1
|
Fok AHK, Lam CHM, Lai CSW. Specific dendritic spine modifications and dendritic transport: From in vitro to in vivo. Neural Regen Res 2026; 21:665-666. [PMID: 39819974 DOI: 10.4103/nrr.nrr-d-24-01159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 12/04/2024] [Indexed: 01/19/2025] Open
Affiliation(s)
- Albert H K Fok
- Center for Research in Neuroscience, Department of Neurology & Neurosurgery, Brain Repair and Integrative Neuroscience Program, The Research Institute of the McGill University Health Centre, Montreal, QC, Canada (Fok AHK)
| | - Charlotte H M Lam
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China (Lam CHM, Lai CSW)
| | - Cora S W Lai
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China (Lam CHM, Lai CSW)
- Advanced Biomedical Instrumentation Center, Hong Kong Science Park, Shatin, New Territories, Hong Kong Special Administrative Region, China (Lai CSW)
- The State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong Special Administrative Region, China (Lai CSW)
| |
Collapse
|
2
|
Zheng Y, Cao L, Zhao D, Yang Q, Gu C, Mao Y, Zhu G, Zhu Y, Zhao J, Xu D. Nerve root magnetic stimulation regulates the synaptic plasticity of injured spinal cord by ascending sensory pathway. Neural Regen Res 2025; 20:3564-3573. [PMID: 40095662 PMCID: PMC11974646 DOI: 10.4103/nrr.nrr-d-24-00628] [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: 06/06/2024] [Revised: 10/21/2024] [Accepted: 12/17/2024] [Indexed: 03/19/2025] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202512000-00026/figure1/v/2025-01-31T122243Z/r/image-tiff Promoting synaptic plasticity and inducing functional reorganization of residual nerve fibers hold clinical significance for restoring motor function following spinal cord injury. Neuromagnetic stimulation targeting the nerve roots has been shown to improve motor function by enhancing nerve conduction in the injured spinal cord and restoring the synaptic ultrastructure of both the sensory and motor cortex. However, our understanding of the neurophysiological mechanisms by which nerve root magnetic stimulation facilitates motor function recovery in the spinal cord is limited, and its role in neuroplasticity remains unclear. In this study, we established a model of spinal cord injury in adult male Sprague-Dawley rats by applying moderate compression at the T10 vertebra. We then performed magnetic stimulation on the L5 nerve root for 3 weeks, beginning on day 3 post-injury. At day 22 post-injury, we observed that nerve root magnetic stimulation downregulated the level of interleukin-6 in the injured spinal cord tissue of rats. Additionally, this treatment reduced neuronal damage and glial scar formation, and increased the number of neurons in the injured spinal cord. Furthermore, nerve root magnetic stimulation decreased the levels of acetylcholine, norepinephrine, and dopamine, and increased the expression of synaptic plasticity-related mRNA and proteins PSD95, GAP43, and Synapsin II. Taken together, these results showed that nerve root magnetic stimulation alleviated neuronal damage in the injured spinal cord, regulated synaptic plasticity, and suppressed inflammatory responses. These findings provide laboratory evidence for the clinical application of nerve root magnetic stimulation in the treatment of spinal cord injury.
Collapse
Affiliation(s)
- Ya Zheng
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Lingyun Cao
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Dan Zhao
- Department of Rehabilitation Medicine, Ruijin Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Qi Yang
- Rehabilitation Medical Center, Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Chunya Gu
- Rehabilitation Medical Center, Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Yeran Mao
- Department of Rehabilitation, Baoshan Branch, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guangyue Zhu
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yulian Zhu
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
| | - Jing Zhao
- Department of Neurology, Minhang Hospital Affiliated to Fudan University, Shanghai, China
| | - Dongsheng Xu
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Engineering Research Center of Traditional Chinese Medicine Intelligent Rehabilitation, Ministry of Education, Shanghai, China
- Institute of Rehabilitation Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
3
|
Ren J, Wang Y, Wang Y, Zhang Y, Xing M, Deng S, Tong S, Wang L, Zheng C, Yang J, Ni G, Ming D. Dynamic changes of hippocampal dendritic spines in Alzheimer's disease mice among the different stages. Exp Neurol 2025; 390:115266. [PMID: 40246009 DOI: 10.1016/j.expneurol.2025.115266] [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: 01/20/2025] [Revised: 03/16/2025] [Accepted: 04/14/2025] [Indexed: 04/19/2025]
Abstract
Alzheimer's disease (AD) is characterized by the accumulation of amyloid-β (Aβ) peptides and a progressive decline in cognitive function. Hippocampus as a crucial brain area for learning and memory, is also adversely affected by AD's pathology. The accumulation of Aβ is often associated with the loss of dendritic spines of the hippocampus. However, the dynamic alterations in dendritic spines throughout AD progression are not fully understood. To investigate it, we conducted in-vivo imaging in two mouse models representing the early and late stages of AD pathology: young mice injected with Aβ1-42 oligomers and APP/PS1 transgenic mice. In the early-stage AD model, imaging was conducted at third- and fifth- weeks post-injection. In the late-stage AD model, a four-month imaging began at 14 months old. The imaging results showed spine elimination in both models. Notably, acute Aβ exposure was linked to heightened spine loss on secondary dendrites, while in the late stage the primary effect was on tertiary dendrites. Concurrently, with the metabolism of Aβ, cognition recovered to some extent by five weeks post Aβ1-42 exposure. These findings suggested that dendritic spine plasticity was impaired during the development of AD, as evidenced by increasing spine loss at different levels. However, the cognitive recovery observed in early-stage AD model mice may indicate a compensatory structural reorganization, highlighting the potential of early intervention to mitigate disease progression. Our results provide novel insights into the neurotoxic effects of Aβ1-42 and may contribute to the development of therapeutic strategies for AD.
Collapse
Affiliation(s)
- Jing Ren
- Medical School of Tianjin University, Tianjin University, Tianjin 300072, China
| | - Yimeng Wang
- Medical School of Tianjin University, Tianjin University, Tianjin 300072, China
| | - Yinuo Wang
- Medical School of Tianjin University, Tianjin University, Tianjin 300072, China
| | - Yiping Zhang
- Medical School of Tianjin University, Tianjin University, Tianjin 300072, China
| | - Mu Xing
- Medical School of Tianjin University, Tianjin University, Tianjin 300072, China
| | - Shouzhe Deng
- Medical School of Tianjin University, Tianjin University, Tianjin 300072, China
| | - Siyi Tong
- Medical School of Tianjin University, Tianjin University, Tianjin 300072, China
| | - Ling Wang
- Medical School of Tianjin University, Tianjin University, Tianjin 300072, China; Tianjin Key Laboratory of Brain Science and Neural Engineering, Tianjin University, Tianjin 300072, China; Haihe Laboratory of Brain-Computer Interaction and Human-Machine Integration, Tianjin 300392, China; State Key Laboratory of Advanced Medical Materials and Devices, Tianjin University, Tianjin 300072, China
| | - Chenguang Zheng
- Medical School of Tianjin University, Tianjin University, Tianjin 300072, China; Tianjin Key Laboratory of Brain Science and Neural Engineering, Tianjin University, Tianjin 300072, China; Haihe Laboratory of Brain-Computer Interaction and Human-Machine Integration, Tianjin 300392, China; State Key Laboratory of Advanced Medical Materials and Devices, Tianjin University, Tianjin 300072, China
| | - Jiajia Yang
- Medical School of Tianjin University, Tianjin University, Tianjin 300072, China; Tianjin Key Laboratory of Brain Science and Neural Engineering, Tianjin University, Tianjin 300072, China; Haihe Laboratory of Brain-Computer Interaction and Human-Machine Integration, Tianjin 300392, China; State Key Laboratory of Advanced Medical Materials and Devices, Tianjin University, Tianjin 300072, China.
| | - Guangjian Ni
- Medical School of Tianjin University, Tianjin University, Tianjin 300072, China; Tianjin Key Laboratory of Brain Science and Neural Engineering, Tianjin University, Tianjin 300072, China; Haihe Laboratory of Brain-Computer Interaction and Human-Machine Integration, Tianjin 300392, China; State Key Laboratory of Advanced Medical Materials and Devices, Tianjin University, Tianjin 300072, China.
| | - Dong Ming
- Medical School of Tianjin University, Tianjin University, Tianjin 300072, China; Tianjin Key Laboratory of Brain Science and Neural Engineering, Tianjin University, Tianjin 300072, China; Haihe Laboratory of Brain-Computer Interaction and Human-Machine Integration, Tianjin 300392, China; State Key Laboratory of Advanced Medical Materials and Devices, Tianjin University, Tianjin 300072, China.
| |
Collapse
|
4
|
Wang X, Wang L, Bu Q, Xiao Y, Zhao Y, Jiang L, Dai Y, Li H, Liu H, Chen Y, Flores AD, Zhao Y, Cen X. LUZP1 Regulates Dendritic Spine Maturation and Synaptic Plasticity in the Hippocampal Dentate Gyrus of Mice. J Neurosci 2025; 45:e1867242025. [PMID: 40180573 PMCID: PMC12079723 DOI: 10.1523/jneurosci.1867-24.2025] [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: 09/30/2024] [Revised: 02/16/2025] [Accepted: 03/18/2025] [Indexed: 04/05/2025] Open
Abstract
Leucine zipper protein 1 (LUZP1) functions in the maintenance and dynamics of the cytoskeleton by interacting with actin and microtubules. Deficiency or mutation of LUZP1 is associated with brain developmental disorders; however, its precise role in brain function remains unclear. We showed that LUZP1 localizes to actin and is highly expressed in CaMKIIα-expressing neurons within the mouse hippocampal dentate gyrus. Depletion of LUZP1 impedes dendritic spine maturation, which is characterized by excess immature filopodia and loss of mature mushroom spines both in vitro and in vivo. LUZP1 knockdown reduces spontaneous electrical activity and synaptic plasticity in hippocampal neurons. Conditional deletion of LUZP1 in CaMKIIα-expressing neurons causes impaired learning and memory behavior in mice of both sexes. Mechanistically, LUZP1 control dendritic maturation by directly interacting with filamin A and modulating the Rac1-PAK1 signaling pathway. These findings shed light on the role of LUZP1 in regulating synaptic plasticity and brain function.
Collapse
Affiliation(s)
- Xiaojie Wang
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Department of Neuroscience, City University of Hong Kong, Hong Kong 999077, China
| | - Liang Wang
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qian Bu
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuzhou Xiao
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yue Zhao
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Linhong Jiang
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yanping Dai
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hongchun Li
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Haxiaoyu Liu
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yaxing Chen
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Angelo D Flores
- Department of Neuroscience, City University of Hong Kong, Hong Kong 999077, China
| | - Yinglan Zhao
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaobo Cen
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
5
|
Dalto JF, Medina JH, Pastor V. Molecular Underpinnings of Memory Persistence and Forgetting. J Neurochem 2025; 169:e70089. [PMID: 40411122 DOI: 10.1111/jnc.70089] [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: 02/19/2025] [Revised: 05/07/2025] [Accepted: 05/08/2025] [Indexed: 05/26/2025]
Abstract
The fate of memories depends mainly on external and internal factors affecting cellular and systems consolidation on the one hand and the decay or weakening of the memory trace on the other hand. Over the past 40 years, research has focused on the mechanisms of memory consolidation, retrieval, and its consequences: extinction and reconsolidation. In contrast, much less is known about the molecular mechanisms required for the maintenance of memory storage and forgetting. These opposing forces are both activity- and time-dependent. Here, we summarize the molecular signatures and inherent mechanisms involved in memory persistence and active forgetting, highlighting recent findings on the role of dopamine neurotransmission, intracellular signaling cascades, and actin cytoskeleton dynamics in these processes.
