learning. However, despite local synaptic inhibition being essential for shaping excitatory synaptic input, less is known about the structural rearrangement of inhibitory synapses following learning. In this study, we co-expressed the structural marker tdTomato and a mEmerald-tagged intrabody against gephyrin to visualize inhibitory synapses in layer 2/3 cortical neurons of wild-type CD1 mice. We found that a 1-day accelerated rotarod paradigm induced robust motor learning in male and female adult CD1 mice. Histological analyses revealed a significant increase in the surface area of gephyrin puncta in neurons within the motor cortex but not in the somatosensory cortex upon motor learning. Furthermore, this learning-induced reorganization of inhibitory synapses only occurred in dendritic shafts and not in the spines. These data suggest that learning induces experience-dependent remodelling of existing inhibitory synapses to fine-tune intrinsic plasticity and input-specific modulation of excitatory connections in the motor cortex.
Collapse
Affiliation(s)
- Nishita Bhembre
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, University of Queensland, Brisbane, Queensland4072, Australia
| | - Annalisa Paolino
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, University of Queensland, Brisbane, Queensland4072, Australia
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, Queensland4072, Australia
| | - Sooraj S. Das
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, University of Queensland, Brisbane, Queensland4072, Australia
| | - Sumasri Guntupalli
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, University of Queensland, Brisbane, Queensland4072, Australia
| | - Laura R. Fenlon
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, Queensland4072, Australia
| | - Victor Anggono
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, University of Queensland, Brisbane, Queensland4072, Australia
| |
Collapse
|
19
|
Assendorp N, Fossati M, Libé-Philippot B, Christopoulou E, Depp M, Rapone R, Dingli F, Loew D, Vanderhaeghen P, Charrier C. CTNND2 moderates the pace of synaptic maturation and links human evolution to synaptic neoteny. Cell Rep 2024; 43:114797. [PMID: 39352808 DOI: 10.1016/j.celrep.2024.114797] [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: 05/16/2023] [Revised: 05/01/2024] [Accepted: 09/10/2024] [Indexed: 10/04/2024] Open
Abstract
Human-specific genes are potential drivers of brain evolution. Among them, SRGAP2C has contributed to the emergence of features characterizing human cortical synapses, including their extended period of maturation. SRGAP2C inhibits its ancestral copy, the postsynaptic protein SRGAP2A, but the synaptic molecular pathways differentially regulated in humans by SRGAP2 proteins remain largely unknown. Here, we identify CTNND2, a protein implicated in severe intellectual disability (ID) in Cri-du-Chat syndrome, as a major partner of SRGAP2. We demonstrate that CTNND2 slows synaptic maturation and promotes neuronal integrity. During postnatal development, CTNND2 moderates neuronal excitation and excitability. In adults, it supports synapse maintenance. While CTNND2 deficiency is deleterious and results in synaptic loss of SYNGAP1, another major ID-associated protein, the human-specific protein SRGAP2C, enhances CTNND2 synaptic accumulation in human neurons. Our findings suggest that CTNND2 regulation by SRGAP2C contributes to synaptic neoteny in humans and link human-specific and ID genes at the synapse.
Collapse
Affiliation(s)
- Nora Assendorp
- Institut de Biologie de l'ENS (IBENS), Ecole Normale Supérieure, CNRS, INSERM, PSL Research University, 75005 Paris, France
| | - Matteo Fossati
- Institut de Biologie de l'ENS (IBENS), Ecole Normale Supérieure, CNRS, INSERM, PSL Research University, 75005 Paris, France
| | - Baptiste Libé-Philippot
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; Department of Neurosciences, Leuven Brain Institute, KUL, 3000 Leuven, Belgium
| | - Eirini Christopoulou
- Institut de Biologie de l'ENS (IBENS), Ecole Normale Supérieure, CNRS, INSERM, PSL Research University, 75005 Paris, France
| | - Marine Depp
- Institut de Biologie de l'ENS (IBENS), Ecole Normale Supérieure, CNRS, INSERM, PSL Research University, 75005 Paris, France
| | - Roberta Rapone
- Institut de Biologie de l'ENS (IBENS), Ecole Normale Supérieure, CNRS, INSERM, PSL Research University, 75005 Paris, France
| | - Florent Dingli
- Institut Curie, PSL Research University, CurieCore Tech Mass Spectrometry Proteomics, 75005 Paris, France
| | - Damarys Loew
- Institut Curie, PSL Research University, CurieCore Tech Mass Spectrometry Proteomics, 75005 Paris, France
| | - Pierre Vanderhaeghen
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; Department of Neurosciences, Leuven Brain Institute, KUL, 3000 Leuven, Belgium
| | - Cécile Charrier
- Institut de Biologie de l'ENS (IBENS), Ecole Normale Supérieure, CNRS, INSERM, PSL Research University, 75005 Paris, France.
| |
Collapse
|
20
|
Zhang X, Huo H, Fu G, Li C, Lin W, Dai H, Xi X, Zhai L, Yuan Q, Zhao G, Huo J. Long-read and short-read RNA-seq reveal the transcriptional regulation characteristics of PICK1 in Baoshan pig testis. Anim Reprod 2024; 21:e20240047. [PMID: 39371543 PMCID: PMC11452158 DOI: 10.1590/1984-3143-ar2024-0047] [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: 04/16/2024] [Accepted: 07/04/2024] [Indexed: 10/08/2024] Open
Abstract
PICK1 plays a crucial role in mammalian spermatogenesis. Here, we integrated single-molecule long-read and short-read sequencing to comprehensively examine PICK1 expression patterns in adult Baoshan pig (BS) testes. We identified the most important transcript ENSSSCT00000000120 of PICK1, obtaining its full-length coding sequence (CDS) spanning 1254 bp. Gene structure analysis located PICK1 on pig chromosome 5 with 14 exons. Protein structure analysis reflected that PICK1 consisted of 417 amino acids containing two conserved domains, PDZ and BAR_PICK1. Phylogenetic analysis underscored the evolutionary conservation and homology of PICK1 across different mammalian species. Evaluation of protein interaction network, KEGG, and GO pathways implied that interacted with 50 proteins, predominantly involved in glutamatergic synapses, amphetamine addiction, neuroactive ligand-receptor interactions, dopaminergic synapses, and synaptic vesicle recycling, and PICK1 exhibited significant correlation with DLG4 and TBC1D20. Functional annotation identified that PICK1 was involved in 9 GOs, including seven cellular components and two molecular functions. ceRNA network analysis suggested BS PICK1 was regulated by seven miRNA targets. Moreover, qPCR expression analysis across 15 tissues highlighted that PICK1 was highly expressed in the bulbourethral gland and testis. Subcellular localization analysis in ST (Swine Tesits) cells demonstrated that PICK1 significantly localized within the cytoplasm. Overall, our findings shed new light on PICK1's role in BS reproduction, providing a foundation for further functional studies of PICK1.
Collapse
Affiliation(s)
- Xia Zhang
- Department of Biological and Food Engineering, Lyuliang University, Lvliang, Shanxi, China
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming, Yunnan, China
| | - Hailong Huo
- Yunnan Open University, Kunming, Yunnan, China
- Yunnan Vocational and Technical College of Agriculture, Kunming, Yunnan, China
| | - Guowen Fu
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, Yunnan, China
| | - Changyao Li
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming, Yunnan, China
| | - Wan Lin
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming, Yunnan, China
| | - Hongmei Dai
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming, Yunnan, China
| | - Xuemin Xi
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming, Yunnan, China
| | - Lan Zhai
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming, Yunnan, China
| | - Qingting Yuan
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming, Yunnan, China
| | - Guiying Zhao
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming, Yunnan, China
| | - Jinlong Huo
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming, Yunnan, China
| |
Collapse
|
21
|
Wlodarczyk J, Bhattacharyya R, Dore K, Ho GPH, Martin DDO, Mejias R, Hochrainer K. Altered Protein Palmitoylation as Disease Mechanism in Neurodegenerative Disorders. J Neurosci 2024; 44:e1225242024. [PMID: 39358031 PMCID: PMC11450541 DOI: 10.1523/jneurosci.1225-24.2024] [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: 06/20/2024] [Revised: 07/12/2024] [Accepted: 07/16/2024] [Indexed: 10/04/2024] Open
Abstract
Palmitoylation, a lipid-based posttranslational protein modification, plays a crucial role in regulating various aspects of neuronal function through altering protein membrane-targeting, stabilities, and protein-protein interaction profiles. Disruption of palmitoylation has recently garnered attention as disease mechanism in neurodegeneration. Many proteins implicated in neurodegenerative diseases and associated neuronal dysfunction, including but not limited to amyloid precursor protein, β-secretase (BACE1), postsynaptic density protein 95, Fyn, synaptotagmin-11, mutant huntingtin, and mutant superoxide dismutase 1, undergo palmitoylation, and recent evidence suggests that altered palmitoylation contributes to the pathological characteristics of these proteins and associated disruption of cellular processes. In addition, dysfunction of enzymes that catalyze palmitoylation and depalmitoylation has been connected to the development of neurological disorders. This review highlights some of the latest advances in our understanding of palmitoylation regulation in neurodegenerative diseases and explores potential therapeutic implications.
Collapse
Affiliation(s)
- Jakub Wlodarczyk
- Laboratory of Cell Biophysics, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw 02-093, Poland
| | - Raja Bhattacharyya
- Genetics and Aging Research Unit, Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Kim Dore
- Department of Neurosciences, Center for Neural Circuits and Behavior, UCSD, La Jolla, California 92093
| | - Gary P H Ho
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115
| | - Dale D O Martin
- Department of Biology, Faculty of Science, University of Waterloo, Waterloo, Ontario N2L 3G1, Canada
| | - Rebeca Mejias
- Department of Physiology, School of Biology, Universidad de Sevilla, Seville, 41012 Spain
- Instituto de Investigaciones Biomédicas de Sevilla, IBIS/Universidad de Sevilla/Hospital Universitario Virgen del Rocío/Junta de Andalucía/CSIC, Seville 41013, Spain
| | - Karin Hochrainer
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York 10065
| |
Collapse
|
22
|
Verma H, Kaur S, Kaur S, Gangwar P, Dhiman M, Mantha AK. Role of Cytoskeletal Elements in Regulation of Synaptic Functions: Implications Toward Alzheimer's Disease and Phytochemicals-Based Interventions. Mol Neurobiol 2024; 61:8320-8343. [PMID: 38491338 DOI: 10.1007/s12035-024-04053-3] [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: 07/03/2023] [Accepted: 02/13/2024] [Indexed: 03/18/2024]
Abstract
Alzheimer's disease (AD), a multifactorial disease, is characterized by the accumulation of neurofibrillary tangles (NFTs) and amyloid beta (Aβ) plaques. AD is triggered via several factors like alteration in cytoskeletal proteins, a mutation in presenilin 1 (PSEN1), presenilin 2 (PSEN2), amyloid precursor protein (APP), and post-translational modifications (PTMs) in the cytoskeletal elements. Owing to the major structural and functional role of cytoskeletal elements, like the organization of axon initial segmentation, dendritic spines, synaptic regulation, and delivery of cargo at the synapse; modulation of these elements plays an important role in AD pathogenesis; like Tau is a microtubule-associated protein that stabilizes the microtubules, and it also causes inhibition of nucleo-cytoplasmic transportation by disrupting the integrity of nuclear pore complex. One of the major cytoskeletal elements, actin and its dynamics, regulate the dendritic spine structure and functions; impairments have been documented towards learning and memory defects. The second major constituent of these cytoskeletal elements, microtubules, are necessary for the delivery of the cargo, like ion channels and receptors at the synaptic membranes, whereas actin-binding protein, i.e., Cofilin's activation form rod-like structures, is involved in the formation of paired helical filaments (PHFs) observed in AD. Also, the glial cells rely on their cytoskeleton to maintain synaptic functionality. Thus, making cytoskeletal elements and their regulation in synaptic structure and function as an important aspect to be focused for better management and targeting AD pathology. This review advocates exploring phytochemicals and Ayurvedic plant extracts against AD by elucidating their neuroprotective mechanisms involving cytoskeletal modulation and enhancing synaptic plasticity. However, challenges include their limited bioavailability due to the poor solubility and the limited potential to cross the blood-brain barrier (BBB), emphasizing the need for targeted strategies to improve therapeutic efficacy.