Collapse
Affiliation(s)
- Juliana F Dalto
- Instituto Tecnológico de Buenos Aires (ITBA), Buenos Aires, Argentina
| | - Jorge H Medina
- Instituto Tecnológico de Buenos Aires (ITBA), Buenos Aires, Argentina
- Instituto de Biología Celular y Neurociencias "Prof. Eduardo De Robertis", CONICET-UBA, Buenos Aires, Argentina
| | - Verónica Pastor
- Instituto de Biología Celular y Neurociencias "Prof. Eduardo De Robertis", CONICET-UBA, Buenos Aires, Argentina
| |
Collapse
|
6
|
Lyukmanova E, Kirichenko A, Kulbatskii D, Isaev A, Kukushkin I, Che Y, Kirpichnikov M, Bychkov M. Water-Soluble Lynx1 Upregulates Dendritic Spine Density and Stimulates Astrocytic Network and Signaling. Mol Neurobiol 2025; 62:5531-5545. [PMID: 39565568 DOI: 10.1007/s12035-024-04627-1] [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: 07/21/2024] [Accepted: 11/09/2024] [Indexed: 11/21/2024]
Abstract
Secreted and membrane-tethered mammalian neuromodulators from the Ly6/uPAR family are involved in regulation of many physiological processes. Some of them are expressed in the CNS in the neurons of different brain regions and target neuronal membrane receptors. Thus, Lynx1 potentiates nicotinic acetylcholine receptors (nAChRs) in the brain, while others like Lypd6 and Lypd6b suppress it. However, the mechanisms underlying the regulation of cognitive processes by these neuromodulators remain unclear. Here, we showed that water-soluble analogue of Lynx1 (ws-Lynx-1) targets α7-nAChRs both in the hippocampal neurons and astrocytes. Incubation of astrocytes with ws-Lynx1 increased expression of connexins 30 and 43; α4, α5, and β4 integrins; and E- and P-cadherins. Ws-Lynx1 reduced secretion of pro-inflammatory adhesion factors ICAM-1, PSGL-1, and VCAM-1 and downregulated secretion of CD44 and NCAM, which inhibit synaptic plasticity. Moreover, increased astrocytic secretion of the dendritic growth activator ALCAM and neurogenesis regulator E-selectin was observed. Incubation of neurons with ws-Lynx1 potentiated α7-nAChRs and upregulated dendritic spine density. Thus, the pro-cognitive activity of ws-Lynx1 observed previously can be explained by stimulation of astrocytic network and signaling together with up-regulation of spinogenesis, potentiation of the α7-nAChRs, and neuronal and synaptic plasticity. For comparison, influence of water-soluble analogues of a set of Ly6/uPAR proteins (SLURP-1, SLURP-2, Lypd6, Lypd6b, and PSCA) on dendritic spine density and diameter was studied. Data obtained give new insights on the role of Ly6/uPAR proteins in the brain and open new prospects for the development of drugs to improve cognitive function.
Collapse
Affiliation(s)
- Ekaterina Lyukmanova
- Faculty of Biology, Shenzhen MSU-BIT University, Shenzhen, China.
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia.
- Moscow Center for Advanced Studies, Moscow, Russia.
- Molecular Technologies of the Living Systems and Synthetic Biology, Faculty of Biology, Interdisciplinary Scientific and Educational School of Moscow University, Lomonosov Moscow State University, Moscow, Russia.
| | - Artem Kirichenko
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Moscow Center for Advanced Studies, Moscow, Russia
| | - Dmitry Kulbatskii
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Aizek Isaev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Moscow Center for Advanced Studies, Moscow, Russia
| | - Ilya Kukushkin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Moscow Center for Advanced Studies, Moscow, Russia
| | - Yuqi Che
- Faculty of Biology, Shenzhen MSU-BIT University, Shenzhen, China
| | - Mikhail Kirpichnikov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Molecular Technologies of the Living Systems and Synthetic Biology, Faculty of Biology, Interdisciplinary Scientific and Educational School of Moscow University, Lomonosov Moscow State University, Moscow, Russia
| | - Maxim Bychkov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
7
|
Ge Q, Zhou S, Porras J, Fu P, Wang T, Du J, Li K. Investigating post-infection anxiety- and depression-like behaviors in a SARS-CoV-2 mouse model. Theranostics 2025; 15:5738-5755. [PMID: 40365287 PMCID: PMC12068287 DOI: 10.7150/thno.102752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 03/27/2025] [Indexed: 05/15/2025] Open
Abstract
Rationale: The COVID-19 pandemic, driven by SARS-CoV-2, has resulted in a wide range of neuropsychiatric symptoms associated with post-acute sequelae (PASC). However, the mechanisms by which SARS-CoV-2 impacts the brain and leads to persistent behavioral changes remain poorly understood. We hypothesize that SARS-CoV-2 exposure induces neuroinflammation and microglial activation, leading to anxiety- and depression-like behaviors in mice. Methods: We established a SARS-CoV-2 mouse model using the virulent SARS2-N501YMA30 strain to investigate its impact on the central nervous system (CNS). We assessed neuroinvasion via immunostaining of dsRNA and markers for neuronal, astrocyte, and microglia in brain slices. Behavioral changes were evaluated at 2 weeks, 2 months, and 4 months post-infection. Molecular and cellular analyses included bulk RNA-seq, Golgi-Cox staining, field excitatory postsynaptic potential (fEPSP) recordings, immunofluorescence, and quantitative real-time PCR (qRT-PCR) to assess gene expression, neuronal morphology, and microglial activation in the brain. Results: We demonstrated that intranasal inoculation of SARS2-N501YMA30 results in viral dissemination to multiple brain regions, including the amygdala and the prefrontal cortex (PFC). Behavioral assays indicated a marked elevation in anxiety- and depression-like behaviors post-infection. A comparative analysis of RNA expression profiles disclosed alterations in the post-infected brains. Additionally, we observed dendritic spine remodeling on neurons within the amygdala after infection. Infection with SARS2-N501YMA30 was associated with microglial activation and a subsequent increase in microglia-dependent neuronal activity in the amygdala. Transcriptomic analysis of infected brains revealed the upregulation of inflammatory and cytokine-related pathways, implicating neuroinflammation in the pathogenesis of neuronal hyperactivity and behavioral abnormality. Conclusion: Our findings provide evidence that SARS-CoV-2 neuroinvasion plays a critical role in the development of lasting behavioral sequelae observed in PASC. These data provide critical insights into the neurological consequences of SARS-CoV-2 infection and underscore microglia as a potential therapeutic target for ameliorating virus-induced neurobehavioral abnormalities.
Collapse
Affiliation(s)
- Qian Ge
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Shan Zhou
- Florida Research and Innovation Center, Cleveland Clinic, Port St. Lucie, FL 34987, USA
| | - Jose Porras
- Florida Research and Innovation Center, Cleveland Clinic, Port St. Lucie, FL 34987, USA
| | - Panfeng Fu
- Center for Translational Science, Florida International University, Port St. Lucie, FL, 34987, USA
| | - Ting Wang
- Center for Translational Science, Florida International University, Port St. Lucie, FL, 34987, USA
| | - Jianyang Du
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Kun Li
- Florida Research and Innovation Center, Cleveland Clinic, Port St. Lucie, FL 34987, USA
| |
Collapse
|
8
|
Schünemann KD, Hattingh RM, Verhoog MB, Yang D, Bak AV, Peter S, van Loo KMJ, Wolking S, Kronenberg-Versteeg D, Weber Y, Schwarz N, Raimondo JV, Melvill R, Tromp SA, Butler JT, Höllig A, Delev D, Wuttke TV, Kampa BM, Koch H. Comprehensive analysis of human dendritic spine morphology and density. J Neurophysiol 2025; 133:1086-1102. [PMID: 40013734 DOI: 10.1152/jn.00622.2024] [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: 01/03/2025] [Revised: 01/14/2025] [Accepted: 02/22/2025] [Indexed: 02/28/2025] Open
Abstract
Dendritic spines, small protrusions on neuronal dendrites, play a crucial role in brain function by changing shape and size in response to neural activity. So far, in-depth analysis of dendritic spines in human brain tissue is lacking. This study presents a comprehensive analysis of human dendritic spine morphology and density using a unique dataset from human brain tissue from 27 patients (8 females, 19 males, aged 18-71 yr) undergoing tumor or epilepsy surgery at three neurosurgery sites. We used acute slices and organotypic brain slice cultures to examine dendritic spines, classifying them into the three main morphological subtypes: mushroom, thin, and stubby, via three-dimensional (3-D) reconstruction using ZEISS arivis Pro software. A deep learning model, trained on 39 diverse datasets, automated spine segmentation and 3-D reconstruction, achieving a 74% F1-score and reducing processing time by over 50%. We show significant differences in spine density by sex, dendrite type, and tissue condition. Females had higher spine densities than males, and apical dendrites were denser in spines than basal ones. Acute tissue showed higher spine densities compared with cultured human brain tissue. With time in culture, mushroom spines decreased, whereas stubby and thin spine percentages increased, particularly from 7-9 to 14 days in vitro, reflecting potential synaptic plasticity changes. Our study underscores the importance of using human brain tissue to understand unique synaptic properties and shows that integrating deep learning with traditional methods enables efficient large-scale analysis, revealing key insights into sex- and tissue-specific dendritic spine dynamics relevant to neurological diseases.NEW & NOTEWORTHY This study presents a dataset of nearly 4,000 morphologically reconstructed human dendritic spines across different ages, gender, and tissue conditions. The dataset was further used to evaluate a deep learning algorithm for three-dimensional spine reconstruction, offering a scalable method for semiautomated spine analysis across various tissues and microscopy setups. The findings enhance understanding of human neurology, indicating potential connections between spine morphology, brain function, and the mechanisms of neurological and psychiatric diseases.
Collapse
Affiliation(s)
- Kerstin D Schünemann
- Department of Epileptology, Neurology, University Hospital RWTH Aachen, Aachen, Germany
| | - Roxanne M Hattingh
- Neuroscience Institute, University of Cape Town, Cape Town,South Africa
- Division of Cell Biology, Department of Human Biology, University of Cape Town, Cape Town, South Africa
| | - Matthijs B Verhoog
- Neuroscience Institute, University of Cape Town, Cape Town,South Africa
- Division of Cell Biology, Department of Human Biology, University of Cape Town, Cape Town, South Africa
| | - Danqing Yang
- Institute of Neuroscience and Medicine 10, Research Center Juelich, Juelich, Germany
| | - Aniella V Bak
- Department of Epileptology, Neurology, University Hospital RWTH Aachen, Aachen, Germany
| | - Sabrina Peter
- Department of Epileptology, Neurology, University Hospital RWTH Aachen, Aachen, Germany
| | - Karen M J van Loo
- Department of Epileptology, Neurology, University Hospital RWTH Aachen, Aachen, Germany
- Department of Neurosurgery, University Hospital RWTH Aachen, Aachen, Germany
| | - Stefan Wolking
- Department of Epileptology, Neurology, University Hospital RWTH Aachen, Aachen, Germany
| | - Deborah Kronenberg-Versteeg
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Yvonne Weber
- Department of Epileptology, Neurology, University Hospital RWTH Aachen, Aachen, Germany
| | - Niklas Schwarz
- Department of Neurology and Epileptology, Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Joseph V Raimondo
- Neuroscience Institute, University of Cape Town, Cape Town,South Africa
- Division of Cell Biology, Department of Human Biology, University of Cape Town, Cape Town, South Africa
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Roger Melvill
- Division of Neurosurgery, Department of Surgery, University of Cape Town, Cape Town, South Africa
| | - Sean A Tromp
- Division of Neurosurgery, Department of Surgery, University of Cape Town, Cape Town, South Africa
| | - James T Butler
- Neuroscience Institute, University of Cape Town, Cape Town,South Africa
- Division of Neurosurgery, Department of Surgery, University of Cape Town, Cape Town, South Africa
| | - Anke Höllig
- Department of Neurosurgery, University Hospital RWTH Aachen, Aachen, Germany
| | - Daniel Delev
- Department of Neurosurgery, University Hospital RWTH Aachen, Aachen, Germany
- Department of Neurosurgery, University Hospital Erlangen, Friedrich-Alexander University Erlangen Nürnberg, Erlangen, Germany
| | - Thomas V Wuttke
- Department of Neurology and Epileptology, Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
- Department of Neurosurgery, University of Tübingen, Tübingen, Germany
| | - Björn M Kampa
- Systems Neurophysiology, Institute of Biology II, RWTH Aachen University, Aachen, Germany
- JARA BRAIN Institute of Neuroscience and Medicine (INM-10), Research Center Juelich, Juelich, Germany
| | - Henner Koch
- Department of Epileptology, Neurology, University Hospital RWTH Aachen, Aachen, Germany
| |
Collapse
|
9
|
Uytiepo M, Zhu Y, Bushong E, Chou K, Polli FS, Zhao E, Kim KY, Luu D, Chang L, Yang D, Ma TC, Kim M, Zhang Y, Walton G, Quach T, Haberl M, Patapoutian L, Shahbazi A, Zhang Y, Beutter E, Zhang W, Dong B, Khoury A, Gu A, McCue E, Stowers L, Ellisman M, Maximov A. Synaptic architecture of a memory engram in the mouse hippocampus. Science 2025; 387:eado8316. [PMID: 40112060 DOI: 10.1126/science.ado8316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 12/17/2024] [Indexed: 03/22/2025]
Abstract
Memory engrams are formed through experience-dependent plasticity of neural circuits, but their detailed architectures remain unresolved. Using three-dimensional electron microscopy, we performed nanoscale reconstructions of the hippocampal CA3-CA1 pathway after chemogenetic labeling of cellular ensembles recruited during associative learning. Neurons with a remote history of activity coinciding with memory acquisition showed no strong preference for wiring with each other. Instead, their connectomes expanded through multisynaptic boutons independently of the coactivation state of postsynaptic partners. The rewiring of ensembles representing an initial engram was accompanied by input-specific, spatially restricted upscaling of individual synapses, as well as remodeling of mitochondria, smooth endoplasmic reticulum, and interactions with astrocytes. Our findings elucidate the physical hallmarks of long-term memory and offer a structural basis for the cellular flexibility of information coding.