Collapse
Affiliation(s)
- Harkomal Verma
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Village Ghudda, VPO - Ghudda, Bathinda, 151 401, Punjab, India
| | - Sharanjot Kaur
- Department of Microbiology, School of Basic Sciences, Central University of Punjab, Village Ghudda, Bathinda, Punjab, India
| | - Sukhchain Kaur
- Department of Microbiology, School of Basic Sciences, Central University of Punjab, Village Ghudda, Bathinda, Punjab, India
| | - Prabhakar Gangwar
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Village Ghudda, VPO - Ghudda, Bathinda, 151 401, Punjab, India
| | - Monisha Dhiman
- Department of Microbiology, School of Basic Sciences, Central University of Punjab, Village Ghudda, Bathinda, Punjab, India
| | - Anil Kumar Mantha
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Village Ghudda, VPO - Ghudda, Bathinda, 151 401, Punjab, India.
| |
Collapse
|
23
|
Carretero-Guillén A, Treviño M, Gómez-Climent MÁ, Dogbevia GK, Bertocchi I, Sprengel R, Larkum ME, Vlachos A, Gruart A, Delgado-García JM, Hasan MT. Dentate gyrus is needed for memory retrieval. Mol Psychiatry 2024; 29:2939-2950. [PMID: 38609585 PMCID: PMC11449802 DOI: 10.1038/s41380-024-02546-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 03/03/2024] [Accepted: 03/26/2024] [Indexed: 04/14/2024]
Abstract
The hippocampus is crucial for acquiring and retrieving episodic and contextual memories. In previous studies, the inactivation of dentate gyrus (DG) neurons by chemogenetic- and optogenetic-mediated hyperpolarization led to opposing conclusions about DG's role in memory retrieval. One study used Designer Receptors Exclusively Activated by Designer Drugs (DREADD)-mediated clozapine N-oxide (CNO)-induced hyperpolarization and reported that the previously formed memory was erased, thus concluding that denate gyrus is needed for memory maintenance. The other study used optogenetic with halorhodopsin induced hyperpolarization and reported and dentate gyrus is needed for memory retrieval. We hypothesized that this apparent discrepancy could be due to the length of hyperpolarization in previous studies; minutes by optogenetics and several hours by DREADD/CNO. Since hyperpolarization interferes with anterograde and retrograde neuronal signaling, it is possible that the memory engram in the dentate gyrus and the entorhinal to hippocampus trisynaptic circuit was erased by long-term, but not with short-term hyperpolarization. We developed and applied an advanced chemogenetic technology to selectively silence synaptic output by blocking neurotransmitter release without hyperpolarizing DG neurons to explore this apparent discrepancy. We performed in vivo electrophysiology during trace eyeblink in a rabbit model of associative learning. Our work shows that the DG output is required for memory retrieval. Based on previous and recent findings, we propose that the actively functional anterograde and retrograde neuronal signaling is necessary to preserve synaptic memory engrams along the entorhinal cortex to the hippocampal trisynaptic circuit.
Collapse
Affiliation(s)
- Alejandro Carretero-Guillén
- Division of Neuroscience, University Pablo de Olavide, Seville, Spain
- Achucarro Basque Center for Neuroscience, Leioa, Spain
| | - Mario Treviño
- Max Planck Institute for Medical Research, Heidelberg, Germany
- Instituto de Neurociencias, Universidad de Guadalajara, Guadalajara, 44130, México
| | | | - Godwin K Dogbevia
- Max Planck Institute for Medical Research, Heidelberg, Germany
- Health Canada, Ottawa, ON, Canada
| | - Ilaria Bertocchi
- Max Planck Institute for Medical Research, Heidelberg, Germany
- Neuroscience Institute Cavalieri-Ottolenghi (NICO), University of Turin, Turin, Italy
| | - Rolf Sprengel
- Max Planck Institute for Medical Research, Heidelberg, Germany
| | | | | | - Agnès Gruart
- Division of Neuroscience, University Pablo de Olavide, Seville, Spain
| | | | - Mazahir T Hasan
- Achucarro Basque Center for Neuroscience, Leioa, Spain.
- Max Planck Institute for Medical Research, Heidelberg, Germany.
- NeuroCure, Charité - Universitätsmedizin, Berlin, Germany.
- Ikerbasque - Basque Foundation for Science, Bilbao, Spain.
| |
Collapse
|
24
|
Guo A, Wu Q, Yan X, Chen K, Liu Y, Liang D, Yang Y, Luo Q, Xiong M, Yu Y, Fei E, Chen F. Differential roles of lysosomal cholesterol transporters in the development of C. elegans NMJs. Life Sci Alliance 2024; 7:e202402584. [PMID: 39084875 PMCID: PMC11291935 DOI: 10.26508/lsa.202402584] [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: 01/10/2024] [Revised: 07/21/2024] [Accepted: 07/22/2024] [Indexed: 08/02/2024] Open
Abstract
Cholesterol homeostasis in neurons is critical for synapse formation and maintenance. Neurons with impaired cholesterol uptake undergo progressive synapse loss and eventual degeneration. To investigate the molecular mechanisms of neuronal cholesterol homeostasis and its role during synapse development, we studied motor neurons of Caenorhabditis elegans because these neurons rely on dietary cholesterol. Combining lipidomic analysis, we discovered that NCR-1, a lysosomal cholesterol transporter, promotes cholesterol absorption and synapse development. Loss of ncr-1 causes smaller synapses, and low cholesterol exacerbates the deficits. Moreover, NCR-1 deficiency hinders the increase in synapses under high cholesterol. Unexpectedly, NCR-2, the NCR-1 homolog, increases the use of cholesterol and sphingomyelins and impedes synapse formation. NCR-2 deficiency causes an increase in synapses regardless of cholesterol concentration. Inhibiting the degradation or synthesis of sphingomyelins can induce or suppress the synaptic phenotypes in ncr-2 mutants. Our findings indicate that neuronal cholesterol homeostasis is differentially controlled by two lysosomal cholesterol transporters and highlight the importance of neuronal cholesterol homeostasis in synapse development.
Collapse
Affiliation(s)
- Amin Guo
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Qi Wu
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Xin Yan
- School of Life Sciences, Nanchang University, Nanchang, China
| | - Kanghua Chen
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yuxiang Liu
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Dingfa Liang
- Queen Mary School of Nanchang University, Jiangxi Medical College, Nanchang, China
| | - Yuxiao Yang
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Qunfeng Luo
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Mingtao Xiong
- Institute of Biomedical Innovation, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yong Yu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Erkang Fei
- Institute of Biomedical Innovation, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Fei Chen
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| |
Collapse
|
25
|
Yan J, Armstrong JPK, Scarpa F, Perriman AW. Hydrogel-Based Artificial Synapses for Sustainable Neuromorphic Electronics. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2403937. [PMID: 39087845 DOI: 10.1002/adma.202403937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 06/16/2024] [Indexed: 08/02/2024]
Abstract
Hydrogels find widespread applications in biomedicine because of their outstanding biocompatibility, biodegradability, and tunable material properties. Hydrogels can be chemically functionalized or reinforced to respond to physical or chemical stimulation, which opens up new possibilities in the emerging field of intelligent bioelectronics. Here, the state-of-the-art in functional hydrogel-based transistors and memristors is reviewed as potential artificial synapses. Within these systems, hydrogels can serve as semisolid dielectric electrolytes in transistors and as switching layers in memristors. These synaptic devices with volatile and non-volatile resistive switching show good adaptability to external stimuli for short-term and long-term synaptic memory effects, some of which are integrated into synaptic arrays as artificial neurons; although, there are discrepancies in switching performance and efficacy. By comparing different hydrogels and their respective properties, an outlook is provided on a new range of biocompatible, environment-friendly, and sustainable neuromorphic hardware. How potential energy-efficient information storage and processing can be achieved using artificial neural networks with brain-inspired architecture for neuromorphic computing is described. The development of hydrogel-based artificial synapses can significantly impact the fields of neuromorphic bionics, biometrics, and biosensing.
Collapse
Affiliation(s)
- Jiongyi Yan
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK
| | - James P K Armstrong
- Department of Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS1 3NY, UK
| | - Fabrizio Scarpa
- Bristol Composites Institute, School of Civil, Aerospace and Design Engineering (CADE), University of Bristol, University Walk, Bristol, BS8 1TR, UK
| | - Adam W Perriman
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK
- Research School of Chemistry, Australian National University, Canberra, Australian Capital Territory, 2601, Australia
- John Curtin School of Medical Research, Australian National University, Canberra, Australian Capital Territory, 2601, Australia
| |
Collapse
|
26
|
Griffin H, Hanson J, Phelan KD, Baldini G. MC4R Localizes at Excitatory Postsynaptic and Peri-Postsynaptic Sites of Hypothalamic Neurons in Primary Culture. Cells 2024; 13:1235. [PMID: 39120267 PMCID: PMC11311852 DOI: 10.3390/cells13151235] [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: 04/28/2024] [Revised: 07/13/2024] [Accepted: 07/18/2024] [Indexed: 08/10/2024] Open
Abstract
The melanocortin-4 receptor (MC4R) is a G protein-coupled receptor (GPCR) that is expressed in several brain locations encompassing the hypothalamus and the brainstem, where the receptor controls several body functions, including metabolism. In a well-defined pathway to decrease appetite, hypothalamic proopiomelanocortin (POMC) neurons localized in the arcuate nucleus (Arc) project to MC4R neurons in the paraventricular nuclei (PVN) to release the natural MC4R agonist α-melanocyte-stimulating hormone (α-MSH). Arc neurons also project excitatory glutamatergic fibers to the MC4R neurons in the PVN for a fast synaptic transmission to regulate a satiety pathway potentiated by α-MSH. By using super-resolution microscopy, we found that in hypothalamic neurons in a primary culture, postsynaptic density protein 95 (PSD95) colocalizes with GluN1, a subunit of the ionotropic N-methyl-D-aspartate receptor (NMDAR). Thus, hypothalamic neurons form excitatory postsynaptic specializations. To study the MC4R distribution at these sites, tagged HA-MC4R under the synapsin promoter was expressed in neurons by adeno-associated virus (AAV) gene transduction. HA-MC4R immunofluorescence peaked at the center and in proximity to the PSD95- and NMDAR-expressing sites. These data provide morphological evidence that MC4R localizes together with glutamate receptors at postsynaptic and peri-postsynaptic sites.