Collapse
Affiliation(s)
- Marco Uytiepo
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
- The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA, USA
- The Skaggs Graduate School of Chemical and Biological Sciences, The Scripps Research Institute, La Jolla, CA, USA
| | - Yongchuan Zhu
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
- The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA, USA
| | - Eric Bushong
- National Center for Microscopy and Imaging Research, University of California, San Diego, San Diego, CA, USA
- Department of Neurosciences, University of California, San Diego, School of Medicine, La Jolla, CA, USA
| | - Katherine Chou
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
- The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA, USA
| | - Filip Souza Polli
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
- The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA, USA
| | - Elise Zhao
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
- The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA, USA
| | - Keun-Young Kim
- National Center for Microscopy and Imaging Research, University of California, San Diego, San Diego, CA, USA
- Department of Neurosciences, University of California, San Diego, School of Medicine, La Jolla, CA, USA
| | - Danielle Luu
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
- The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA, USA
| | - Lyanne Chang
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
- The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA, USA
| | - Dong Yang
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
- The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA, USA
| | - Tsz Ching Ma
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
- The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA, USA
| | - Mingi Kim
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
- The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA, USA
- The Skaggs Graduate School of Chemical and Biological Sciences, The Scripps Research Institute, La Jolla, CA, USA
| | - Yuting Zhang
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
- The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA, USA
- The Skaggs Graduate School of Chemical and Biological Sciences, The Scripps Research Institute, La Jolla, CA, USA
| | - Grant Walton
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
- The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA, USA
| | - Tom Quach
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
- The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA, USA
| | - Matthias Haberl
- National Center for Microscopy and Imaging Research, University of California, San Diego, San Diego, CA, USA
- Department of Neurosciences, University of California, San Diego, School of Medicine, La Jolla, CA, USA
| | - Luca Patapoutian
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
- The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA, USA
| | - Arya Shahbazi
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
- The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA, USA
| | - Yuxuan Zhang
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
- The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA, USA
| | - Elizabeth Beutter
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
- The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA, USA
| | - Weiheng Zhang
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
- The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA, USA
| | - Brian Dong
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
- The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA, USA
| | - Aureliano Khoury
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
- The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA, USA
| | - Alton Gu
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
- The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA, USA
| | - Elle McCue
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
- The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA, USA
| | - Lisa Stowers
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
- The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA, USA
| | - Mark Ellisman
- National Center for Microscopy and Imaging Research, University of California, San Diego, San Diego, CA, USA
- Department of Neurosciences, University of California, San Diego, School of Medicine, La Jolla, CA, USA
| | - Anton Maximov
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
- The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA, USA
| |
Collapse
|
10
|
Nemat P, Semenova S, van der Loo RJ, Smit AB, Spijker S, van den Oever MC, Rao-Ruiz P. Structural synaptic signatures of contextual memory retrieval-reactivated hippocampal engram cells. Neurobiol Learn Mem 2025; 218:108033. [PMID: 39923960 DOI: 10.1016/j.nlm.2025.108033] [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: 09/29/2024] [Revised: 01/24/2025] [Accepted: 02/05/2025] [Indexed: 02/11/2025]
Abstract
Learning enhances hippocampal engram cell synaptic connectivity which is crucial for engram reactivation and recall to natural cues. Memory retrieval engages only a subset of the learning-activated ensemble, indicating potential differences in synaptic connectivity signatures of reactivated and non-reactivated cells. We probed these differences in structural synaptic connectivity patterns after recent memory retrieval, 72 h after either neutral Context Exploration (CE) or aversive Contextual Fear Conditioning (CFC). Using a combination of eGRASP (enhanced green fluorescent protein (GFP) reconstitution across synaptic partners) and viral-TRAP (targeted recombination in activated populations) to label CA3 synapses onto CA1 engram cells, we investigated differences in spine density, clusters, and morphology between the reactivated and non-reactivated population of the learning ensemble. In doing so, we developed a pipeline for reconstruction and analysis of dendrites and spines, taking nested data structure into account. Our data demonstrate an interplay between reactivation status, context valence or both factors on the number, distribution, and morphology of CA1 engram cell synapses. Despite a lack of differences in spine density, reactivated engram cells encoding an aversive context were characterised by a higher probability of forming spine clusters and a more dynamic spine type signature compared to their non-reactivated counterparts or engram cells encoding a neutral context. Together, our data indicate that the learning-activated ensemble undergoes different trajectories in structural synaptic connectivity during engram refinement.
Collapse
Affiliation(s)
- Panthea Nemat
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, The Netherlands
| | - Salimat Semenova
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, The Netherlands
| | - Rolinka J van der Loo
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, The Netherlands
| | - August B Smit
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, The Netherlands
| | - Sabine Spijker
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, The Netherlands
| | - Michel C van den Oever
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, The Netherlands.
| | - Priyanka Rao-Ruiz
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, The Netherlands.
| |
Collapse
|
11
|
Caffino L, Targa G, Mottarlini F, Thielens S, Rizzi B, Villers A, Ris L, Gainetdinov RR, Leo D, Fumagalli F. Memantine-induced functional rewiring of the glutamate synapse in the striatum of dopamine transporter knockout rats. Br J Pharmacol 2025; 182:1377-1393. [PMID: 39653030 DOI: 10.1111/bph.17403] [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: 06/11/2024] [Revised: 09/27/2024] [Accepted: 10/29/2024] [Indexed: 02/11/2025] Open
Abstract
BACKGROUND AND PURPOSE Slow-acting biogenic amines, such as dopamine, are known to modulate fast neurotransmitters e.g. glutamate. In the striatum, dopamine (DA) interacts with glutamate, influencing neural excitability and promoting synaptic plasticity. The exact mechanism of such interaction is not fully understood. This study investigates, in detail, how dopamine overactivity in dopamine transporter knockout (DAT-/-) rats, alters the homeostasis of the striatal glutamate synapse from a molecular, behavioural and functional point of view. EXPERIMENTAL APPROACH The expression, localisation, retention and electrophysiological properties of N-methyl-D-aspartate (NMDA) receptors as well as dendritic spine density and morphology were investigated in the striatum of DAT-/- rats, at baseline and after treatment with the non-competitive NMDA receptor antagonist memantine (30 mg kg-1). KEY RESULTS Dopamine overactivity dramatically reorganises the striatal glutamate synapse, redistributing NMDA receptors in the synapse as typified by reduced synaptic availability and reduced expression of NMDA scaffolding proteins, as well as by increased GluN2B-containing NMDA receptors in the extra synapse. Such changes are accompanied by reduced spine density, suggesting dopamine-induced structural rearrangements. These results converge into a compromised plasticity, as shown by the impaired ability to promote long-term depression (LTD) in the striatum of DAT-/-rats. Notably, memantine counteracts hyperlocomotion, reverses spine alterations and abolishes the extrasynaptic movements of NMDA receptors in the striatum of DAT-/- rats, thus restoring functional LTD. CONCLUSION AND IMPLICATIONS A hyperdopaminergic condition seems to alter striatal homeostasis by increasing extrasynaptic NMDA receptors. These findings may be relevant to manipulate disorders characterised by elevated dopaminergic activity.
Collapse
Affiliation(s)
- Lucia Caffino
- Department of Pharmacological and Biomolecular Sciences 'Rodolfo Paoletti', Università degli Studi di Milano, Milan, Italy
| | - Giorgia Targa
- Department of Pharmacological and Biomolecular Sciences 'Rodolfo Paoletti', Università degli Studi di Milano, Milan, Italy
| | - Francesca Mottarlini
- Department of Pharmacological and Biomolecular Sciences 'Rodolfo Paoletti', Università degli Studi di Milano, Milan, Italy
| | - Sarah Thielens
- Department of Neurosciences, University of Mons, Mons, Belgium
| | - Beatrice Rizzi
- Department of Pharmacological and Biomolecular Sciences 'Rodolfo Paoletti', Università degli Studi di Milano, Milan, Italy
- Center for Neuroscience, University of Camerino, Camerino, Italy
| | - Agnes Villers
- Department of Neurosciences, University of Mons, Mons, Belgium
| | - Laurence Ris
- Department of Neurosciences, University of Mons, Mons, Belgium
| | - Raul R Gainetdinov
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
- St. Petersburg University Hospital, St. Petersburg State University, St. Petersburg, Russia
| | - Damiana Leo
- Department of Neurosciences, University of Mons, Mons, Belgium
| | - Fabio Fumagalli
- Department of Pharmacological and Biomolecular Sciences 'Rodolfo Paoletti', Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
12
|
Dickstein DL, Zhang R, Ru N, Vozenin MC, Perry BC, Wang J, Baulch JE, Acharya MM, Limoli CL. Structural plasticity of pyramidal cell neurons measured after FLASH and conventional dose-rate irradiation. Brain Struct Funct 2025; 230:41. [PMID: 40024988 PMCID: PMC11872753 DOI: 10.1007/s00429-025-02902-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 02/17/2025] [Indexed: 03/04/2025]
Abstract
Evidence shows that ultra-high dose-rate FLASH-radiotherapy (FLASH-RT) provides relative protection against normal tissue complications and functional decrements in the irradiated brain. Past work has shown that radiation-induced cognitive impairment, neuroinflammation and reduced structural complexity ofgranule cell neurons were not observed to the same extent after FLASH-RT (> MGy/s) compared to conventional dose-rate (CONV, 0.1 Gy/s) delivery. In this study, we explored the sensitivity of hippocampal CA1 and medial prefrontal cortex (mPFC) pyramidal neurons to cranial irradiation and dose-rate modulation using electron and confocal microscopy. Neuron ultrastructural analyses by electron microscopy after 10 Gy FLASH- or CONV-RT exposures indicated that irradiation had little impact on dendritic complexity and synapse density in the CA1, but did increase the length and head diameter of smaller non-perforated synapses. Similarly, irradiation caused no change in mPFC prelimbic/infralimbic axospinous synapse density, but reductions in non-perforated synapse diameters. While irradiation resulted in thinner myelin sheaths compared to controls, none of these metrics were dose-rate sensitive. Analysis of fluorescently labeled CA1 neurons revealed no radiation-induced or dose-rate-dependent changes in overall dendritic complexity or spine density, in contrast to our past analysis of granule cell neurons. Super-resolution confocal microscopy following a clinical dosing paradigm (3 × 10 Gy) showed significant reductions in excitatory vesicular glutamate transporter 1 and inhibitory vesicular GABA transporter puncta density within the CA1 that were largely dose-rate independent. Collectively, these data reveal that, compared to granule cell neurons, CA1 and mPFC neurons are relatively more radioresistant irrespective of radiation dose-rate.
Collapse
Affiliation(s)
- Dara L Dickstein
- Department of Pathology, Uniformed Services University of Health Sciences, Bethesda, MD, 20814, USA.
- The Henry M. Jackson Foundation for the Advancement of Military Health Inc, Bethesda, MD, USA.
| | - Richard Zhang
- Department of Radiation Oncology, University of California, Irvine School of Medicine, Irvine, CA, USA
| | - Ning Ru
- Department of Radiation Oncology, University of California, Irvine School of Medicine, Irvine, CA, USA
| | - Marie-Catherine Vozenin
- Secteur Radio-Oncologie et Radiobiologie, Hôpitaux Universitaires de Genève, Geneva, Switzerland
| | - Bayley C Perry
- Department of Pathology, Uniformed Services University of Health Sciences, Bethesda, MD, 20814, USA
- The Henry M. Jackson Foundation for the Advancement of Military Health Inc, Bethesda, MD, USA
| | - Juan Wang
- Department of Pathology, Uniformed Services University of Health Sciences, Bethesda, MD, 20814, USA
- The Henry M. Jackson Foundation for the Advancement of Military Health Inc, Bethesda, MD, USA
| | - Janet E Baulch
- Department of Radiation Oncology, University of California, Irvine School of Medicine, Irvine, CA, USA
| | - Munjal M Acharya
- Department of Radiation Oncology, University of California, Irvine School of Medicine, Irvine, CA, USA
- Department of Anatomy and Neurobiology, University of California, Irvine School of Medicine, Irvine, CA, USA
| | - Charles L Limoli
- Department of Radiation Oncology, University of California, Irvine School of Medicine, Irvine, CA, USA.
| |
Collapse
|
13
|
Hoffe B, Kang G, Thomson H, Banton R, Piehler T, Petel OE, Holahan MR. The link between impact-induced tensile strain and dendritic spine morphology in porcine brain tissue. PLoS One 2025; 20:e0318932. [PMID: 39992913 PMCID: PMC11849840 DOI: 10.1371/journal.pone.0318932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 01/24/2025] [Indexed: 02/26/2025] Open
Abstract
Brain tissue as a material presents unique properties with a multitude of cell types and densities, varying degrees of axonal fiber diameters and blood vessels. These neural components are contained within a very viscous environment that upon impact, can result in a variety of tensile, compressive and rotational forces. The depths of the sulcus appear to be particularly vulnerable to biomechanical forces following an impact. The movement and subsequent forces loaded on to the brain have been shown to produce a variety of biomechanical responses that impair neurophysiological functioning at the cellular level. We recently reported a decrease in microtubule associated protein 2 (MAP2) within the depths of the porcine sulcus in an ex vivo model, along with elevated tensile strain in this region within 1 hour after impact. In the current work, using the same impact model, we explored whether changes in spine morphology and density occurred within the same timeframe following impact. The Golgi-Cox method was used to visualize dendritic spine morphology. Cortical pyramidal neurons within the depths and the arms of the sulcus were reconstructed. One hour after impact, there was a change in the distribution of spine type resulting in an increased proportion of mushroom-type spines compared to nonimpacted tissue. The increased proportion of mushroom-type spines was proportional to tensile strain measurements in the apical dendrites. These results demonstrate the sensitivity of dendritic spine morphology to tensile strain within the porcine cortex and suggest a state of hyperexcitability during the hyperacute phase following an impact.
Collapse
Affiliation(s)
- Brendan Hoffe
- Departement of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Gia Kang
- Department of Mechanical and Aerospace Engineering, Carleton University, Ottawa, Ontario, Canada
| | - Hannah Thomson
- Department of Mechanical and Aerospace Engineering, Carleton University, Ottawa, Ontario, Canada
| | - Rohan Banton
- U.S. Army Research Laboratory, Aberdeen Proving Ground, Maryland, United States
| | - Thuvan Piehler
- U.S. Army Research Laboratory, Aberdeen Proving Ground, Maryland, United States
| | - Oren E. Petel
- Department of Mechanical and Aerospace Engineering, Carleton University, Ottawa, Ontario, Canada
| | - Matthew R. Holahan
- Departement of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| |
Collapse
|
14
|
Hanauske T, Koretz CC, Jungenitz T, Roeper J, Drakew A, Deller T. Electrophysiologically calibrated optogenetic stimulation of dentate granule cells mitigates dendritic spine loss in denervated organotypic entorhino-hippocampal slice cultures. Sci Rep 2025; 15:4563. [PMID: 39915664 PMCID: PMC11802742 DOI: 10.1038/s41598-025-88536-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: 02/01/2024] [Accepted: 01/29/2025] [Indexed: 02/09/2025] Open
Abstract
Organotypic slice cultures (OTCs) are versatile tools for studying long-term structure-function relationships of neurons within a defined network (e.g. hippocampus). We developed a method for repeated experimenter-controlled activation of hippocampal granule cells (GCs) in OTCs within the incubator. After several days of contact-free photonic stimulation, we were able to ameliorate entorhinal denervation-induced structural damage in GCs. To achieve this outcome, we had to calibrate the intensity and duration of optogenetic (light) pulses using whole-cell electrophysiological recordings and multi-cell calcium imaging. Our findings showed that ChR2-expressing cells generated action potentials (APs) or calcium transients in response to illumination but were otherwise functionally indistinguishable from non-transduced GCs within the same neural circuit. However, the threshold for AP firing in single GCs varied based on the stimulus light intensity and the expression levels of ChR2. This information allowed us to calibrate light intensity for chronic stimulations. Denervated GCs exhibited significant spine loss four days post-denervation, but this detrimental effect was mitigated when AP firing was induced at a physiological GC bursting rate. Phototoxic damage caused by chronic light exposure was significantly reduced if illuminated with longer wavelength and by adding antioxidants to the culture medium. Our study presents a versatile approach for concurrent non-invasive manipulation and observation of neural circuit activity and remodeling in vitro.
Collapse
Affiliation(s)
- Tijana Hanauske
- Institute for Clinical Neuroanatomy, Dr. Senckenberg Anatomy, Faculty of Medicine, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany.
| | - Carolin Christina Koretz
- Institute for Clinical Neuroanatomy, Dr. Senckenberg Anatomy, Faculty of Medicine, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
- Institute for Neurophysiology, Faculty of Medicine, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Tassilo Jungenitz
- Institute for Clinical Neuroanatomy, Dr. Senckenberg Anatomy, Faculty of Medicine, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Jochen Roeper
- Institute for Neurophysiology, Faculty of Medicine, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Alexander Drakew
- Institute for Clinical Neuroanatomy, Dr. Senckenberg Anatomy, Faculty of Medicine, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Thomas Deller
- Institute for Clinical Neuroanatomy, Dr. Senckenberg Anatomy, Faculty of Medicine, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| |
Collapse
|
15
|
Jenks KR, Cai Y, Nayan ME, Tsimring K, Li K, Zepeda JC, Heller GR, Delepine C, Shih J, Yuan S, Zhu Y, Wang Y, Duan Y, Fu AKY, Ku T, Yun DH, Chung K, Mellios N, Sur M, Ip JPK. The noncoding circular RNA circHomer1 regulates synaptic development and experience-dependent plasticity in mouse visual cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.07.19.603416. [PMID: 39091722 PMCID: PMC11291094 DOI: 10.1101/2024.07.19.603416] [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: 08/04/2024]
Abstract
Circular RNAs (circRNAs) are a class of closed-loop, single stranded RNAs whose expression is particularly enriched in the brain. Despite this enrichment and evidence that the expression of circRNAs are altered by synaptic development and in response to synaptic plasticity in vitro, the regulation by and function of the majority of circRNAs in experience-dependent plasticity in vivo remain unexplored. Here, we employed transcriptome-wide analysis comparing differential expression of both mRNAs and circRNAs in juvenile mouse primary visual cortex (V1) following monocular deprivation (MD), a model of experience-dependent developmental plasticity. Among the differentially expressed mRNAs and circRNAs following 3-day MD, the circular and the activity-dependent mRNA forms of the Homer1 gene, circHomer1 and Homer1a respectively, were of interest as their expression changed in opposite directions: circHomer1 expression increased while the expression of Homer1a decreased following 3-day MD. Knockdown of circHomer1 delayed the depression of closed-eye responses normally observed after 3-day MD. circHomer1-knockdown also led to a reduction in average dendritic spine size prior to MD but critically there was no further reduction after 3-day MD, consistent with impaired structural plasticity. circHomer1-knockdown also prevented the reduction of surface AMPA receptors after 3-day MD. Synapse-localized puncta of the AMPA receptor endocytic protein Arc increased in volume after MD but were smaller in circHomer1-knockdown neurons, suggesting that circHomer1 knockdown impairs experience-dependent AMPA receptor endocytosis. Thus, the expression of multiple circRNAs are regulated by experience-dependent developmental plasticity, and our findings highlight the essential role of circHomer1 in V1 synaptic development and experience-dependent plasticity.
Collapse
Affiliation(s)
- Kyle R. Jenks
- Department of Brain and Cognitive Sciences, The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, USA
- These authors contributed equally
| | - Ying Cai
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
- These authors contributed equally
| | - Marvin Eduarte Nayan
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, USA
- These authors contributed equally
| | - Katya Tsimring
- Department of Brain and Cognitive Sciences, The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, USA
| | - Keji Li
- Department of Brain and Cognitive Sciences, The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, USA
| | - José C. Zepeda
- Department of Brain and Cognitive Sciences, The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, USA
| | - Gregg R. Heller
- Department of Brain and Cognitive Sciences, The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, USA
| | - Chloe Delepine
- Department of Brain and Cognitive Sciences, The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, USA
| | - Jennifer Shih
- Department of Brain and Cognitive Sciences, The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, USA
| | - Shiyang Yuan
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Yao Zhu
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Ye Wang
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Yangyang Duan
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Amy K. Y. Fu
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Taeyun Ku
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Korea
| | - Dae Hee Yun
- Department of Brain and Cognitive Sciences, The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, USA
| | - Kwanghun Chung
- Department of Brain and Cognitive Sciences, The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, USA
| | - Nikolaos Mellios
- Circular Genomics Inc, Albuquerque, New Mexico, 87110, USA
- Previously at: University of New Mexico, Department of Neurosciences, Albuquerque, New Mexico, 87131, USA
| | - Mriganka Sur
- Department of Brain and Cognitive Sciences, The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, USA
| | - Jacque Pak Kan Ip
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
- Gerald Choa Neuroscience Institute, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
16
|
Judák L, Dobos G, Ócsai K, Báthory E, Szebik H, Tarján B, Maák P, Szadai Z, Takács I, Chiovini B, Lőrincz T, Szepesi Á, Roska B, Szalay G, Rózsa B. Moculus: an immersive virtual reality system for mice incorporating stereo vision. Nat Methods 2025; 22:386-398. [PMID: 39668210 PMCID: PMC11810792 DOI: 10.1038/s41592-024-02554-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 10/29/2024] [Indexed: 12/14/2024]
Abstract
Due to technical roadblocks, it is unclear how visual circuits represent multiple features or how behaviorally relevant representations are selected for long-term memory. Here we developed Moculus, a head-mounted virtual reality platform for mice that covers the entire visual field, and allows binocular depth perception and full visual immersion. This controllable environment, with three-dimensional (3D) corridors and 3D objects, in combination with 3D acousto-optical imaging, affords rapid visual learning and the uncovering of circuit substrates in one measurement session. Both the control and reinforcement-associated visual cue coding neuronal assemblies are transiently expanded by reinforcement feedback to near-saturation levels. This increases computational capability and allows competition among assemblies that encode behaviorally relevant information. The coding assemblies form partially orthogonal and overlapping clusters centered around hub cells with higher and earlier ramp-like responses, as well as locally increased functional connectivity.
Collapse
Grants
- ERC 682426 (VISONby3DSTIM), 2018-1.3.1-VKE-00032, 2018-1.1.2-KFI-00097, PM/20453-15/2020, 712821-NEURAM, 2020-1.1.5-GYORSÍTÓSÁV-2021-00004, GINOP-1.2.15-21-2021-00061. 2020-2.1.1-ED-2021-00190, 2020-2.1.1-ED-2022-00208, 2022-1.1.1-KK- 2022-00005, 2022-2.1.1-NL-2022-00012, 2021-1.1.4-GYORSÍTÓSÁV-2022-00064, NUMBER 871277 — AMPLITUDE, GINOP_PLUSZ-2.1.1-21-2022-00143,NKFIH/143650
Collapse
Affiliation(s)
- Linda Judák
- Laboratory of 3D Functional Network and Dendritic Imaging, Institute of Experimental Medicine, Budapest, Hungary
- BrainVisionCenter Research Institute and Competence Center, Budapest, Hungary
| | - Gergely Dobos
- Bay Zoltán Nonprofit for Applied Research, Budapest, Hungary
| | - Katalin Ócsai
- BrainVisionCenter Research Institute and Competence Center, Budapest, Hungary
- Department of Algebra and Geometry, Institute of Mathematics, Budapest University of Technology and Economics, Budapest, Hungary
| | - Eszter Báthory
- BrainVisionCenter Research Institute and Competence Center, Budapest, Hungary
| | - Huba Szebik
- BrainVisionCenter Research Institute and Competence Center, Budapest, Hungary
| | - Balázs Tarján
- BrainVisionCenter Research Institute and Competence Center, Budapest, Hungary
- Doctoral School, Semmelweis University, Budapest, Hungary
| | - Pál Maák
- Department of Atomic Physics, Budapest University of Technology and Economics, Budapest, Hungary
| | - Zoltán Szadai
- Laboratory of 3D Functional Network and Dendritic Imaging, Institute of Experimental Medicine, Budapest, Hungary
- BrainVisionCenter Research Institute and Competence Center, Budapest, Hungary
| | - István Takács
- Laboratory of 3D Functional Network and Dendritic Imaging, Institute of Experimental Medicine, Budapest, Hungary
| | - Balázs Chiovini
- Laboratory of 3D Functional Network and Dendritic Imaging, Institute of Experimental Medicine, Budapest, Hungary
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary
| | - Tibor Lőrincz
- Laboratory of 3D Functional Network and Dendritic Imaging, Institute of Experimental Medicine, Budapest, Hungary
- BrainVisionCenter Research Institute and Competence Center, Budapest, Hungary
| | - Áron Szepesi
- Laboratory of 3D Functional Network and Dendritic Imaging, Institute of Experimental Medicine, Budapest, Hungary
- BrainVisionCenter Research Institute and Competence Center, Budapest, Hungary
| | - Botond Roska
- BrainVisionCenter Research Institute and Competence Center, Budapest, Hungary
- Department of Ophthalmology, University of Basel, Basel, Switzerland
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
| | - Gergely Szalay
- Laboratory of 3D Functional Network and Dendritic Imaging, Institute of Experimental Medicine, Budapest, Hungary
- BrainVisionCenter Research Institute and Competence Center, Budapest, Hungary
| | - Balázs Rózsa
- Laboratory of 3D Functional Network and Dendritic Imaging, Institute of Experimental Medicine, Budapest, Hungary.