Collapse
Affiliation(s)
- Haven Griffin
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (H.G.); (J.H.)
| | - Jude Hanson
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (H.G.); (J.H.)
| | - Kevin D. Phelan
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| | - Giulia Baldini
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (H.G.); (J.H.)
| |
Collapse
|
27
|
Held RG, Liang J, Brunger AT. Nanoscale architecture of synaptic vesicles and scaffolding complexes revealed by cryo-electron tomography. Proc Natl Acad Sci U S A 2024; 121:e2403136121. [PMID: 38923992 PMCID: PMC11228483 DOI: 10.1073/pnas.2403136121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Accepted: 05/16/2024] [Indexed: 06/28/2024] Open
Abstract
The spatial distribution of proteins and their arrangement within the cellular ultrastructure regulates the opening of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors in response to glutamate release at the synapse. Fluorescence microscopy imaging revealed that the postsynaptic density (PSD) and scaffolding proteins in the presynaptic active zone (AZ) align across the synapse to form a trans-synaptic "nanocolumn," but the relation to synaptic vesicle release sites is uncertain. Here, we employ focused-ion beam (FIB) milling and cryoelectron tomography to image synapses under near-native conditions. Improved image contrast, enabled by FIB milling, allows simultaneous visualization of supramolecular nanoclusters within the AZ and PSD and synaptic vesicles. Surprisingly, membrane-proximal synaptic vesicles, which fuse to release glutamate, are not preferentially aligned with AZ or PSD nanoclusters. These synaptic vesicles are linked to the membrane by peripheral protein densities, often consistent in size and shape with Munc13, as well as globular densities bridging the synaptic vesicle and plasma membrane, consistent with prefusion complexes of SNAREs, synaptotagmins, and complexin. Monte Carlo simulations of synaptic transmission events using biorealistic models guided by our tomograms predict that clustering AMPARs within PSD nanoclusters increases the variability of the postsynaptic response but not its average amplitude. Together, our data support a model in which synaptic strength is tuned at the level of single vesicles by the spatial relationship between scaffolding nanoclusters and single synaptic vesicle fusion sites.
Collapse
Affiliation(s)
- Richard G. Held
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA94305
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA94305
- Department of Structural Biology, Stanford University, Stanford, CA94305
- Department of Photon Science, Stanford University, Stanford, CA94305
- HHMI, Stanford University, Stanford, CA94305
| | - Jiahao Liang
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA94305
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA94305
- Department of Structural Biology, Stanford University, Stanford, CA94305
- Department of Photon Science, Stanford University, Stanford, CA94305
- HHMI, Stanford University, Stanford, CA94305
| | - Axel T. Brunger
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA94305
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA94305
- Department of Structural Biology, Stanford University, Stanford, CA94305
- Department of Photon Science, Stanford University, Stanford, CA94305
- HHMI, Stanford University, Stanford, CA94305
| |
Collapse
|
28
|
Badal KK, Zhao Y, Raveendra BL, Lozano-Villada S, Miller KE, Puthanveettil SV. PKA Activity-Driven Modulation of Bidirectional Long-Distance transport of Lysosomal vesicles During Synapse Maintenance. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.28.601272. [PMID: 38979384 PMCID: PMC11230415 DOI: 10.1101/2024.06.28.601272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
The bidirectional long-distance transport of organelles is crucial for cell body-synapse communication. However, the mechanisms by which this transport is modulated for synapse formation, maintenance, and plasticity are not fully understood. Here, we demonstrate through quantitative analyses that maintaining sensory neuron-motor neuron synapses in the Aplysia gill-siphon withdrawal reflex is linked to a sustained reduction in the retrograde transport of lysosomal vesicles in sensory neurons. Interestingly, while mitochondrial transport in the anterograde direction increases within 12 hours of synapse formation, the reduction in lysosomal vesicle retrograde transport appears three days after synapse formation. Moreover, we find that formation of new synapses during learning induced by neuromodulatory neurotransmitter serotonin further reduces lysosomal vesicle transport within 24 hours, whereas mitochondrial transport increases in the anterograde direction within one hour of exposure. Pharmacological inhibition of several signaling pathways pinpoints PKA as a key regulator of retrograde transport of lysosomal vesicles during synapse maintenance. These results demonstrate that synapse formation leads to organelle-specific and direction specific enduring changes in long-distance transport, offering insights into the mechanisms underlying synapse maintenance and plasticity.
Collapse
Affiliation(s)
- Kerriann. K. Badal
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, 130 Scripps Way, Jupiter, FL 33458, USA
- Integrative Biology PhD Program, Charles E. Schmidt College of Science, Florida Atlantic University, Jupiter, FL 33458, USA
| | - Yibo. Zhao
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, 130 Scripps Way, Jupiter, FL 33458, USA
| | - Bindu L Raveendra
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, 130 Scripps Way, Jupiter, FL 33458, USA
| | - Sebastian Lozano-Villada
- Harriet L. Wilkes Honors College, Florida Atlantic University, 5353 Parkside Drive, Jupiter, FL 33458, USA
| | - Kyle. E. Miller
- Harriet L. Wilkes Honors College, Florida Atlantic University, 5353 Parkside Drive, Jupiter, FL 33458, USA
| | - Sathyanarayanan V. Puthanveettil
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, 130 Scripps Way, Jupiter, FL 33458, USA
| |
Collapse
|
29
|
Eckmann S, Young EJ, Gjorgjieva J. Synapse-type-specific competitive Hebbian learning forms functional recurrent networks. Proc Natl Acad Sci U S A 2024; 121:e2305326121. [PMID: 38870059 PMCID: PMC11194505 DOI: 10.1073/pnas.2305326121] [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: 04/04/2023] [Accepted: 04/25/2024] [Indexed: 06/15/2024] Open
Abstract
Cortical networks exhibit complex stimulus-response patterns that are based on specific recurrent interactions between neurons. For example, the balance between excitatory and inhibitory currents has been identified as a central component of cortical computations. However, it remains unclear how the required synaptic connectivity can emerge in developing circuits where synapses between excitatory and inhibitory neurons are simultaneously plastic. Using theory and modeling, we propose that a wide range of cortical response properties can arise from a single plasticity paradigm that acts simultaneously at all excitatory and inhibitory connections-Hebbian learning that is stabilized by the synapse-type-specific competition for a limited supply of synaptic resources. In plastic recurrent circuits, this competition enables the formation and decorrelation of inhibition-balanced receptive fields. Networks develop an assembly structure with stronger synaptic connections between similarly tuned excitatory and inhibitory neurons and exhibit response normalization and orientation-specific center-surround suppression, reflecting the stimulus statistics during training. These results demonstrate how neurons can self-organize into functional networks and suggest an essential role for synapse-type-specific competitive learning in the development of cortical circuits.
Collapse
Affiliation(s)
- Samuel Eckmann
- Computation in Neural Circuits Group, Max Planck Institute for Brain Research, Frankfurt am Main60438, Germany
- Computational and Biological Learning Lab, Department of Engineering, University of Cambridge, CambridgeCB2 1PZ, United Kingdom
| | - Edward James Young
- Computational and Biological Learning Lab, Department of Engineering, University of Cambridge, CambridgeCB2 1PZ, United Kingdom
| | - Julijana Gjorgjieva
- Computation in Neural Circuits Group, Max Planck Institute for Brain Research, Frankfurt am Main60438, Germany
- School of Life Sciences, Technical University Munich, Freising85354, Germany
| |
Collapse
|
30
|
Matúš D, Lopez JM, Sando RC, Südhof TC. Essential Role of Latrophilin-1 Adhesion GPCR Nanoclusters in Inhibitory Synapses. J Neurosci 2024; 44:e1978232024. [PMID: 38684366 PMCID: PMC11154861 DOI: 10.1523/jneurosci.1978-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 02/03/2024] [Accepted: 02/20/2024] [Indexed: 05/02/2024] Open
Abstract
Latrophilin-1 (Lphn1, aka CIRL1 and CL1; gene symbol Adgrl1) is an adhesion GPCR that has been implicated in excitatory synaptic transmission as a candidate receptor for α-latrotoxin. Here we analyzed conditional knock-in/knock-out mice for Lphn1 that contain an extracellular myc epitope tag. Mice of both sexes were used in all experiments. Surprisingly, we found that Lphn1 is localized in cultured neurons to synaptic nanoclusters that are present in both excitatory and inhibitory synapses. Conditional deletion of Lphn1 in cultured neurons failed to elicit a detectable impairment in excitatory synapses but produced a decrease in inhibitory synapse numbers and synaptic transmission that was most pronounced for synapses close to the neuronal soma. No changes in axonal or dendritic outgrowth or branching were observed. Our data indicate that Lphn1 is among the few postsynaptic adhesion molecules that are present in both excitatory and inhibitory synapses and that Lphn1 by itself is not essential for excitatory synaptic transmission but is required for some inhibitory synaptic connections.
Collapse
Affiliation(s)
- Daniel Matúš
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, California 94305
| | - Jaybree M Lopez
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee 37240
| | - Richard C Sando
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, California 94305
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee 37240
| | - Thomas C Südhof
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, California 94305
- Hughes Medical Institute, Stanford University School of Medicine, Stanford, California 94305
| |
Collapse
|
31
|
Fatemi SH, Eschenlauer A, Aman J, Folsom TD, Chekouo T. Quantitative proteomics of dorsolateral prefrontal cortex reveals an early pattern of synaptic dysmaturation in children with idiopathic autism. Cereb Cortex 2024; 34:161-171. [PMID: 38696595 PMCID: PMC11484494 DOI: 10.1093/cercor/bhae044] [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: 12/26/2023] [Accepted: 01/23/2024] [Indexed: 05/04/2024] Open
Abstract
Autism spectrum disorder (ASD) is a developmental disorder with a rising prevalence and unknown etiology presenting with deficits in cognition and abnormal behavior. We hypothesized that the investigation of the synaptic component of prefrontal cortex may provide proteomic signatures that may identify the biological underpinnings of cognitive deficits in childhood ASD. Subcellular fractions of synaptosomes from prefrontal cortices of age-, brain area-, and postmortem-interval-matched samples from children and adults with idiopathic ASD vs. controls were subjected to HPLC-tandem mass spectrometry. Analysis of data revealed the enrichment of ASD risk genes that participate in slow maturation of the postsynaptic density (PSD) structure and function during early brain development. Proteomic analysis revealed down regulation of PSD-related proteins including AMPA and NMDA receptors, GRM3, DLG4, olfactomedins, Shank1-3, Homer1, CaMK2α, NRXN1, NLGN2, Drebrin1, ARHGAP32, and Dock9 in children with autism (FDR-adjusted P < 0.05). In contrast, PSD-related alterations were less severe or unchanged in adult individuals with ASD. Network analyses revealed glutamate receptor abnormalities. Overall, the proteomic data support the concept that idiopathic autism is a synaptopathy involving PSD-related ASD risk genes. Interruption in evolutionarily conserved slow maturation of the PSD complex in prefrontal cortex may lead to the development of ASD in a susceptible individual.
Collapse
Affiliation(s)
- S Hossein Fatemi
- Department of Psychiatry and Behavioral Sciences, University of Minnesota Medical School, 420 Delaware Street SE, Minneapolis, MN 55455, USA
| | - Arthur Eschenlauer
- Minnesota Supercomputing Institute, 599 Walter Library, 117 Pleasant Street, Minneapolis, MN 55455, USA
| | - Justin Aman
- Department of Psychiatry and Behavioral Sciences, University of Minnesota Medical School, 420 Delaware Street SE, Minneapolis, MN 55455, USA
| | - Timothy D Folsom
- Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota Medical School, Minneapolis, MN 55455, USA
- Center for Genome Engineering, University of Minnesota Medical School, Minneapolis, MN 55455, USA
- Stem Cell Institute, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Thierry Chekouo
- University of Minnesota School of Public Health, Minneapolis, MN 55455, USA
| |
Collapse
|
32
|
González-Cota AL, Martínez-Flores D, Rosendo-Pineda MJ, Vaca L. NMDA receptor-mediated Ca 2+ signaling: Impact on cell cycle regulation and the development of neurodegenerative diseases and cancer. Cell Calcium 2024; 119:102856. [PMID: 38408411 DOI: 10.1016/j.ceca.2024.102856] [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: 09/11/2023] [Revised: 01/08/2024] [Accepted: 02/07/2024] [Indexed: 02/28/2024]
Abstract
NMDA receptors are Ca2+-permeable ligand-gated ion channels that mediate fast excitatory transmission in the central nervous system. NMDA receptors regulate the proliferation and differentiation of neural progenitor cells and also play critical roles in neural plasticity, memory, and learning. In addition to their physiological role, NMDA receptors are also involved in glutamate-mediated excitotoxicity, which results from excessive glutamate stimulation, leading to Ca2+ overload, and ultimately to neuronal death. Thus, NMDA receptor-mediated excitotoxicity has been linked to several neurodegenerative diseases such as Alzheimer's, Parkinson's, Huntington's, dementia, and stroke. Interestingly, in addition to its effects on cell death, aberrant expression or activation of NMDA receptors is also involved in pathological cellular proliferation, and is implicated in the invasion and proliferation of various types of cancer. These disorders are thought to be related to the contribution of NMDA receptors to cell proliferation and cell death through cell cycle modulation. This review aims to discuss the evidence implicating NMDA receptor activity in cell cycle regulation and the link between aberrant NMDA receptor activity and the development of neurodegenerative diseases and cancer due to cell cycle dysregulation. The information presented here will provide insights into the signaling pathways and the contribution of NMDA receptors to these diseases, and suggests that NMDA receptors are promising targets for the prevention and treatment of these diseases, which are leading causes of death and disability worldwide.
Collapse
Affiliation(s)
- Ana L González-Cota
- Instituto de Fisiología Celular, Departamento de Biología Celular y Desarrollo, Universidad Nacional Autónoma de México, Ciudad Universitaria, Coyoacán, Ciudad de México, 04510, Mexico
| | - Daniel Martínez-Flores
- Instituto de Fisiología Celular, Departamento de Biología Celular y Desarrollo, Universidad Nacional Autónoma de México, Ciudad Universitaria, Coyoacán, Ciudad de México, 04510, Mexico
| | - Margarita Jacaranda Rosendo-Pineda
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad Universitaria, Coyoacán, Ciudad de México, 04510, Mexico
| | - Luis Vaca
- Instituto de Fisiología Celular, Departamento de Biología Celular y Desarrollo, Universidad Nacional Autónoma de México, Ciudad Universitaria, Coyoacán, Ciudad de México, 04510, Mexico.
| |
Collapse
|
33
|
Mansouri Z, Khodagholi F, Zaringhalam J, Abbaszadeh F, Ghasemi R, Maghsoudi N. Intranasal CEPO-FC prevents attention deficits in streptozotocin-induced rat model of Alzheimer's disease: Focus on synaptic plasticity-related factors. EXCLI JOURNAL 2024; 23:491-508. [PMID: 38741725 PMCID: PMC11089095 DOI: 10.17179/excli2023-6818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 04/17/2024] [Indexed: 05/16/2024]
Abstract
Alzheimer's disease remains an issue of great controversy due to its pathology. It is characterized by cognitive impairments and neuropsychiatric symptoms. The FDA approved medications for this disease, can only mitigate the symptoms. One reason for the lack of effective medications is the inaccessibility of the brain which is encompassed by the blood-brain barrier, making intranasal (IN) route of administration potentially advantageous. Male Wistar rats underwent stereotaxic surgery to induce an Alzheimer's disease model via intracerebroventricular (ICV) streptozotocin injection, and Carbamylated Erythropoietin-Fc (CEPO-FC), a derivative of Erythropoietin without its harmful characteristics, was administered intranasally for ten consecutive days. Cognition performance for memory and attention was assessed using the Novel Object Recognition Test and the Object-Based Attention Test respectively. Depression like behavior was evaluated using the Forced Swim Test. Western blotting was done on the extracted hippocampus to quantify STIM proteins. Calbindin, PSD-95, Neuroplastin, Synaptophysin and GAP-43 genes were assessed by Realtime PCR. Behavioral tests demonstrated that IN CEPO-FC could halt cognition deficits and molecular investigations showed that, STIM proteins were decreased in Alzheimer's model, and increased after IN CEPO-FC treatment. Calbindin and PSD-95 were downregulated in our disease model and upregulated when treated with IN CEPO-FC. While Neuroplastin, and GAP-43 expressions remained unchanged. This study suggests that IN CEPO-FC in low doses could be promising for improving cognition and synaptic plasticity deficits in Alzheimer's disease and since IN route of administration is a convenient way, choosing IN CEPO-FC for clinical trial might worth consideration. See also the graphical abstract(Fig. 1).
Collapse
Affiliation(s)
- Zahra Mansouri
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fariba Khodagholi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Jalal Zaringhalam
- Neurophysiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Physiology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Abbaszadeh
- Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Rasoul Ghasemi
- Neurophysiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Physiology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nader Maghsoudi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
34
|
Janelt K, Poprawa I. Organisation of the nervous system in cysts of the freshwater tardigrade Thulinius ruffoi (Parachela, Isohypsibioidea: Doryphoribiidae). J Anat 2024; 244:654-666. [PMID: 38131103 PMCID: PMC10941535 DOI: 10.1111/joa.13994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 11/26/2023] [Accepted: 11/27/2023] [Indexed: 12/23/2023] Open
Abstract
Encystment is a natural process that involves cyst formation, and at least some species of tardigrades can form cysts. However, the encystment process and cyst structure among tardigrades are still poorly understood. Despite some aspects of the encysted animals' systems organisation being examined in the past, the morphology and structure of the nervous system have never been thoroughly investigated. This study covers anatomical, histological and morphological details and proposes physiological aspects of the nervous system in encysted Thulinius ruffoi up to 11 months duration in encystment. This is the first record of the nervous system organisation in a species belonging to the family Doryphoribiidae. The cyst formation results in morphological changes in the nervous system. It comprises central and peripheral elements, which may be observable even after many months since the cyst formation. Based on the nervous system's organisation in cysts, there is no sign that histolysis is a part of encystment.
Collapse
Affiliation(s)
- Kamil Janelt
- Institute of Biology, Biotechnology and Environmental Protection, Faculty of Natural Sciences, University of Silesia in Katowice, Katowice, Poland
- Department of Medical Genetics, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Izabela Poprawa
- Institute of Biology, Biotechnology and Environmental Protection, Faculty of Natural Sciences, University of Silesia in Katowice, Katowice, Poland
| |
Collapse
|
35
|
Kassabian B, Levy AM, Gardella E, Aledo-Serrano A, Ananth AL, Brea-Fernández AJ, Caumes R, Chatron N, Dainelli A, De Wachter M, Denommé-Pichon AS, Dye TJ, Fazzi E, Felt R, Fernández-Jaén A, Fernández-Prieto M, Gantz E, Gasperowicz P, Gil-Nagel A, Gómez-Andrés D, Greiner HM, Guerrini R, Haanpää MK, Helin M, Hoyer J, Hurst ACE, Kallish S, Karkare SN, Khan A, Kleinendorst L, Koch J, Kothare SV, Koudijs SM, Lagae L, Lakeman P, Leppig KA, Lesca G, Lopergolo D, Lusk L, Mackenzie A, Mei D, Møller RS, Pereira EM, Platzer K, Quelin C, Revah-Politi A, Rheims S, Rodríguez-Palmero A, Rossi A, Santorelli F, Seinfeld S, Sell E, Stephenson D, Szczaluba K, Trinka E, Umair M, Van Esch H, van Haelst MM, Veenma DCM, Weber S, Weckhuysen S, Zacher P, Tümer Z, Rubboli G. Developmental epileptic encephalopathy in DLG4-related synaptopathy. Epilepsia 2024; 65:1029-1045. [PMID: 38135915 DOI: 10.1111/epi.17876] [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: 10/13/2023] [Revised: 12/05/2023] [Accepted: 12/20/2023] [Indexed: 12/24/2023]
Abstract
OBJECTIVE The postsynaptic density protein of excitatory neurons PSD-95 is encoded by discs large MAGUK scaffold protein 4 (DLG4), de novo pathogenic variants of which lead to DLG4-related synaptopathy. The major clinical features are developmental delay, intellectual disability (ID), hypotonia, sleep disturbances, movement disorders, and epilepsy. Even though epilepsy is present in 50% of the individuals, it has not been investigated in detail. We describe here the phenotypic spectrum of epilepsy and associated comorbidities in patients with DLG4-related synaptopathy. METHODS We included 35 individuals with a DLG4 variant and epilepsy as part of a multicenter study. The DLG4 variants were detected by the referring laboratories. The degree of ID, hypotonia, developmental delay, and motor disturbances were evaluated by the referring clinician. Data on awake and sleep electroencephalography (EEG) and/or video-polygraphy and brain magnetic resonance imaging were collected. Antiseizure medication response was retrospectively assessed by the referring clinician. RESULTS A large variety of seizure types was reported, although focal seizures were the most common. Encephalopathy related to status epilepticus during slow-wave sleep (ESES)/developmental epileptic encephalopathy with spike-wave activation during sleep (DEE-SWAS) was diagnosed in >25% of the individuals. All but one individual presented with neurodevelopmental delay. Regression in verbal and/or motor domains was observed in all individuals who suffered from ESES/DEE-SWAS, as well as some who did not. We could not identify a clear genotype-phenotype relationship even between individuals with the same DLG4 variants. SIGNIFICANCE Our study shows that a subgroup of individuals with DLG4-related synaptopathy have DEE, and approximately one fourth of them have ESES/DEE-SWAS. Our study confirms DEE as part of the DLG4-related phenotypic spectrum. Occurrence of ESES/DEE-SWAS in DLG4-related synaptopathy requires proper investigation with sleep EEG.