- BrainVisionCenter Research Institute and Competence Center, Budapest, Hungary.
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary.
| |
Collapse
|
17
|
Wang X, Wu S, Zuo J, Li K, Chen Y, Fan Z, Wu Z, Yang JX, Song W, Cao JL, Cui M. Selective activation of SIGMAR1 in anterior cingulate cortex glutamatergic neurons facilitates comorbid pain in depression in male mice. Commun Biol 2025; 8:150. [PMID: 39890921 PMCID: PMC11785782 DOI: 10.1038/s42003-025-07590-2] [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: 07/01/2024] [Accepted: 01/21/2025] [Indexed: 02/03/2025] Open
Abstract
Depression and comorbid pain are frequently encountered clinically, and the comorbidity complicates the overall medical management. However, the mechanism whereby depression triggers development of pain needs to be further elucidated. Here, by using the chronic restraint stress (CRS) mouse model of depression and comorbid pain, we showed that CRS hyperactivated the glutamatergic neurons in the anterior cingulate cortex (ACC), as well as increasing the dendrite complexity and number. Chemogenetic activation of these neurons can induce depression and pain, while chemogenetic blockade can reverse such depression-induced pain. Moreover, we utilized translating ribosome affinity purification (TRAP) in combination with c-Fos-tTA strategy and pharmacological approaches and identified SIGMAR1 as a potential therapeutic molecular target. These results revealed a previously unknown neural mechanism for depression and pain comorbidity and provided new mechanistic insights into the antidepressive and analgesic effects of the disease.
Collapse
Affiliation(s)
- Xianlei Wang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Shulin Wu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Junsheng Zuo
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Keying Li
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
- School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Yutong Chen
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Zhijie Fan
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Zhou Wu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Jun-Xia Yang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Weiyi Song
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
- School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Jun-Li Cao
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China.
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China.
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China.
- Department of Anesthesiology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China.
| | - Mengqiao Cui
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China.
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China.
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China.
| |
Collapse
|
18
|
Qiao D, Mu C, Chen H, Wen D, Wang Z, Zhang B, Guo F, Wang C, Zhang R, Wang C, Cui H, Li S. Implications of prenatal exposure to hyperandrogen for hippocampal neurodevelopment and autism-like behavior in offspring. Prog Neuropsychopharmacol Biol Psychiatry 2025; 136:111219. [PMID: 39694316 DOI: 10.1016/j.pnpbp.2024.111219] [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: 01/27/2024] [Revised: 08/24/2024] [Accepted: 12/09/2024] [Indexed: 12/20/2024]
Abstract
Autism spectrum disorder (ASD) is a highly heterogeneous neurodevelopmental disorder that significantly jeopardizes the physical and mental well-being of children. Autism spectrum disorder results from a combination of environmental and genetic factors. Hyperandrogenic exposure during pregnancy increases their risk of developing autism. Nevertheless, the prenatal exposure to androgens affects offspring neurodevelopment and the underlying mechanisms have not been fully elucidated. In the present study, administration of excessive dihydrotestosterone (DHT) to pregnant mice was found to impair neuronal development and dendritic spine formation in offspring, inducing autism-like behaviors. Furthermore, through mRNA transcriptome sequencing technology, the key molecule Nr4a2 was identified during this process of change. Overexpression of Nr4a2 and treatment with amodiaquine (AQ) significantly improved the abnormal phenotypes in offspring caused by prenatal exposure to androgens. Overall, Nr4a2 emerges as a crucial molecule involved in the impairment of offspring neurodevelopment due to prenatal androgen exposure, which provides a new perspective for the in-depth study of the influencing factors and underlying mechanisms.
Collapse
Affiliation(s)
- Dan Qiao
- Department of Human Anatomy; Neuroscience Research Center, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Shijiazhuang 050017, China
| | - Chenyu Mu
- Department of Human Anatomy; Neuroscience Research Center, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Shijiazhuang 050017, China
| | - Huan Chen
- Department of Human Anatomy; Neuroscience Research Center, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Shijiazhuang 050017, China
| | - Di Wen
- College of Forensic Medicine, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Shijiazhuang 050017, China
| | - Zhao Wang
- Department of Human Anatomy; Neuroscience Research Center, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Shijiazhuang 050017, China
| | - Bohan Zhang
- Department of Human Anatomy; Neuroscience Research Center, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Shijiazhuang 050017, China
| | - Fangzhen Guo
- Department of Human Anatomy; Neuroscience Research Center, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Shijiazhuang 050017, China
| | - Chang Wang
- Department of Human Anatomy; Neuroscience Research Center, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Shijiazhuang 050017, China
| | - Rong Zhang
- Autism Research Center; Neuroscience Research Institute, Key Laboratory for Neuroscience, Ministry of Education of China, Key Laboratory for Neuroscience, National Committee of Health and Family Planning of China, Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Chongying Wang
- Autism Research Center, School of Sociology, Nankai University, Tianjin 300071, China
| | - Huixian Cui
- Department of Human Anatomy; Neuroscience Research Center, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Shijiazhuang 050017, China
| | - Sha Li
- Department of Human Anatomy; Neuroscience Research Center, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Shijiazhuang 050017, China; The Key Laboratory of Neural and Vascular Biology of Ministry of Education, Shijiazhuang 050017, China.
| |
Collapse
|
19
|
Matsuki T, Tabata H, Ueda M, Ito H, Nagata KI, Tsuneura Y, Eda S, Kasai K, Nakayama A. The MCPH7 Gene Product STIL Is Essential for Dendritic Spine Formation. Cells 2025; 14:62. [PMID: 39851490 PMCID: PMC11764357 DOI: 10.3390/cells14020062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 12/14/2024] [Accepted: 12/23/2024] [Indexed: 01/26/2025] Open
Abstract
Dendritic spine formation/maintenance is highly dependent on actin cytoskeletal dynamics, which is regulated by small GTPases Rac1 and Cdc42 through their downstream p21-activated kinase/LIM-kinase-I/cofilin pathway. ARHGEF7, also known as ß-PIX, is a guanine nucleotide exchange factor for Rac1 and Cdc42, thereby activating Rac1/Cdc42 and the downstream pathway, leading to the upregulation of spine formation/maintenance. We found that STIL, one of the primary microcephaly gene products, is associated with ARHGEF7 in dendritic spines and that knockdown of Stil resulted in a significant reduction in dendritic spines in neurons both in vitro and in vivo. Rescue experiments indicated that the STIL requirement for spine formation/maintenance depended on its coiled coil domain that mediates the association with ARHGEF7. The overexpression of Rac1/Cdc42 compensated for the spine reduction caused by STIL knockdown. FRET experiments showed that Rac activation is impaired in STIL knockdown neurons. Chemical long-term potentiation, which triggers Rac activation, promoted STIL accumulation in the spine and its association with ARHGEF7. The dynamics of these proteins further supported their coordinated involvement in spine formation/maintenance. Based on these findings, we concluded that the centrosomal protein STIL is a novel regulatory factor essential for spine formation/maintenance by activating Rac and its downstream pathway, possibly through the association with ARHGEF7.
Collapse
Affiliation(s)
- Tohru Matsuki
- Department of Cellular Pathology, Institute for Developmental Research, Aichi Developmental Disability Center, Kasugai 480-0392, Aichi, Japan (S.E.)
| | - Hidenori Tabata
- Department of Molecular Neurobiology, Institute for Developmental Research, Aichi Developmental Disability Center, Kasugai 480-0392, Aichi, Japan; (H.T.); (K.-i.N.)
| | - Masashi Ueda
- Department of Cellular Pathology, Institute for Developmental Research, Aichi Developmental Disability Center, Kasugai 480-0392, Aichi, Japan (S.E.)
| | - Hideaki Ito
- Department of Pathology, Aichi Medical University School of Medicine, Nagakute 480-1195, Aichi, Japan (K.K.)
| | - Koh-ichi Nagata
- Department of Molecular Neurobiology, Institute for Developmental Research, Aichi Developmental Disability Center, Kasugai 480-0392, Aichi, Japan; (H.T.); (K.-i.N.)
- Department of Neurochemistry, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Aichi, Japan
| | - Yumi Tsuneura
- Department of Cellular Pathology, Institute for Developmental Research, Aichi Developmental Disability Center, Kasugai 480-0392, Aichi, Japan (S.E.)
| | - Shima Eda
- Department of Cellular Pathology, Institute for Developmental Research, Aichi Developmental Disability Center, Kasugai 480-0392, Aichi, Japan (S.E.)
| | - Kenji Kasai
- Department of Pathology, Aichi Medical University School of Medicine, Nagakute 480-1195, Aichi, Japan (K.K.)
| | - Atsuo Nakayama
- Department of Cellular Pathology, Institute for Developmental Research, Aichi Developmental Disability Center, Kasugai 480-0392, Aichi, Japan (S.E.)
- Department of Neurochemistry, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Aichi, Japan
| |
Collapse
|
20
|
Pereira-Iglesias M, Maldonado-Teixido J, Melero A, Piriz J, Galea E, Ransohoff RM, Sierra A. Microglia as hunters or gatherers of brain synapses. Nat Neurosci 2025; 28:15-23. [PMID: 39663381 DOI: 10.1038/s41593-024-01818-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 10/02/2024] [Indexed: 12/13/2024]
Abstract
Over a decade ago, it was discovered that microglia, the brain's immune cells, engulf synaptic material in a process named microglial pruning. This term suggests that microglia actively sculpt brain circuits by tagging and phagocytosing unwanted synapses. However, live imaging studies have yet to demonstrate how microglial synapse elimination occurs. To address this issue, we propose a new conceptual framework distinguishing between two potential mechanisms of synapse elimination, culling and scavenging. During culling, microglia may use a contractile ring to sever the neuronal plasma membrane, removing the unwanted synapse. During scavenging, synapse elimination is neuronal-driven, and the neuronal plasma membrane fission machinery sheds off synapses that are later phagocytosed by microglia. We will discuss the current limitations of studying microglial synapse elimination and evaluate evidence supporting either culling or scavenging. Discerning between these mechanisms is essential for determining the therapeutic value of phagocytosis modulators in diseases with altered brain connectivity.
Collapse
Affiliation(s)
- Marta Pereira-Iglesias
- Achucarro Basque Center for Neuroscience, Leioa, Spain
- Department of Neuroscience, University of the Basque Country EHU/UPV, Leioa, Spain
| | - Joel Maldonado-Teixido
- Achucarro Basque Center for Neuroscience, Leioa, Spain
- Department of Pharmacology, University of the Basque Country EHU/UPV, Leioa, Spain
| | | | - Joaquin Piriz
- Achucarro Basque Center for Neuroscience, Leioa, Spain
- Ikerbasque Foundation, Bilbao, Spain
| | - Elena Galea
- Institut de Neurociències and Departament de Bioquímica i Biologia Molecular, Unitat de Bioquímica de Medicina, Universitat Autònoma de Barcelona UAB, Barcelona, Spain
- ICREA, Barcelona, Spain
| | | | - Amanda Sierra
- Achucarro Basque Center for Neuroscience, Leioa, Spain.
- Ikerbasque Foundation, Bilbao, Spain.