Collapse
Affiliation(s)
- Benedetta Kassabian
- Department of Epilepsy Genetics and Precision Medicine, Danish Epilepsy Center Filadelfia, member of the European Reference Network EpiCARE, Dianalund, Denmark
- Neurology Unit, Department of Neurosciences, University of Padua, Padua, Italy
| | - Amanda M Levy
- Department of Clinical Genetics, Kennedy Center, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Elena Gardella
- Department of Epilepsy Genetics and Precision Medicine, Danish Epilepsy Center Filadelfia, member of the European Reference Network EpiCARE, Dianalund, Denmark
- Department of Regional Health Research, University of Southern Denmark, Odense, Denmark
| | - Angel Aledo-Serrano
- Epilepsy and Neurogenetics Unit, Vithas la Milagrosa University Hospital, Vithas Hospital Group, Madrid, Spain
| | - Amitha L Ananth
- Division of Pediatric Neurology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Alejandro J Brea-Fernández
- Grupo de Genómica y Bioinformática, Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CiMUS), Centro de Investigación Biomédica en Red de Enfermedades Raras del Instituto de Salud Carlos III (CIBERER-ISCIII), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
- Grupo de Genética, Fundación Pública Galega de Medicina Xenómica, Instituto de Investigación Biomédica de Santiago (IDIS), Santiago de Compostela, Spain
| | | | - Nicolas Chatron
- Service de Genetique, Hospices Civils de Lyon, Bron, France
- Institute NeuroMyoGène, Laboratoire Physiopathologie et Génétique du Neurone et du Muscle, Centre National de la recherche scientifique (CNRS) Unité mixte de recherche (UMR) 5261- L'Institut national de la santé et de la recherche médicale (INSERM) U1315, Université de Lyon-Université Claude Bernard Lyon 1, Lyon, France
| | - Alice Dainelli
- Neuroscience Department, Meyer Children's Hospital IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico), member of the European Reference Network EpiCARE, Florence, Italy
| | - Matthias De Wachter
- Department of Pediatric Neurology, Antwerp University Hospital, University of Antwerp, Edegem, Belgium
| | - Anne-Sophie Denommé-Pichon
- Functional Unit for Diagnostic Innovation in Rare Diseases, Fédération Hospitalo-Universitaire Médecine TRANSLationnelle et Anomalies du Développement (FHU-TRANSLAD), Dijon Bourgogne University Hospital, Dijon, France
- L'Institut national de la santé et de la recherche médicale (INSERM) Unité mixte de recherche (UMR) 1231, Génétique des Anomalies du Développement (GAD), Fédération Hospitalo-Universitaire Médecine TRANSLationnelle et Anomalies du Développement (FHU-TRANSLAD), University of Burgundy, Dijon, France
| | - Thomas J Dye
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Elisa Fazzi
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Unit of Child Neurology and Psychiatry, Azienda Socio Sanitaria Territoriale (ASST) Spedali Civili of Brescia, Brescia, Italy
| | - Roxanne Felt
- Department of Neurology, Kaiser Permanente Bellevue Medical Center, Bellevue, Washington, USA
| | - Alberto Fernández-Jaén
- Department of Pediatric Neurology, Neurogenetics Section, Hospital Universitario Quirónsalud, Madrid, Spain
- Facultad de Medicina, Universidad Europea, Madrid, Spain
| | - Montse Fernández-Prieto
- Grupo de Genómica y Bioinformática, Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CiMUS), Centro de Investigación Biomédica en Red de Enfermedades Raras del Instituto de Salud Carlos III (CIBERER-ISCIII), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
- Grupo de Genética, Fundación Pública Galega de Medicina Xenómica, Instituto de Investigación Biomédica de Santiago (IDIS), Santiago de Compostela, Spain
| | - Emily Gantz
- Division of Pediatric Neurology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Piotr Gasperowicz
- Department of Medical Genetics, Medical University of Warsaw, Warsaw, Poland
| | - Antonio Gil-Nagel
- Neurology Department, Epilepsy Program, Ruber Internacional Hospital, Madrid, Spain
| | - David Gómez-Andrés
- Child Neurology Unit, Hospital Universitari Vall d'Hebron, Vall d'Hebron Research Institute, Barcelona, Spain
| | - Hansel M Greiner
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Renzo Guerrini
- Neuroscience Department, Meyer Children's Hospital IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico), member of the European Reference Network EpiCARE, Florence, Italy
| | - Maria K Haanpää
- Department of Genomics, Turku University Hospital, Turku, Finland
| | - Minttu Helin
- Department of Pediatric Neurology, Turku University Hospital, Turku, Finland
| | - Juliane Hoyer
- Friedrich-Alexander-Universität Erlangen Nürnberg, Institute of Human Genetics, Erlangen, Germany
| | - Anna C E Hurst
- Department of Genetics, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Staci Kallish
- Division of Translational Medicine and Human Genetics, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Shefali N Karkare
- Division of Pediatric Neurology, Department of Pediatrics, Cohen Children's Medical Center, New Hyde Park, New York, USA
| | - Amjad Khan
- Department of Zoology, Faculty of Biological Sciences, University of Lakki Marwat, Lakki Marwat, Pakistan
- Institute for Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Lotte Kleinendorst
- Department of Human Genetics, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, the Netherlands
- Emma Center for Personalized Medicine, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, the Netherlands
| | - Johannes Koch
- University Children's Hospital, Paracelsus Medical University, Salzburg, Austria
| | - Sanjeev V Kothare
- Division of Pediatric Neurology, Department of Pediatrics, Cohen Children's Medical Center, New Hyde Park, New York, USA
| | - Suzanna M Koudijs
- Department of Neurology, Erasmus Medical Center (MC) Sophia Children's Hospital, Rotterdam, the Netherlands
- Erfelijke Neuro-Cognitieve Ontwikkelingsstoornissen, Rotterdam, Erasmus Medical Center (ENCORE)-GRIN Expertise Center, Rotterdam, the Netherlands
| | - Lieven Lagae
- Department of Development and Regeneration, Section Paediatric Neurology, member of the European Reference Network EpiCARE, University Hospitals Leuven, Leuven, Belgium
| | - Phillis Lakeman
- Department of Human Genetics, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, the Netherlands
| | - Kathleen A Leppig
- Genetic Services, Kaiser Permanente of Washington, Seattle, Washington, USA
| | - Gaetan Lesca
- Service de Genetique, Hospices Civils de Lyon, Bron, France
- Institute NeuroMyoGène, Laboratoire Physiopathologie et Génétique du Neurone et du Muscle, Centre National de la recherche scientifique (CNRS) Unité mixte de recherche (UMR) 5261- L'Institut national de la santé et de la recherche médicale (INSERM) U1315, Université de Lyon-Université Claude Bernard Lyon 1, Lyon, France
| | - Diego Lopergolo
- Department of Medicine, Surgery, and Neurosciences, University of Siena, Siena, Italy
- Molecular Medicine for Neurodegenerative and Neuromuscular Diseases Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Stella Maris Foundation, Pisa, Italy
| | - Laina Lusk
- Division of Neurology, Epilepsy Neurogenetics Initiative, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Alex Mackenzie
- Research Institute, Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada
- Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Davide Mei
- Neuroscience Department, Meyer Children's Hospital IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico), member of the European Reference Network EpiCARE, Florence, Italy
| | - Rikke S Møller
- Department of Epilepsy Genetics and Precision Medicine, Danish Epilepsy Center Filadelfia, member of the European Reference Network EpiCARE, Dianalund, Denmark
- Department of Regional Health Research, University of Southern Denmark, Odense, Denmark
| | - Elaine M Pereira
- Division of Clinical Genetics, Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons and New York-Presbyterian Morgan Stanley Children's Hospital, New York, New York, USA
| | - Konrad Platzer
- Institute of Human Genetics, University of Leipzig Medical Center, Leipzig, Germany
| | - Chloe Quelin
- Department of Medical Genetics, CHU de Rennes, Rennes, France
| | - Anya Revah-Politi
- Institute for Genomic Medicine, Columbia University Irving Medical Center, New York, New York, USA
| | - Sylvain Rheims
- Department of Functional Neurology and Epileptology, member of the European Reference Network EpiCARE, Hospices Civils de Lyon and Lyon 1 University, Lyon, France
| | - Agustí Rodríguez-Palmero
- Paediatric Neurology Unit, Department of Pediatrics, Hospital Universitari Germans Trias i Pujol, Universitat Autònoma de Barcelona, Barcelona, Spain
- Grupo de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain
| | - Andrea Rossi
- Unit of Child Neurology and Psychiatry, Azienda Socio Sanitaria Territoriale (ASST) Spedali Civili of Brescia, Brescia, Italy
| | - Filippo Santorelli
- Molecular Medicine for Neurodegenerative and Neuromuscular Diseases Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Stella Maris Foundation, Pisa, Italy
| | - Syndi Seinfeld
- Department of Pediatric Neurology, Neuroscience Center, Joe DiMaggio Children's Hospital, Hollywood, Florida, USA
| | - Erick Sell
- Division of Neurology, Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada
| | - Donna Stephenson
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Krzysztof Szczaluba
- Department of Medical Genetics, Medical University of Warsaw, Warsaw, Poland
- Center of Excellence for Rare and Undiagnosed Diseases, Medical University of Warsaw, Warsaw, Poland
| | - Eugen Trinka
- Department of Neurology, Neurointensive Care and Neurorehabilitation, Christian Doppler University Hospital, member of the European Reference Network EpiCARE, Paracelsus Medical University, Center for Cognitive Neuroscience, Salzburg, Austria
- Neuroscience Institute, Christian Doppler University Hospital, member of the European Reference Network EpiCARE, Paracelsus Medical University, Center for Cognitive Neuroscience, Salzburg, Austria
| | - Muhammad Umair
- Medical Genomics Research Department, King Abdullah International Medical Research Center, King Saud Bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
- Department of Life Sciences, School of Science, University of Management and Technology, Lahore, Pakistan
| | - Hilde Van Esch
- Center for Human Genetics, University Hospitals Leuven, Leuven, Belgium
| | - Mieke M van Haelst
- Department of Human Genetics, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, the Netherlands
- Emma Center for Personalized Medicine, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, the Netherlands
| | - Danielle C M Veenma
- Erfelijke Neuro-Cognitieve Ontwikkelingsstoornissen, Rotterdam, Erasmus Medical Center (ENCORE)-GRIN Expertise Center, Rotterdam, the Netherlands
- Department of Pediatrics, Erasmus Medical Center (MC)-Sophia Hospital, Rotterdam, the Netherlands
| | - Sacha Weber
- Service de Génétique, Centre Hospitalier Universitaire (CHU) de Caen-Normandie, Caen, France
- Service de Neurologie, Centre Hospitalier Universitaire (CHU) de Caen-Normandie, Caen, France
| | - Sarah Weckhuysen
- Applied and Translational Neurogenomics Group, Vlaams Instituut voor Biotechnologie (VIB) Center for Molecular Neurology, Antwerp, Belgium
- Department of Neurology, Antwerp University Hospital, Antwerp, Belgium
| | - Pia Zacher
- Center for Adults with Disability (MZEB), Epilepsy Center Kleinwachau, Radeberg, Germany
| | - Zeynep Tümer
- Department of Clinical Genetics, Kennedy Center, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Guido Rubboli
- Department of Epilepsy Genetics and Precision Medicine, Danish Epilepsy Center Filadelfia, member of the European Reference Network EpiCARE, Dianalund, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
36
|
Zhao J, Luo J, Deng C, Fan Y, Liu N, Cao J, Chen D, Diao Y. Volatile oil of Angelica sinensis Radix improves cognitive function by inhibiting miR-301a-3p targeting Ppp2ca in cerebral ischemia mice. JOURNAL OF ETHNOPHARMACOLOGY 2024; 322:117621. [PMID: 38154524 DOI: 10.1016/j.jep.2023.117621] [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: 11/08/2023] [Revised: 12/11/2023] [Accepted: 12/18/2023] [Indexed: 12/30/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Angelica Sinensis Radix (ASR) is a commonly used Chinese medicine known for its effects on tonifying blood, promoting blood circulation, and alleviating pain associated with menstrual regulation. Additionally, it has been used in the treatment of vascular cognitive impairment (VCI). The primary pharmacodynamic agent within ASR is volatile oil of Angelica Sinensis Radix (VOASR), which has demonstrated efficacy in combating cognitive impairment, although its mechanism remains unclear. OBJECTIVE This study aimed to elucidate the potential molecular mechanisms underlying VOASR's improvement of cognitive function in cerebral ischemic mice. METHODS A model of cerebral ischemic mice was established through unilateral common carotid artery occlusion (UCCAO) surgery, followed by intervention with VOASR. Cognitive function was assessed using the Morris water maze (MWM) test, while RT-qPCR was utilized to measure the differential expression of miR-301a-3p in the hippocampus. To evaluate cognitive function and hippocampal protein differences, wild-type mice and miR-301a-3p knockout mice were subjected to the MWM test and iTRAQ protein profiling. The relationship between miR-301a-3p and potential target genes was validated through a Dual-Luciferase Reporter experiment. RT-qPCR and Western blot were employed to determine the differential expression of Ppp2ca and synaptic plasticity-related proteins in the mouse hippocampus. RESULTS Intervention with VOASR significantly improved cognitive impairment in cerebral ischemic mice and reduced the expression of miR-301a-3p in the hippocampus. Our findings suggest that miR-301a-3p may regulate cognitive function by targeting Ppp2ca. Furthermore, VOASR intervention led to an increase in the expression of Ppp2ca and synaptic plasticity-related proteins. CONCLUSION Our study indicates that VOASR may be involved in regulating cognitive function by inhibiting miR-301a-3p, consequently increasing the expression of Ppp2ca and synaptic plasticity proteins. These results provide a new target and direction for the treatment of cognitive dysfunction.