- Department of Biochemistry and Molecular Biology, University of the Basque Country EHU/UPV, Leioa, Spain.
| |
Collapse
|
21
|
Yan HH, He JJ, Fu C, Chen JH, Tang AH. ATAD1 Regulates Neuronal Development and Synapse Formation Through Tuning Mitochondrial Function. Int J Mol Sci 2024; 26:44. [PMID: 39795902 PMCID: PMC11719905 DOI: 10.3390/ijms26010044] [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: 11/04/2024] [Revised: 12/20/2024] [Accepted: 12/21/2024] [Indexed: 01/13/2025] Open
Abstract
Mitochondrial function is essential for synaptic function. ATAD1, an AAA+ protease involved in mitochondrial quality control, governs fission-fusion dynamics within the organelle. However, the distribution and functional role of ATAD1 in neurons remain poorly understood. In this study, we demonstrate that ATAD1 is primarily localized to mitochondria in dendrites and, to a lesser extent, in spines in cultured hippocampal neurons. We found that ATAD1 deficiency disrupts the mitochondrial fission-fusion balance, resulting in mitochondrial fragmentation. This deficiency also impairs dendritic branching, hinders dendritic spine maturation, and reduces glutamatergic synaptic transmission in hippocampal neuron. To further investigate the underlying mechanism, we employed an ATP hydrolysis-deficient mutant of ATAD1 to rescue the neuronal deficits associated with ATAD1 loss. We discovered that the synaptic deficits are independent of the mitochondrial morphology changes but rely on its ATP hydrolysis. Furthermore, we show that ATAD1 loss leads to impaired mitochondrial function, including decreased ATP production, impaired membrane potential, and elevated oxidative stress. In conclusion, our results provide evidence that ATAD1 is crucial for maintaining mitochondrial function and regulating neurodevelopment and synaptic function.
Collapse
Affiliation(s)
- Hao-Hao Yan
- Hefei National Laboratory for Physical Sciences at the Microscale, MOE Key Laboratory for Membrane-Less Organelles & Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (H.-H.Y.); (J.-J.H.); (C.F.)
- Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230088, China
- Neurodegenerative Disorder Research Center and Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei 230027, China
| | - Jia-Jia He
- Hefei National Laboratory for Physical Sciences at the Microscale, MOE Key Laboratory for Membrane-Less Organelles & Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (H.-H.Y.); (J.-J.H.); (C.F.)
| | - Chuanhai Fu
- Hefei National Laboratory for Physical Sciences at the Microscale, MOE Key Laboratory for Membrane-Less Organelles & Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (H.-H.Y.); (J.-J.H.); (C.F.)
| | - Jia-Hui Chen
- Department of Anatomy, School of Basic Medicine, Anhui Medical University, Hefei 230032, China
| | - Ai-Hui Tang
- Hefei National Laboratory for Physical Sciences at the Microscale, MOE Key Laboratory for Membrane-Less Organelles & Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (H.-H.Y.); (J.-J.H.); (C.F.)
- Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230088, China
- Neurodegenerative Disorder Research Center and Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei 230027, China
| |
Collapse
|
22
|
Jiang W, Zhang J, Wang M, Zou Y, Liu Q, Song Y, Sun G, Gong Y, Zhang F, Jiang B. The X-linked intellectual disability gene CUL4B is critical for memory and synaptic function. Acta Neuropathol Commun 2024; 12:188. [PMID: 39633474 PMCID: PMC11619648 DOI: 10.1186/s40478-024-01903-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] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 11/25/2024] [Indexed: 12/07/2024] Open
Abstract
Cullin 4B (CUL4B) is the scaffold protein in the CUL4B-RING E3 ubiquitin ligase (CRL4B) complex. Loss-of-function mutations in the human CUL4B gene lead to syndromic X-linked intellectual disability (XLID). Till now, the mechanism of intellectual disability caused by CUL4B mutation still needs to be elucidated. In this study, we used single-nucleus RNA sequencing (snRNA-seq) to investigate the impact of CUL4B deficiency on the transcriptional programs of diverse cell types. The results revealed that depletion of CUL4B resulted in impaired intercellular communication and elicited cell type-specific transcriptional changes relevant to synapse dysfunction. Golgi-Cox staining of brain slices and immunostaining of in vitro cultured neurons revealed remarkable synapse loss in CUL4B-deficient mice. Ultrastructural analysis via transmission electron microscopy (TEM) showed that the width of the synaptic cleft was significantly greater in CUL4B-deficient mice. Electrophysiological experiments found a decrease in the amplitude of AMPA receptor-mediated EPSCs in the hippocampal CA1 pyramidal neurons of CUL4B-deficient mice. These results indicate that depletion of CUL4B in mice results in morphological and functional abnormalities in synapses. Furthermore, behavioral tests revealed that depletion of CUL4B in the mouse nervous system results in impaired spatial learning and memory. Taken together, the findings of this study reveal the pathogenesis of neurological disorders associated with CUL4B mutations and promote the identification of therapeutic targets that can halt synaptic abnormalities and preserve memory in individuals.
Collapse
Affiliation(s)
- Wei Jiang
- The Key Laboratory of Experimental Teratology of the Ministry of Education and Department of Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Jian Zhang
- The Key Laboratory of Experimental Teratology of the Ministry of Education and Department of Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Molin Wang
- The Key Laboratory of Experimental Teratology of the Ministry of Education and Department of Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yongxin Zou
- The Key Laboratory of Experimental Teratology of the Ministry of Education and Department of Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Qiao Liu
- The Key Laboratory of Experimental Teratology of the Ministry of Education and Department of Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yu Song
- The Key Laboratory of Experimental Teratology of the Ministry of Education and Department of Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Gongping Sun
- The Key Laboratory of Experimental Teratology of the Ministry of Education, Department of Histology and Embryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yaoqin Gong
- The Key Laboratory of Experimental Teratology of the Ministry of Education and Department of Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Fan Zhang
- Medical Morphology Teaching Laboratory, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.
| | - Baichun Jiang
- The Key Laboratory of Experimental Teratology of the Ministry of Education and Department of Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.
| |
Collapse
|
23
|
Kim N, Yun H, Lee H, Yoo JY. Interplay between membranes and biomolecular condensates in the regulation of membrane-associated cellular processes. Exp Mol Med 2024; 56:2357-2364. [PMID: 39482532 PMCID: PMC11612285 DOI: 10.1038/s12276-024-01337-5] [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: 02/19/2024] [Revised: 08/04/2024] [Accepted: 08/11/2024] [Indexed: 11/03/2024] Open
Abstract
Liquid‒liquid phase separation (LLPS) has emerged as a key mechanism for organizing cellular spaces independent of membranes. Biomolecular condensates, which assemble through LLPS, exhibit distinctive liquid droplet-like behavior and can exchange constituents with their surroundings. The regulation of condensate phases, including transitions from a liquid state to gel or irreversible aggregates, is important for their physiological functions and for controlling pathological progression, as observed in neurodegenerative diseases and cancer. While early studies on biomolecular condensates focused primarily on those in fluidic environments such as the cytosol, recent discoveries have revealed their existence in close proximity to, on, or even comprising membranes. The aim of this review is to provide an overview of the properties of membrane-associated condensates in a cellular context and their biological functions in relation to membranes.
Collapse
Affiliation(s)
- Nari Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea.
| | - Hyeri Yun
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Hojin Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Joo-Yeon Yoo
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea.
| |
Collapse
|
24
|
Brill J, Linden DJ. Chronic Aromatase Inhibition Attenuates Synaptic Plasticity in Ovariectomized Mice. eNeuro 2024; 11:ENEURO.0346-24.2024. [PMID: 39592220 PMCID: PMC11594935 DOI: 10.1523/eneuro.0346-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 10/11/2024] [Accepted: 10/23/2024] [Indexed: 11/28/2024] Open
Abstract
Brain-derived estrogen (17β-estradiol, E2) is a neuromodulator that plays important roles in neural plasticity and network excitability. Chronic inhibition of estrogen synthesis is used in adjuvant breast cancer therapy for estrogen receptor-positive tumors and may have been associated with cognitive and affective side effects. Here, we have developed a model of adjuvant therapy in female ovariectomized mice in which the E2 biosynthetic enzyme aromatase is inhibited by letrozole (1 mg/kg/day, i.p., for up to 3 weeks), Using two-photon longitudinal in vivo imaging in Thy1-GFP-M mice, we found that spine density in the apical dendrites of neocortical layer 5 pyramidal cells was unaffected by letrozole treatment but spine turnover was reduced. LTP in layer 4 to layer 2/3 synapses in the somatosensory cortex was also reduced in slices from letrozole-treated mice, showing deficits in structural and functional plasticity resulting from aromatase inhibition. Ovariectomized mice performed worse than intact control mice in the novel object recognition test but, surprisingly, letrozole treatment rescued this deficit.
Collapse
Affiliation(s)
- Julia Brill
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, Maryland 21210
| | - David J Linden
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, Maryland 21210
- Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, Maryland 21210
| |
Collapse
|
25
|
Bharmauria V, Ramezanpour H, Ouelhazi A, Yahia Belkacemi Y, Flouty O, Molotchnikoff S. KETAMINE: Neural- and network-level changes. Neuroscience 2024; 559:188-198. [PMID: 39245312 DOI: 10.1016/j.neuroscience.2024.09.010] [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: 07/08/2024] [Revised: 08/30/2024] [Accepted: 09/03/2024] [Indexed: 09/10/2024]
Abstract
Ketamine is a widely used clinical drug that has several functional and clinical applications, including its use as an anaesthetic, analgesic, anti-depressive, anti-suicidal agent, among others. Among its diverse behavioral effects, it influences short-term memory and induces psychedelic effects. At the neural level across different brain areas, it modulates neural firing rates, neural tuning, brain oscillations, and modularity, while promoting hypersynchrony and random connectivity between neurons. In our recent studies we demonstrated that topical application of ketamine on the visual cortex alters neural tuning and promotes vigorous connectivity between neurons by decreasing their firing variability. Here, we begin with a brief review of the literature, followed by results from our lab, where we synthesize a dendritic model of neural tuning and network changes following ketamine application. This model has potential implications for focused modulation of cortical networks in clinical settings. Finally, we identify current gaps in research and suggest directions for future studies, particularly emphasizing the need for more animal experiments to establish a platform for effective translation and synergistic therapies combining ketamine with other protocols such as training and adaptation. In summary, investigating ketamine's broader systemic effects, not only provides deeper insight into cognitive functions and consciousness but also paves the way to advance therapies for neuropsychiatric disorders.
Collapse
Affiliation(s)
- Vishal Bharmauria
- The Tampa Human Neurophysiology Lab & Department of Neurosurgery and Brain Repair, Morsani College of Medicine, 2 Tampa General Circle, University of South Florida, Tampa, FL 33606, USA; Centre for Vision Research and Centre for Integrative and Applied Neuroscience, York University, 4700 Keele Street, Toronto, Ontario M3J 1P3, Canada.
| | - Hamidreza Ramezanpour
- Department of Biology, York University, 4700 Keele Street, Toronto, Ontario M3J 1P3, Canada
| | - Afef Ouelhazi
- Neurophysiology of the Visual system, Département de Sciences Biologiques, 1375 Av. Thérèse-Lavoie-Roux, Université de Montréal, Montréal, Québec H2V 0B3, Canada
| | - Yassine Yahia Belkacemi
- Neurophysiology of the Visual system, Département de Sciences Biologiques, 1375 Av. Thérèse-Lavoie-Roux, Université de Montréal, Montréal, Québec H2V 0B3, Canada
| | - Oliver Flouty
- The Tampa Human Neurophysiology Lab & Department of Neurosurgery and Brain Repair, Morsani College of Medicine, 2 Tampa General Circle, University of South Florida, Tampa, FL 33606, USA
| | - Stéphane Molotchnikoff
- Neurophysiology of the Visual system, Département de Sciences Biologiques, 1375 Av. Thérèse-Lavoie-Roux, Université de Montréal, Montréal, Québec H2V 0B3, Canada
| |
Collapse
|
26
|
Lin B, Jin Z, Park G, Ge Q, Singh K, Ryan V WG, Imami AS, Naghavi F, Miller OA, Khan S, Lu H, McCullumsmith RE, Du J. Mice lacking acid-sensing ion channel 2 in the medial prefrontal cortex exhibit social dominance. SCIENCE ADVANCES 2024; 10:eadn7573. [PMID: 39453995 PMCID: PMC11506137 DOI: 10.1126/sciadv.adn7573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 09/23/2024] [Indexed: 10/27/2024]
Abstract
Social dominance is essential for maintaining a stable society and has both positive and negative impacts on social animals, including humans. However, the regulatory mechanisms governing social dominance, as well as the crucial regulators and biomarkers involved, remain poorly understood. We discover that mice lacking acid-sensing ion channel 2 (ASIC2) exhibit persistently higher social dominance than their wild-type cagemates. Conversely, overexpression of ASIC2 in the medial prefrontal cortex reverses the dominance hierarchy observed in ASIC2 knockout (Asic2-/-) mice. Asic2-/- neurons exhibit increased synaptic transmission and plasticity, potentially mediated by protein kinase A signaling pathway. Furthermore, ASIC2 plays distinct functional roles in excitatory and inhibitory neurons, thereby modulating the balance of neuronal activities underlying social dominance behaviors-a phenomenon suggestive of a cell subtype-specific mechanism. This research lays the groundwork for understanding the mechanisms of social dominance, offering potential insights for managing social disorders, such as depression and anxiety.
Collapse
Affiliation(s)
- Boren Lin
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Department of Biological Sciences, University of Toledo, Toledo, OH 43606, USA
| | - Zhen Jin
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Gyeongah Park
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Qian Ge
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Kritika Singh
- Department of Biological Sciences, University of Toledo, Toledo, OH 43606, USA
| | - William G. Ryan V
- Department of Neuroscience, University of Toledo, Toledo, OH 43606, USA
| | - Ali Sajid Imami
- Department of Neuroscience, University of Toledo, Toledo, OH 43606, USA
| | - Farzaneh Naghavi
- Department of Neuroscience, University of Toledo, Toledo, OH 43606, USA
| | - Olivia Ann Miller
- Department of Biological Sciences, University of Toledo, Toledo, OH 43606, USA
| | - Saira Khan
- Department of Biological Sciences, University of Toledo, Toledo, OH 43606, USA
| | - Hui Lu
- Department of Pharmacology and Physiology, George Washington University School of Medicine, Washington, DC 20037, USA
| | - Robert E. McCullumsmith
- Department of Neuroscience, University of Toledo, Toledo, OH 43606, USA
- Neurosciences Institute, ProMedica, Toledo, OH 43614, USA
| | - Jianyang Du
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| |
Collapse
|
27
|
Bauer J, Lewin U, Herbert E, Gjorgjieva J, Schoonover CE, Fink AJP, Rose T, Bonhoeffer T, Hübener M. Sensory experience steers representational drift in mouse visual cortex. Nat Commun 2024; 15:9153. [PMID: 39443498 PMCID: PMC11499870 DOI: 10.1038/s41467-024-53326-x] [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: 12/20/2023] [Accepted: 10/08/2024] [Indexed: 10/25/2024] Open
Abstract
Representational drift-the gradual continuous change of neuronal representations-has been observed across many brain areas. It is unclear whether drift is caused by synaptic plasticity elicited by sensory experience, or by the intrinsic volatility of synapses. Here, using chronic two-photon calcium imaging in primary visual cortex of female mice, we find that the preferred stimulus orientation of individual neurons slowly drifts over the course of weeks. By using cylinder lens goggles to limit visual experience to a narrow range of orientations, we show that the direction of drift, but not its magnitude, is biased by the statistics of visual input. A network model suggests that drift of preferred orientation largely results from synaptic volatility, which under normal visual conditions is counteracted by experience-driven Hebbian mechanisms, stabilizing preferred orientation. Under deprivation conditions these Hebbian mechanisms enable adaptation. Thus, Hebbian synaptic plasticity steers drift to match the statistics of the environment.
Collapse
Affiliation(s)
- Joel Bauer
- Max Planck Institute for Biological Intelligence, Martinsried, Germany.
- International Max Planck Research School for Molecular Life Sciences, Martinsried, Germany.
- Sainsbury Wellcome Centre for Neural Circuits and Behaviour, University College London, London, UK.
| | - Uwe Lewin
- Max Planck Institute for Biological Intelligence, Martinsried, Germany
- Graduate School of Systemic Neurosciences, Ludwig-Maximilians-Universität München, Planegg, Germany
| | - Elizabeth Herbert
- School of Life Sciences, Technical University of Munich, Freising, Germany
| | | | - Carl E Schoonover
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Department of Neuroscience, Columbia University, New York, NY, USA
- Allen Institute for Neural Dynamics, Seattle, WA, USA
| | - Andrew J P Fink
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Department of Neuroscience, Columbia University, New York, NY, USA
- Department of Neurobiology, Northwestern University, Evanston, IL, USA
| | - Tobias Rose
- Max Planck Institute for Biological Intelligence, Martinsried, Germany
- Institute for Experimental Epileptology and Cognition Research, University of Bonn, Medical Center, Bonn, Germany
| | - Tobias Bonhoeffer
- Max Planck Institute for Biological Intelligence, Martinsried, Germany
| | - Mark Hübener
- Max Planck Institute for Biological Intelligence, Martinsried, Germany.
| |
Collapse
|
28
|
Barrantes FJ. Cognitive synaptopathy: synaptic and dendritic spine dysfunction in age-related cognitive disorders. Front Aging Neurosci 2024; 16:1476909. [PMID: 39420927 PMCID: PMC11484076 DOI: 10.3389/fnagi.2024.1476909] [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: 08/06/2024] [Accepted: 09/20/2024] [Indexed: 10/19/2024] Open
Abstract
Cognitive impairment is a leading component of several neurodegenerative and neurodevelopmental diseases, profoundly impacting on the individual, the family, and society at large. Cognitive pathologies are driven by a multiplicity of factors, from genetic mutations and genetic risk factors, neurotransmitter-associated dysfunction, abnormal connectomics at the level of local neuronal circuits and broader brain networks, to environmental influences able to modulate some of the endogenous factors. Otherwise healthy older adults can be expected to experience some degree of mild cognitive impairment, some of which fall into the category of subjective cognitive deficits in clinical practice, while many neurodevelopmental and neurodegenerative diseases course with more profound alterations of cognition, particularly within the spectrum of the dementias. Our knowledge of the underlying neuropathological mechanisms at the root of this ample palette of clinical entities is far from complete. This review looks at current knowledge on synaptic modifications in the context of cognitive function along healthy ageing and cognitive dysfunction in disease, providing insight into differential diagnostic elements in the wide range of synapse alterations, from those associated with the mild cognitive changes of physiological senescence to the more profound abnormalities occurring at advanced clinical stages of dementia. I propose the term "cognitive synaptopathy" to encompass the wide spectrum of synaptic pathologies associated with higher brain function disorders.
Collapse
Affiliation(s)
- Francisco J. Barrantes
- Laboratory of Molecular Neurobiology, Biomedical Research Institute, Pontifical Catholic University of Argentina (UCA), Argentine Scientific and Technological Research Council (CONICET), Buenos Aires, Argentina
| |
Collapse
|
29
|
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
|
30
|
Silva RH, Pedro LC, Manosso LM, Gonçalves CL, Réus GZ. Pre- and Post-Synaptic protein in the major depressive Disorder: From neurobiology to therapeutic targets. Neuroscience 2024; 556:14-24. [PMID: 39103041 DOI: 10.1016/j.neuroscience.2024.07.050] [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: 04/17/2024] [Revised: 07/22/2024] [Accepted: 07/31/2024] [Indexed: 08/07/2024]
Abstract
Major depressive disorder (MDD) has demonstrated its negative impact on various aspects of the lives of those affected. Although several therapies have been developed over the years, it remains a challenge for mental health professionals. Thus, understanding the pathophysiology of MDD is necessary to improve existing treatment options or seek new therapeutic alternatives. Clinical and preclinical studies in animal models of depression have shown the involvement of synaptic plasticity in both the development of MDD and the response to available drugs. However, synaptic plasticity involves a cascade of events, including the action of presynaptic proteins such as synaptophysin and synapsins and postsynaptic proteins such as postsynaptic density-95 (PSD-95). Additionally, several factors can negatively impact the process of spinogenesis/neurogenesis, which are related to many outcomes, including MDD. Thus, this narrative review aims to deepen the understanding of the involvement of synaptic formations and their components in the pathophysiology and treatment of MDD.
Collapse
Affiliation(s)
- Ritele H Silva
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil; Department of Health Sciences, Campus Araranguá, Federal University of Santa Catarina, 88906-072 Araranguá, SC, Brazil
| | - Lucas C Pedro
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Luana M Manosso
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Cinara L Gonçalves
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Gislaine Z Réus
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil.
| |
Collapse
|
31
|
Li DC, Hinton EA, Guo J, Knight KA, Sequeira MK, Wynne ME, Dighe NM, Gourley SL. Social experience in adolescence shapes prefrontal cortex structure and function in adulthood. Mol Psychiatry 2024; 29:2787-2798. [PMID: 38580810 PMCID: PMC11567502 DOI: 10.1038/s41380-024-02540-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 03/15/2024] [Accepted: 03/18/2024] [Indexed: 04/07/2024]
Abstract
During adolescence, the prefrontal cortex (PFC) undergoes dramatic reorganization. PFC development is profoundly influenced by the social environment, disruptions to which may prime the emergence of psychopathology across the lifespan. We investigated the neurobehavioral consequences of isolation experienced in adolescence in mice, and in particular, the long-term consequences that were detectable even despite normalization of the social milieu. Isolation produced biases toward habit-like behavior at the expense of flexible goal seeking, plus anhedonic-like reward deficits. Behavioral phenomena were accompanied by neuronal dendritic spine over-abundance and hyper-excitability in the ventromedial PFC (vmPFC), which was necessary for the expression of isolation-induced habits and sufficient to trigger behavioral inflexibility in socially reared controls. Isolation activated cytoskeletal regulatory pathways otherwise suppressed during adolescence, such that repression of constituent elements prevented long-term isolation-induced neurosequelae. Altogether, our findings unveil an adolescent critical period and multi-model mechanism by which social experiences facilitate prefrontal cortical maturation.
Collapse
Affiliation(s)
- Dan C Li
- Medical Scientist Training Program, Emory University School of Medicine, Atlanta, GA, USA.
- Emory National Primate Research Center, Emory University, Atlanta, GA, USA.
| | - Elizabeth A Hinton
- Emory National Primate Research Center, Emory University, Atlanta, GA, USA
- Department of Pediatrics, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
| | - Jidong Guo
- Emory National Primate Research Center, Emory University, Atlanta, GA, USA
- Department of Psychiatry and Behavioral sciences, Emory University School of Medicine, Atlanta, GA, USA
| | | | - Michelle K Sequeira
- Emory National Primate Research Center, Emory University, Atlanta, GA, USA
- Department of Pediatrics, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
| | - Meghan E Wynne
- Emory National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Niharika M Dighe
- Emory National Primate Research Center, Emory University, Atlanta, GA, USA
- Department of Pediatrics, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
| | - Shannon L Gourley
- Emory National Primate Research Center, Emory University, Atlanta, GA, USA.
- Department of Pediatrics, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA.
- Department of Psychiatry and Behavioral sciences, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
32
|
Arshavsky YI. Autoimmune hypothesis of Alzheimer's disease: unanswered question. J Neurophysiol 2024; 132:929-942. [PMID: 39163023 DOI: 10.1152/jn.00259.2024] [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: 06/18/2024] [Revised: 07/24/2024] [Accepted: 07/25/2024] [Indexed: 08/21/2024] Open
Abstract
Alzheimer's disease (AD) was described more than a century ago. However, there are still no effective approaches to its treatment, which may suggest that the search for the cure is not being conducted in the most productive direction. AD begins as selective impairments of declarative memory with no deficits in other cognitive functions. Therefore, understanding of the AD pathogenesis has to include the understanding of this selectivity. Currently, the main efforts aimed at prevention and treatment of AD are based on the dominating hypothesis for the AD pathogenesis: the amyloid hypothesis. But this hypothesis does not explain selective memory impairments since β-amyloid accumulates extracellularly and should be toxic to all types of cerebral neurons, not only to "memory engram neurons." To explain selective memory impairment, I propose the autoimmune hypothesis of AD, based on the analysis of risk factors for AD and molecular mechanisms of memory formation. Memory formation is associated with epigenetic modifications of chromatin in memory engram neurons and, therefore, might be accompanied by the expression of memory-specific proteins recognized by the adaptive immune system as "non-self" antigens. Normally, the brain is protected by the blood-brain barrier (BBB). All risk factors for AD provoke BBB disruptions, possibly leading to an autoimmune reaction against memory engram neurons. This reaction would make them selectively sensitive to tauopathy. If this hypothesis is confirmed, the strategies for AD prevention and treatment would be radically changed.