Collapse
Affiliation(s)
- Jie Zhao
- Research Centre of Basic Intergrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| | - Jing Luo
- Shenzhen Hospital of Integrated Traditional and Western Medicine, ShenZhen, 518000, China.
| | - Cuili Deng
- Research Centre of Basic Intergrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| | - Yueying Fan
- Research Centre of Basic Intergrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| | - Na Liu
- Research Centre of Basic Intergrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| | - Jiahui Cao
- Research Centre of Basic Intergrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| | - Dongfeng Chen
- Research Centre of Basic Intergrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| | - Yuanming Diao
- Research Centre of Basic Intergrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| |
Collapse
|
37
|
Dhawka L, Palfini V, Hambright E, Blanco I, Poon C, Kahl A, Resch U, Bhawal R, Benakis C, Balachandran V, Holder A, Zhang S, Iadecola C, Hochrainer K. Post-ischemic ubiquitination at the postsynaptic density reversibly influences the activity of ischemia-relevant kinases. Commun Biol 2024; 7:321. [PMID: 38480905 PMCID: PMC10937959 DOI: 10.1038/s42003-024-06009-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 03/04/2024] [Indexed: 03/17/2024] Open
Abstract
Ubiquitin modifications alter protein function and stability, thereby regulating cell homeostasis and viability, particularly under stress. Ischemic stroke induces protein ubiquitination at the ischemic periphery, wherein cells remain viable, however the identity of ubiquitinated proteins is unknown. Here, we employed a proteomics approach to identify these proteins in mice undergoing ischemic stroke. The data are available in a searchable web interface ( https://hochrainerlab.shinyapps.io/StrokeUbiOmics/ ). We detected increased ubiquitination of 198 proteins, many of which localize to the postsynaptic density (PSD) of glutamatergic neurons. Among these were proteins essential for maintaining PSD architecture, such as PSD95, as well as NMDA and AMPA receptor subunits. The largest enzymatic group at the PSD with elevated post-ischemic ubiquitination were kinases, such as CaMKII, PKC, Cdk5, and Pyk2, whose aberrant activities are well-known to contribute to post-ischemic neuronal death. Concurrent phospho-proteomics revealed altered PSD-associated phosphorylation patterns, indicative of modified kinase activities following stroke. PSD-located CaMKII, PKC, and Cdk5 activities were decreased while Pyk2 activity was increased after stroke. Removal of ubiquitin restored kinase activities to pre-stroke levels, identifying ubiquitination as the responsible molecular mechanism for post-ischemic kinase regulation. These findings unveil a previously unrecognized role of ubiquitination in the regulation of essential kinases involved in ischemic injury.
Collapse
Affiliation(s)
- Luvna Dhawka
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Victoria Palfini
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Emma Hambright
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Ismary Blanco
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Carrie Poon
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Anja Kahl
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Ulrike Resch
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Ruchika Bhawal
- Institute of Biotechnology, Cornell University, Ithaca, NY, USA
| | - Corinne Benakis
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
- Institute for Stroke and Dementia Research, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Vaishali Balachandran
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Alana Holder
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Sheng Zhang
- Institute of Biotechnology, Cornell University, Ithaca, NY, USA
| | - Costantino Iadecola
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Karin Hochrainer
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
38
|
Araki Y, Rajkovich KE, Gerber EE, Gamache TR, Johnson RC, Tran THN, Liu B, Zhu Q, Hong I, Kirkwood A, Huganir R. SynGAP regulates synaptic plasticity and cognition independently of its catalytic activity. Science 2024; 383:eadk1291. [PMID: 38422154 PMCID: PMC11188940 DOI: 10.1126/science.adk1291] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 12/28/2023] [Indexed: 03/02/2024]
Abstract
SynGAP is an abundant synaptic GTPase-activating protein (GAP) critical for synaptic plasticity, learning, memory, and cognition. Mutations in SYNGAP1 in humans result in intellectual disability, autistic-like behaviors, and epilepsy. Heterozygous Syngap1-knockout mice display deficits in synaptic plasticity, learning, and memory and exhibit seizures. It is unclear whether SynGAP imparts structural properties at synapses independently of its GAP activity. Here, we report that inactivating mutations within the GAP domain do not inhibit synaptic plasticity or cause behavioral deficits. Instead, SynGAP modulates synaptic strength by physically competing with the AMPA-receptor-TARP excitatory receptor complex in the formation of molecular condensates with synaptic scaffolding proteins. These results have major implications for developing therapeutic treatments for SYNGAP1-related neurodevelopmental disorders.
Collapse
Affiliation(s)
| | | | | | | | - Richard C. Johnson
- Department of Neuroscience, Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Thanh Hai N. Tran
- Department of Neuroscience, Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Bian Liu
- Department of Neuroscience, Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Qianwen Zhu
- Department of Neuroscience, Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ingie Hong
- Department of Neuroscience, Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Alfredo Kirkwood
- Department of Neuroscience, Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Richard Huganir
- Department of Neuroscience, Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
39
|
Ulu E, Demirci E, Sener EF, Özmen S, Gul MK, Tahtasakal R, Dal F. Role of Glutamate Receptor-related Biomarkers in the Etiopathogenesis of ADHD. CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE : THE OFFICIAL SCIENTIFIC JOURNAL OF THE KOREAN COLLEGE OF NEUROPSYCHOPHARMACOLOGY 2024; 22:79-86. [PMID: 38247414 PMCID: PMC10811385 DOI: 10.9758/cpn.23.1056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 03/28/2023] [Accepted: 05/02/2023] [Indexed: 01/23/2024]
Abstract
Objective : Pathways associated with glutamate receptors are known to play a role in the pathophysiology of attention-deficit hyperactivity disorder (ADHD). However, cyclin-dependent kinase 5 (CDK5), microtubule-associated protein-2 (MAP2), guanylate kinase-associated protein (GKAP), and postsynaptic density 95 (PSD95), all of which are biomarkers involved in neurodevelopmental processes closely related to glutamatergic pathways, have not previously been studied in patients with ADHD. The main purpose of this study was to evaluate the plasma levels of CDK5, MAP2, GKAP, and PSD95 in children with ADHD and investigate whether these markers have a role in the etiology of ADHD. Methods : Ninety-six children with ADHD between 6 and 15 years of age and 72 healthy controls were included in the study. Five milliliters of blood samples were taken from all participants. The samples were stored at -80°C until analyzed by the enzyme-linked immunosorbent assay method. Results : Statistically significantly lower CDK5 levels were observed in children with ADHD than in healthy controls (p = 0.037). The MAP2, GKAP, and PSD95 levels were found to be statistically significantly higher in the ADHD group than in healthy controls (p = 0.012, p = 0.009, and p = 0.024, respectively). According to binary regression analysis, CDK5 and MAP2 levels were found to be predictors of ADHD. Conclusion : In conclusion, we found that a close relationship existed between ADHD and glutamatergic pathways, and low levels of CDK5 and high levels of MAP2 and GKAP played a role in the etiopathogenesis of ADHD.
Collapse
Affiliation(s)
- Ebru Ulu
- Department of Child and Adolescent Psychiatry, Erciyes University School of Medicine, Kayseri, Turkey
| | - Esra Demirci
- Department of Child and Adolescent Psychiatry, Erciyes University School of Medicine, Kayseri, Turkey
| | - Elif Funda Sener
- Erciyes University Genome and Stem Cell Center (GENKOK), Department of Medical Biology, Erciyes University Faculty of Medicine, Kayseri, Turkey
| | - Sevgi Özmen
- Department of Child and Adolescent Psychiatry, Erciyes University School of Medicine, Kayseri, Turkey
| | - Melike Kevser Gul
- Department of Child and Adolescent Psychiatry, Kayseri City Hospital, Kayseri, Turkey
| | - Reyhan Tahtasakal
- Erciyes University Genome and Stem Cell Center (GENKOK), Department of Medical Biology, Erciyes University Faculty of Medicine, Kayseri, Turkey
| | - Fatma Dal
- Erciyes University Genome and Stem Cell Center (GENKOK), Department of Medical Biology, Erciyes University Faculty of Medicine, Kayseri, Turkey
| |
Collapse
|
40
|
Tokunaga S, Shimomura H, Taniguchi N, Yanagi K, Kaname T, Okamoto N, Takeshima Y. A novel DLG4 variant causes DLG4-related synaptopathy with intellectual regression. Hum Genome Var 2024; 11:1. [PMID: 38182567 PMCID: PMC10770362 DOI: 10.1038/s41439-023-00260-x] [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: 08/25/2023] [Revised: 11/09/2023] [Accepted: 11/21/2023] [Indexed: 01/07/2024] Open
Abstract
DLG4-related synaptopathy is a neurodevelopmental disorder caused by a DLG4 variant. We identified a novel de novo heterozygous frameshift variant, NM_001321075.3(DLG4):c.554_563del, in a Japanese girl. Intellectual regression without motor delay was observed at 2 years of age, and she was diagnosed with autism spectrum disorder and attention-deficit/hyperactivity disorder. Recognizing the possibility of DLG4-related synaptopathy in patients with intellectual regression is important for ensuring an accurate diagnosis.
Collapse
Affiliation(s)
- Sachi Tokunaga
- Department of Pediatrics, Hyogo Medical University School of Medicine, Nishinomiya, Hyogo, Japan.
| | - Hideki Shimomura
- Department of Pediatrics, Hyogo Medical University School of Medicine, Nishinomiya, Hyogo, Japan
| | - Naoko Taniguchi
- Department of Pediatrics, Hyogo Medical University School of Medicine, Nishinomiya, Hyogo, Japan
| | - Kumiko Yanagi
- Department of Genome Medicine, National Center for Child Health and Development, Tokyo, Japan
| | - Tadashi Kaname
- Department of Genome Medicine, National Center for Child Health and Development, Tokyo, Japan
| | - Nobuhiko Okamoto
- Department of Medical Genetics, Osaka Women's and Children's Hospital, Osaka, Japan
| | - Yasuhiro Takeshima
- Department of Pediatrics, Hyogo Medical University School of Medicine, Nishinomiya, Hyogo, Japan
| |
Collapse
|
41
|
Ito H, Morishita R, Nagata KI. Simple Method for the Preparation of Postsynaptic Density Fraction from Mouse Brain. Methods Mol Biol 2024; 2794:71-78. [PMID: 38630221 DOI: 10.1007/978-1-0716-3810-1_7] [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] [Indexed: 04/19/2024]
Abstract
Postsynaptic density (PSD) is a morphologically and functionally specialized postsynaptic membrane structure of excitatory synapses. It contains hundreds of proteins such as neurotransmitter receptors, adhesion molecules, cytoskeletal proteins, and signaling enzymes. The study of the molecular architecture of the PSD is one of the most intriguing issues in neuroscience research. The isolation of the PSD from the brain of an animal is necessary for subsequent biochemical and morphological analyses. Many laboratories have developed methods to isolate PSD from the animal brain. In this chapter, we present a simple method to isolate PSD from the mouse brain using sucrose density gradient-based purification of synaptosomes followed by detergent extraction.
Collapse
Affiliation(s)
- Hidenori Ito
- Department of Molecular Neurobiology, Institute for Developmental Research, Aichi Developmental Disability Center, Aichi, Japan.
| | - Rika Morishita
- Department of Molecular Neurobiology, Institute for Developmental Research, Aichi Developmental Disability Center, Aichi, Japan
| | - Koh-Ichi Nagata
- Department of Molecular Neurobiology, Institute for Developmental Research, Aichi Developmental Disability Center, Aichi, Japan
- Department of Neurochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
42
|
Imrie G, Gray MB, Raghuraman V, Farhy-Tselnicker I. Gene Expression at the Tripartite Synapse: Bridging the Gap Between Neurons and Astrocytes. ADVANCES IN NEUROBIOLOGY 2024; 39:95-136. [PMID: 39190073 DOI: 10.1007/978-3-031-64839-7_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
Astrocytes, a major class of glial cells, are an important element at the synapse where they engage in bidirectional crosstalk with neurons to regulate numerous aspects of neurotransmission, circuit function, and behavior. Mutations in synapse-related genes expressed in both neurons and astrocytes are central factors in a vast number of neurological disorders, making the proteins that they encode prominent targets for therapeutic intervention. Yet, while the roles of many of these synaptic proteins in neurons are well established, the functions of the same proteins in astrocytes are largely unknown. This gap in knowledge must be addressed to refine therapeutic approaches. In this chapter, we integrate multiomic meta-analysis and a comprehensive overview of current literature to show that astrocytes express an astounding number of genes that overlap with the neuronal and synaptic transcriptomes. Further, we highlight recent reports that characterize the expression patterns and potential novel roles of these genes in astrocytes in both physiological and pathological conditions, underscoring the importance of considering both cell types when investigating the function and regulation of synaptic proteins.
Collapse
Affiliation(s)
- Gillian Imrie
- Department of Biology, Texas A&M University, College Station, TX, USA
| | - Madison B Gray
- Department of Biology, Texas A&M University, College Station, TX, USA
| | - Vishnuvasan Raghuraman
- Department of Biology, Texas A&M University, College Station, TX, USA
- Department of Computer Science and Engineering, Texas A&M University, College Station, TX, USA
| | - Isabella Farhy-Tselnicker
- Department of Biology, Texas A&M University, College Station, TX, USA.
- Texas A&M Institute for Neuroscience (TAMIN), Texas A&M University, College Station, TX, USA.
- Center for Biological Clocks Research, Texas A&M University, College Station, TX, USA.
| |
Collapse
|
43
|
Mirmosayyeb O, Yazdan Panah M, Mokary Y, Ghaffary EM, Ghoshouni H, Zivadinov R, Weinstock-Guttman B, Jakimovski D. Optical coherence tomography (OCT) measurements and disability in multiple sclerosis (MS): A systematic review and meta-analysis. J Neurol Sci 2023; 454:120847. [PMID: 37924591 DOI: 10.1016/j.jns.2023.120847] [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: 08/23/2023] [Revised: 09/28/2023] [Accepted: 10/18/2023] [Indexed: 11/06/2023]
Abstract
BACKGROUND Studies have demonstrated that people with multiple sclerosis (pwMS) experience visual impairments and neurodegenerative retinal processes. The disability progression in pwMS may be associated with retinal changes assessed with optical coherence tomography (OCT). This meta-analysis aims at synthesizing the correlations between OCT measurements of disability in pwMS. METHODS We systematically searched four databases (PubMed/MEDLINE, Embase, Scopus, and Web of Science) from inception to November 2022, then conducted a meta-analysis using a random effects model to determine the pooled correlation coefficient(r) between OCT measurements and disability scales by R version 4.2.3 with the meta version 6.2-1 package. RESULTS From 3129 studies, 100 studies were included. Among 9051 pwMS, the female-to-male ratio was 3.15:1, with a mean age of 39.57 ± 6.07 years. The mean disease duration and Expanded Disability Status Scale (EDSS) were 8.5 ± 3.7 and 2.7 ± 1.1, respectively. Among the pooled subgroup analyses, macular ganglion cell inner plexiform layer (mGCIPL) in patients with relapsing-remitting (pwRRMS) and peripapillary retinal nerve fiber layer (pRNFL) in patients with progressive MS (pwPMS) had strong correlations with EDSS, r = -0.33 (95% CI: -0.45 to -0.20, I2 = 45%, z-score = -4.86, p < 0.001) and r = -0.20 (95% CI:-0.58 to 0.26, I2 = 76%, z-score = -0.85, p = 0.395), respectively. According to subgroup analysis on pwMS without optic neuritis (ON) history, the largest correlation was seen between EDSS and macular ganglion cell complex (mGCC): r = -0.39 (95% CI: -0.70 to 0.04, I2 = 79%, z-score = -1.79, p = 0.073). CONCLUSION OCT measurements are correlated with disability in pwMS, and they can complement the comprehensive neurological visit as an additional paraclinical test.
Collapse
Affiliation(s)
- Omid Mirmosayyeb
- Department of Neurology, Jacobs Comprehensive MS Treatment and Research Center, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Mohammad Yazdan Panah
- Isfahan Neurosciences Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Yousef Mokary
- Isfahan Neurosciences Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Elham Moases Ghaffary
- Isfahan Neurosciences Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hamed Ghoshouni
- Isfahan Neurosciences Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Robert Zivadinov
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, 100 High Street, Buffalo, NY 14203, USA; Center for Biomedical Imaging at the Clinical Translational Science Institute, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Bianca Weinstock-Guttman
- Department of Neurology, Jacobs Comprehensive MS Treatment and Research Center, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Dejan Jakimovski
- Department of Neurology, Jacobs Comprehensive MS Treatment and Research Center, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA; Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, 100 High Street, Buffalo, NY 14203, USA.
| |
Collapse
|
44
|
Woelfle S, Pedro MT, Wagner J, Schön M, Boeckers TM. Expression profiles of the autism-related SHANK proteins in the human brain. BMC Biol 2023; 21:254. [PMID: 37953224 PMCID: PMC10641957 DOI: 10.1186/s12915-023-01712-0] [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: 03/17/2023] [Accepted: 09/25/2023] [Indexed: 11/14/2023] Open
Abstract
BACKGROUND SHANKs are major scaffolding proteins at postsynaptic densities (PSDs) in the central nervous system. Mutations in all three family members have been associated with neurodevelopmental disorders such as autism spectrum disorders (ASDs). Despite the pathophysiological importance of SHANK2 and SHANK3 mutations in humans, research on the expression of these proteins is mostly based on rodent model organisms. RESULTS In the present study, cellular and neuropil SHANK2 expression was analyzed by immunofluorescence (IF) staining of post mortem human brain tissue from four male individuals (19 brain regions). Mouse brains were analyzed in comparison to evaluate the degree of phylogenetic conservation. Furthermore, SHANK2 and SHANK3 isoform patterns were compared in human and mouse brain lysates. While isoform expression and subcellular distribution were largely conserved, differences in neuropil levels of SHANK2 were found by IF staining: Maximum expression was concordantly measured in the cerebellum; however, higher SHANK2 expression was detected in the human brainstem and thalamus when compared to mice. One of the lowest SHANK2 levels was found in the human amygdala, a moderately expressing region in mouse. Quantification of SHANK3 IF in mouse brains unveiled a distribution comparable to humans. CONCLUSIONS In summary, these data show that the overall expression pattern of SHANK is largely conserved in defined brain regions; however, differences do exist, which need to be considered in the translation of rodent studies. The summarized expression patterns of SHANK2 and SHANK3 should serve as a reference for future studies.
Collapse
Affiliation(s)
- Sarah Woelfle
- Institute for Anatomy and Cell Biology, Ulm University, Albert-Einstein-Allee 11, 89081, Ulm, Germany
| | - Maria T Pedro
- Department of Neurosurgery, Ulm University, Campus Günzburg, Lindenallee 2, 89312, Günzburg, Germany
| | - Jan Wagner
- Department of Neurology, Ulm University and Universitäts- and Rehabilitationskliniken Ulm, 89081, Ulm, Germany
| | - Michael Schön
- Institute for Anatomy and Cell Biology, Ulm University, Albert-Einstein-Allee 11, 89081, Ulm, Germany.
| | - Tobias M Boeckers
- Institute for Anatomy and Cell Biology, Ulm University, Albert-Einstein-Allee 11, 89081, Ulm, Germany.
- Deutsches Zentrum Für Neurodegenerative Erkrankungen, DZNE, Ulm Site, 89081, Ulm, Germany.
| |
Collapse
|
45
|
Noh YW, Kim Y, Lee S, Kim Y, Shin JJ, Kang H, Kim IH, Kim E. The PFC-LH-VTA pathway contributes to social deficits in IRSp53-mutant mice. Mol Psychiatry 2023; 28:4642-4654. [PMID: 37730842 PMCID: PMC10914623 DOI: 10.1038/s41380-023-02257-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 09/01/2023] [Accepted: 09/08/2023] [Indexed: 09/22/2023]
Abstract
Dopamine (DA) neurons in the ventral tegmental area (VTA) promote social brain functions by releasing DA onto nucleus accumbens neurons, but it remains unclear how VTA neurons communicate with cortical neurons. Here, we report that the medial prefrontal cortex (mPFC)-lateral hypothalamus (LH)-VTA pathway contributes to social deficits in mice with IRSp53 deletion restricted to cortical excitatory neurons (Emx1-Cre;Irsp53fl/fl mice). LH-projecting mutant mPFC neurons display abnormally increased excitability involving decreased potassium channel gene expression, leading to excessive excitatory synaptic input to LH-GABA neurons. A circuit-specific IRSp53 deletion in LH-projecting mPFC neurons also increases neuronal excitability and induces social deficits. LH-GABA neurons with excessive mPFC excitatory synaptic input show a compensatory decrease in excitability, weakening the inhibitory LHGABA-VTAGABA pathway and subsequently over-activating VTA-GABA neurons and over-inhibiting VTA-DA neurons. Accordingly, optogenetic activation of the LHGABA-VTAGABA pathway improves social deficits in Emx1-Cre;Irsp53fl/fl mice. Therefore, the mPFC-LHGABA-VTAGABA-VTADA pathway contributes to the social deficits in Emx1-Cre;Irsp53fl/fl mice.
Collapse
Affiliation(s)
- Young Woo Noh
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Korea
| | - Yangsik Kim
- Department of Psychiatry, Inha University Hospital, Incheon, 22332, Korea
| | - Soowon Lee
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, 34141, Korea
| | - Yeonghyeon Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Korea
| | - Jae Jin Shin
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science, Daejeon, 34141, Korea
| | - Hyojin Kang
- Division of National Supercomputing, Korea Institute of Science and Technology Information (KISTI), Daejeon, 34141, Korea
| | - Il Hwan Kim
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Eunjoon Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Korea.
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science, Daejeon, 34141, Korea.
| |
Collapse
|
46
|
Wang L, Pang K, Zhou L, Cebrián-Silla A, González-Granero S, Wang S, Bi Q, White ML, Ho B, Li J, Li T, Perez Y, Huang EJ, Winkler EA, Paredes MF, Kovner R, Sestan N, Pollen AA, Liu P, Li J, Piao X, García-Verdugo JM, Alvarez-Buylla A, Liu Z, Kriegstein AR. A cross-species proteomic map reveals neoteny of human synapse development. Nature 2023; 622:112-119. [PMID: 37704727 PMCID: PMC10576238 DOI: 10.1038/s41586-023-06542-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 08/15/2023] [Indexed: 09/15/2023]
Abstract
The molecular mechanisms and evolutionary changes accompanying synapse development are still poorly understood1,2. Here we generate a cross-species proteomic map of synapse development in the human, macaque and mouse neocortex. By tracking the changes of more than 1,000 postsynaptic density (PSD) proteins from midgestation to young adulthood, we find that PSD maturation in humans separates into three major phases that are dominated by distinct pathways. Cross-species comparisons reveal that human PSDs mature about two to three times slower than those of other species and contain higher levels of Rho guanine nucleotide exchange factors (RhoGEFs) in the perinatal period. Enhancement of RhoGEF signalling in human neurons delays morphological maturation of dendritic spines and functional maturation of synapses, potentially contributing to the neotenic traits of human brain development. In addition, PSD proteins can be divided into four modules that exert stage- and cell-type-specific functions, possibly explaining their differential associations with cognitive functions and diseases. Our proteomic map of synapse development provides a blueprint for studying the molecular basis and evolutionary changes of synapse maturation.
Collapse
Affiliation(s)
- Li Wang
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA.
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA.
| | - Kaifang Pang
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Li Zhou
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Arantxa Cebrián-Silla
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Susana González-Granero
- Laboratory of Comparative Neurobiology, Cavanilles Institute of Biodiversity and Evolutionary Biology, University of Valencia and CIBERNED, Valencia, Spain
| | - Shaohui Wang
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Qiuli Bi
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Matthew L White
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Brandon Ho
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Jiani Li
- Gilead Sciences, Foster City, CA, USA
| | - Tao Li
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
| | - Yonatan Perez
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Eric J Huang
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA
| | - Ethan A Winkler
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Mercedes F Paredes
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Rothem Kovner
- Department of Neuroscience, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Nenad Sestan
- Department of Neuroscience, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Alex A Pollen
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Pengyuan Liu
- Department of Chemistry, University of Massachusetts Lowell, Lowell, MA, USA
| | - Jingjing Li
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Xianhua Piao
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Division of Neonatology, Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
- Newborn Brain Research Institute, University of California San Francisco, San Francisco, CA, USA
| | - José Manuel García-Verdugo
- Laboratory of Comparative Neurobiology, Cavanilles Institute of Biodiversity and Evolutionary Biology, University of Valencia and CIBERNED, Valencia, Spain
| | - Arturo Alvarez-Buylla
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Zhandong Liu
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Arnold R Kriegstein
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA.
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
47
|
Zhuang Y, Xu X, Li H, Niu F, Yang M, Ge Q, Lu S, Deng Y, Wu H, Zhang B, Liu B. Megf10-related engulfment of excitatory postsynapses by astrocytes following severe brain injury. CNS Neurosci Ther 2023; 29:2873-2883. [PMID: 37081759 PMCID: PMC10493650 DOI: 10.1111/cns.14223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 01/13/2023] [Accepted: 01/18/2023] [Indexed: 04/22/2023] Open
Abstract
AIMS To investigate astrocyte-related phagocytosis of synapses in the ipsilateral hippocampus after traumatic brain injury (TBI). METHODS We performed controlled cortical impact to simulate TBI in mice. Seven days postinjury, we performed cognitive tests, synapse quantification, and examination of astrocytic phagocytosis in association with Megf10 expression. RESULTS During the subacute stage post-TBI, we found a reduction in excitatory postsynaptic materials in the ipsilateral hippocampus, which was consistent with poor performance in the cognitive test. The transcriptome data suggested that robust phagocytosis was responsible for this process. Coincidently, we identified phagocytic astrocytes containing secondary lysosomes that were wrapped around the synapses in the ipsilateral hippocampus. Moreover, a significant increase in the co-location of GFAP and PSD-95 in the CA1 region suggested astrocytic engulfment of excitatory postsynaptic proteins. After examining the reported phagocytic pathways, we found that both the transcription level and protein expression of Megf10 were elevated. Co-immunofluorescence of GFAP and Megf10 demonstrated that the expression of Megf10 was spatially upregulated in astrocytes, exclusively in the CA1 region, and was related to the astrocytic engulfment of PSD-95. CONCLUSION Our study elaborated that the Megf10-related astrocytic engulfment of PSD-95 in the CA1 region of the ipsilateral hippocampus aggravated cognitive dysfunction following severe TBI.
Collapse
Affiliation(s)
- Yuan Zhuang
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Xiaojian Xu
- Beijing Key Laboratory of Central Nervous System InjuryBeijing Neurosurgical Institute, Capital Medical UniversityBeijingChina
| | - Hao Li
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Fei Niu
- Beijing Key Laboratory of Central Nervous System InjuryBeijing Neurosurgical Institute, Capital Medical UniversityBeijingChina
| | - Mengshi Yang
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Qianqian Ge
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Shenghua Lu
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Yu Deng
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Hongbin Wu
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Bin Zhang
- Department of Intensive Care Unit, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Baiyun Liu
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
- Beijing Key Laboratory of Central Nervous System InjuryBeijing Neurosurgical Institute, Capital Medical UniversityBeijingChina
- Center for Nerve Injury and RepairBeijing Institute of Brain DisordersBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
| |
Collapse
|
48
|
Dunot J, Ribera A, Pousinha PA, Marie H. Spatiotemporal insights of APP function. Curr Opin Neurobiol 2023; 82:102754. [PMID: 37542943 DOI: 10.1016/j.conb.2023.102754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 06/29/2023] [Accepted: 07/07/2023] [Indexed: 08/07/2023]
Abstract
The amyloid-β precursor protein (APP) is a ubiquitous protein with a strong genetic link to Alzheimer's disease. Although the protein was identified more than forty years ago, its physiological function is still unclear. In recent years, advances in technology have allowed researchers to tackle APP functions in greater depth. In this review, we discuss the latest research pertaining to APP functions from development to aging. We also address the different roles that APP could play in specific types of cells of the central and peripheral nervous system and in other organs of the body. We argue that, until we fully identify the functions of APP in space and time, we will be missing important pieces of the puzzle to solve its pathological implication in Alzheimer's disease and beyond.
Collapse
Affiliation(s)
- Jade Dunot
- Université Côte d'Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275, 06560, Valbonne, France. https://twitter.com/DunotJade
| | - Aurore Ribera
- Université Côte d'Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275, 06560, Valbonne, France. https://twitter.com/aurore_et_al_
| | - Paula A Pousinha
- Université Côte d'Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275, 06560, Valbonne, France.
| | - Hélène Marie
- Université Côte d'Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275, 06560, Valbonne, France.
| |
Collapse
|
49
|
Edwards N, Combrinck C, McCaughey-Chapman A, Connor B. Directly reprogrammed fragile X syndrome dorsal forebrain precursor cells generate cortical neurons exhibiting impaired neuronal maturation. Front Cell Neurosci 2023; 17:1254412. [PMID: 37810261 PMCID: PMC10552551 DOI: 10.3389/fncel.2023.1254412] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 09/01/2023] [Indexed: 10/10/2023] Open
Abstract
Introduction The neurodevelopmental disorder fragile X syndrome (FXS) is the most common monogenic cause of intellectual disability associated with autism spectrum disorder. Inaccessibility to developing human brain cells is a major barrier to studying FXS. Direct-to-neural precursor reprogramming provides a unique platform to investigate the developmental profile of FXS-associated phenotypes throughout neural precursor and neuron generation, at a temporal resolution not afforded by post-mortem tissue and in a patient-specific context not represented in rodent models. Direct reprogramming also circumvents the protracted culture times and low efficiency of current induced pluripotent stem cell strategies. Methods We have developed a chemically modified mRNA (cmRNA) -based direct reprogramming protocol to generate dorsal forebrain precursors (hiDFPs) from FXS patient-derived fibroblasts, with subsequent differentiation to glutamatergic cortical neurons and astrocytes. Results We observed differential expression of mature neuronal markers suggesting impaired neuronal development and maturation in FXS- hiDFP-derived neurons compared to controls. FXS- hiDFP-derived cortical neurons exhibited dendritic growth and arborization deficits characterized by reduced neurite length and branching consistent with impaired neuronal maturation. Furthermore, FXS- hiDFP-derived neurons exhibited a significant decrease in the density of pre- and post- synaptic proteins and reduced glutamate-induced calcium activity, suggesting impaired excitatory synapse development and functional maturation. We also observed a reduced yield of FXS- hiDFP-derived neurons with a significant increase in FXS-affected astrocytes. Discussion This study represents the first reported derivation of FXS-affected cortical neurons following direct reprogramming of patient fibroblasts to dorsal forebrain precursors and subsequently neurons that recapitulate the key molecular hallmarks of FXS as it occurs in human tissue. We propose that direct to hiDFP reprogramming provides a unique platform for further study into the pathogenesis of FXS as well as the identification and screening of new drug targets for the treatment of FXS.
Collapse
Affiliation(s)
| | | | | | - Bronwen Connor
- Department of Pharmacology and Clinical Pharmacology, Centre for Brain Research, School of Medical Science, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
50
|
Belapurkar V, Mahadeva Swamy HS, Singh N, Kedia S, Setty SRG, Jose M, Nair D. Real-time heterogeneity of supramolecular assembly of amyloid precursor protein is modulated by an endocytic risk factor PICALM. Cell Mol Life Sci 2023; 80:295. [PMID: 37726569 PMCID: PMC11072284 DOI: 10.1007/s00018-023-04939-w] [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: 04/05/2023] [Revised: 07/10/2023] [Accepted: 07/25/2023] [Indexed: 09/21/2023]
Abstract
Recently, the localization of amyloid precursor protein (APP) into reversible nanoscale supramolecular assembly or "nanodomains" has been highlighted as crucial towards understanding the onset of the molecular pathology of Alzheimer's disease (AD). Surface expression of APP is regulated by proteins interacting with it, controlling its retention and lateral trafficking on the synaptic membrane. Here, we evaluated the involvement of a key risk factor for AD, PICALM, as a critical regulator of nanoscale dynamics of APP. Although it was enriched in the postsynaptic density, PICALM was also localized to the presynaptic active zone and the endocytic zone. PICALM colocalized with APP and formed nanodomains with distinct morphological properties in different subsynaptic regions. Next, we evaluated if this localization to subsynaptic compartments was regulated by the C-terminal sequences of APP, namely, the "Y682ENPTY687" domain. Towards this, we found that deletion of C-terminal regions of APP with partial or complete deletion of Y682ENPTY687, namely, APP-Δ9 and APP-Δ14, affected the lateral diffusion and nanoscale segregation of APP. Lateral diffusion of APP mutant APP-Δ14 sequence mimicked that of a detrimental Swedish mutant of APP, namely, APP-SWE, while APP-Δ9 diffused similar to wild-type APP. Interestingly, elevated expression of PICALM differentially altered the lateral diffusion of the APP C-terminal deletion mutants. These observations confirm that the C-terminal sequence of APP regulates its lateral diffusion and the formation of reversible nanoscale domains. Thus, when combined with autosomal dominant mutations, it generates distinct molecular patterns leading to onset of Alzheimer's disease (AD).
Collapse
Affiliation(s)
- Vivek Belapurkar
- Centre for Neuroscience, Indian Institute of Science, Bangalore, 560012, India
- Interdisciplinary Institute for Neuroscience CNRS UMR5297, University of Bordeaux, Bordeaux, France
| | - H S Mahadeva Swamy
- Centre for Neuroscience, Indian Institute of Science, Bangalore, 560012, India
- Tata Institute for Genetics and Society, Bengaluru, India
| | - Nivedita Singh
- Centre for Neuroscience, Indian Institute of Science, Bangalore, 560012, India
| | - Shekhar Kedia
- Centre for Neuroscience, Indian Institute of Science, Bangalore, 560012, India
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Subba Rao Gangi Setty
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, 560012, India
| | - Mini Jose
- Centre for Neuroscience, Indian Institute of Science, Bangalore, 560012, India
| | - Deepak Nair
- Centre for Neuroscience, Indian Institute of Science, Bangalore, 560012, India.
| |
Collapse
|