Collapse
Affiliation(s)
- Yuri I Arshavsky
- BioCircuits Institute, University of California, San Diego, La Jolla, California, United States
| |
Collapse
|
33
|
Onimus O, Arrivet F, Souza INDO, Bertrand B, Castel J, Luquet S, Mothet JP, Heck N, Gangarossa G. The gut-brain vagal axis scales hippocampal memory processes and plasticity. Neurobiol Dis 2024; 199:106569. [PMID: 38885849 DOI: 10.1016/j.nbd.2024.106569] [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: 04/28/2024] [Revised: 06/05/2024] [Accepted: 06/13/2024] [Indexed: 06/20/2024] Open
Abstract
The vagus nerve serves as an interoceptive relay between the body and the brain. Despite its well-established role in feeding behaviors, energy metabolism, and cognitive functions, the intricate functional processes linking the vagus nerve to the hippocampus and its contribution to learning and memory dynamics remain still elusive. Here, we investigated whether and how the gut-brain vagal axis contributes to hippocampal learning and memory processes at behavioral, functional, cellular, and molecular levels. Our results indicate that the integrity of the vagal axis is essential for long-term recognition memories, while sparing other forms of memory. In addition, by combing multi-scale approaches, our findings show that the gut-brain vagal tone exerts a permissive role in scaling intracellular signaling events, gene expressions, hippocampal dendritic spines density as well as functional long-term plasticities (LTD and LTP). These results highlight the critical role of the gut-brain vagal axis in maintaining the spontaneous and homeostatic functions of hippocampal ensembles and in regulating their learning and memory functions. In conclusion, our study provides comprehensive insights into the multifaceted involvement of the gut-brain vagal axis in shaping time-dependent hippocampal learning and memory dynamics. Understanding the mechanisms underlying this interoceptive body-brain neuronal communication may pave the way for novel therapeutic approaches in conditions associated with cognitive decline, including neurodegenerative disorders.
Collapse
Affiliation(s)
- Oriane Onimus
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, F-75013 Paris, France
| | - Faustine Arrivet
- Sorbonne Université, CNRS, INSERM, Neurosciences Paris Seine, Institut de Biologie Paris Seine, F-75005 Paris, France
| | - Isis Nem de Oliveira Souza
- Biophotonics and Synapse Physiopathology Team, Laboratoire LuMIn UMR9024 Université Paris-Saclay, ENS Paris-Saclay, CNRS, CentraleSupelec, 91190 Gif-sur-Yvette, France; Laboratory of Molecular Pharmacology, Institute of Biomedical Sciences, Universidade Federal do Rio de Janeiro, Brazil
| | - Benoit Bertrand
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, F-75013 Paris, France
| | - Julien Castel
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, F-75013 Paris, France
| | - Serge Luquet
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, F-75013 Paris, France
| | - Jean-Pierre Mothet
- Biophotonics and Synapse Physiopathology Team, Laboratoire LuMIn UMR9024 Université Paris-Saclay, ENS Paris-Saclay, CNRS, CentraleSupelec, 91190 Gif-sur-Yvette, France
| | - Nicolas Heck
- Sorbonne Université, CNRS, INSERM, Neurosciences Paris Seine, Institut de Biologie Paris Seine, F-75005 Paris, France
| | - Giuseppe Gangarossa
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, F-75013 Paris, France; Institut Universitaire de France, France.
| |
Collapse
|
34
|
Yaeger CE, Vardalaki D, Zhang Q, Pham TLD, Brown NJ, Ji N, Harnett MT. A dendritic mechanism for balancing synaptic flexibility and stability. Cell Rep 2024; 43:114638. [PMID: 39167486 PMCID: PMC11403626 DOI: 10.1016/j.celrep.2024.114638] [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: 04/16/2024] [Revised: 06/28/2024] [Accepted: 07/30/2024] [Indexed: 08/23/2024] Open
Abstract
Biological and artificial neural networks learn by modifying synaptic weights, but it is unclear how these systems retain previous knowledge and also acquire new information. Here, we show that cortical pyramidal neurons can solve this plasticity-versus-stability dilemma by differentially regulating synaptic plasticity at distinct dendritic compartments. Oblique dendrites of adult mouse layer 5 cortical pyramidal neurons selectively receive monosynaptic thalamic input, integrate linearly, and lack burst-timing synaptic potentiation. In contrast, basal dendrites, which do not receive thalamic input, exhibit conventional NMDA receptor (NMDAR)-mediated supralinear integration and synaptic potentiation. Congruently, spiny synapses on oblique branches show decreased structural plasticity in vivo. The selective decline in NMDAR activity and expression at synapses on oblique dendrites is controlled by a critical period of visual experience. Our results demonstrate a biological mechanism for how single neurons can safeguard a set of inputs from ongoing plasticity by altering synaptic properties at distinct dendritic domains.
Collapse
Affiliation(s)
- Courtney E Yaeger
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Dimitra Vardalaki
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Qinrong Zhang
- Department of Physics, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Trang L D Pham
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Norma J Brown
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Na Ji
- Department of Physics, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA; Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Mark T Harnett
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
35
|
Xu C, Nedergaard M, Fowell DJ, Friedl P, Ji N. Multiphoton fluorescence microscopy for in vivo imaging. Cell 2024; 187:4458-4487. [PMID: 39178829 PMCID: PMC11373887 DOI: 10.1016/j.cell.2024.07.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/18/2024] [Accepted: 07/22/2024] [Indexed: 08/26/2024]
Abstract
Multiphoton fluorescence microscopy (MPFM) has been a game-changer for optical imaging, particularly for studying biological tissues deep within living organisms. MPFM overcomes the strong scattering of light in heterogeneous tissue by utilizing nonlinear excitation that confines fluorescence emission mostly to the microscope focal volume. This enables high-resolution imaging deep within intact tissue and has opened new avenues for structural and functional studies. MPFM has found widespread applications and has led to numerous scientific discoveries and insights into complex biological processes. Today, MPFM is an indispensable tool in many research communities. Its versatility and effectiveness make it a go-to technique for researchers investigating biological phenomena at the cellular and subcellular levels in their native environments. In this Review, the principles, implementations, capabilities, and limitations of MPFM are presented. Three application areas of MPFM, neuroscience, cancer biology, and immunology, are reviewed in detail and serve as examples for applying MPFM to biological research.
Collapse
Affiliation(s)
- Chris Xu
- School of Applied and Engineering Physics, Cornell University, Ithaca, NY 14850, USA
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Nørre Alle 3B, 2200 Copenhagen, Denmark; University of Rochester Medical School, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | - Deborah J Fowell
- Department of Microbiology & Immunology, Cornell University, Ithaca, NY 14853, USA
| | - Peter Friedl
- Department of Medical BioSciences, Radboud University Medical Centre, Geert Grooteplein 26-28, Nijmegen HB 6500, the Netherlands
| | - Na Ji
- Department of Neuroscience, Department of Physics, University of California Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
36
|
Roth RH, Ding JB. Cortico-basal ganglia plasticity in motor learning. Neuron 2024; 112:2486-2502. [PMID: 39002543 PMCID: PMC11309896 DOI: 10.1016/j.neuron.2024.06.014] [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: 02/29/2024] [Revised: 05/29/2024] [Accepted: 06/17/2024] [Indexed: 07/15/2024]
Abstract
One key function of the brain is to control our body's movements, allowing us to interact with the world around us. Yet, many motor behaviors are not innate but require learning through repeated practice. Among the brain's motor regions, the cortico-basal ganglia circuit is particularly crucial for acquiring and executing motor skills, and neuronal activity in these regions is directly linked to movement parameters. Cell-type-specific adaptations of activity patterns and synaptic connectivity support the learning of new motor skills. Functionally, neuronal activity sequences become structured and associated with learned movements. On the synaptic level, specific connections become potentiated during learning through mechanisms such as long-term synaptic plasticity and dendritic spine dynamics, which are thought to mediate functional circuit plasticity. These synaptic and circuit adaptations within the cortico-basal ganglia circuitry are thus critical for motor skill acquisition, and disruptions in this plasticity can contribute to movement disorders.
Collapse
Affiliation(s)
- Richard H Roth
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA.
| | - Jun B Ding
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA; Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305, USA; The Phil & Penny Knight Initiative for Brain Resilience at the Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
37
|
Goniotaki D, Tamagnini F, Biasetti L, Rumpf S, Troakes C, Pollack SJ, Ukwesa S, Sun H, Kraev I, Serpell LC, Noble W, Staras K, Hanger DP. Tau-mediated synaptic dysfunction is coupled with HCN channelopathy. Alzheimers Dement 2024; 20:5629-5646. [PMID: 38994745 PMCID: PMC11350046 DOI: 10.1002/alz.14074] [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: 01/29/2024] [Revised: 05/01/2024] [Accepted: 05/25/2024] [Indexed: 07/13/2024]
Abstract
INTRODUCTION In tauopathies, altered tau processing correlates with impairments in synaptic density and function. Changes in hyperpolarization-activated cyclic nucleotide-gated (HCN) channels contribute to disease-associated abnormalities in multiple neurodegenerative diseases. METHODS To investigate the link between tau and HCN channels, we performed histological, biochemical, ultrastructural, and functional analyses of hippocampal tissues from Alzheimer's disease (AD), age-matched controls, Tau35 mice, and/or Tau35 primary hippocampal neurons. RESULTS Expression of specific HCN channels is elevated in post mortem AD hippocampus. Tau35 mice develop progressive abnormalities including increased phosphorylated tau, enhanced HCN channel expression, decreased dendritic branching, reduced synapse density, and vesicle clustering defects. Tau35 primary neurons show increased HCN channel expression enhanced hyperpolarization-induced membrane voltage "sag" and changes in the frequency and kinetics of spontaneous excitatory postsynaptic currents. DISCUSSION Our findings are consistent with a model in which pathological changes in tauopathies impact HCN channels to drive network-wide structural and functional synaptic deficits. HIGHLIGHTS Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels are functionally linked to the development of tauopathy. Expression of specific HCN channels is elevated in the hippocampus in Alzheimer's disease and the Tau35 mouse model of tauopathy. Increased expression of HCN channels in Tau35 mice is accompanied by hyperpolarization-induced membrane voltage "sag" demonstrating a detrimental effect of tau abnormalities on HCN channel function. Tau35 expression alters synaptic organization, causing a loosened vesicle clustering phenotype in Tau35 mice.
Collapse
Affiliation(s)
- Despoina Goniotaki
- Department of Basic and Clinical NeuroscienceInstitute of PsychiatryPsychology & NeuroscienceMaurice Wohl Clinical Neuroscience InstituteKing's College LondonLondonUK
| | - Francesco Tamagnini
- Department of PharmacySchool of ChemistryFood and PharmacyUniversity of ReadingReadingUK
| | - Luca Biasetti
- Sussex NeuroscienceSchool of Life SciencesUniversity of SussexBrightonUK
| | - Svenja‐Lotta Rumpf
- Department of Basic and Clinical NeuroscienceInstitute of PsychiatryPsychology & NeuroscienceMaurice Wohl Clinical Neuroscience InstituteKing's College LondonLondonUK
| | - Claire Troakes
- Department of Basic and Clinical NeuroscienceInstitute of PsychiatryPsychology & NeuroscienceMaurice Wohl Clinical Neuroscience InstituteKing's College LondonLondonUK
| | - Saskia J. Pollack
- Department of Basic and Clinical NeuroscienceInstitute of PsychiatryPsychology & NeuroscienceMaurice Wohl Clinical Neuroscience InstituteKing's College LondonLondonUK
| | - Shalom Ukwesa
- Department of Basic and Clinical NeuroscienceInstitute of PsychiatryPsychology & NeuroscienceMaurice Wohl Clinical Neuroscience InstituteKing's College LondonLondonUK
| | - Haoyue Sun
- Department of Basic and Clinical NeuroscienceInstitute of PsychiatryPsychology & NeuroscienceMaurice Wohl Clinical Neuroscience InstituteKing's College LondonLondonUK
| | - Igor Kraev
- Electron Microscopy SuiteSTEM FacultyThe Open UniversityMilton KeynesUK
| | - Louise C. Serpell
- Sussex NeuroscienceSchool of Life SciencesUniversity of SussexBrightonUK
| | - Wendy Noble
- Department of Clinical and Biomedical SciencesUniversity of ExeterExeterUK
| | - Kevin Staras
- Sussex NeuroscienceSchool of Life SciencesUniversity of SussexBrightonUK
| | - Diane P. Hanger
- Department of Basic and Clinical NeuroscienceInstitute of PsychiatryPsychology & NeuroscienceMaurice Wohl Clinical Neuroscience InstituteKing's College LondonLondonUK
| |
Collapse
|
38
|
Cheng X, Nareddula S, Gao HC, Chen Y, Xiao T, Nadew YY, Xu F, Edens PA, Quinn CJ, Kimbrough A, Huang F, Chubykin AA. Impaired Experience-Dependent Theta Oscillation Synchronization and Inter-Areal Synaptic Connectivity in the Visual Cortex of Fmr1 KO Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.23.601989. [PMID: 39211264 PMCID: PMC11360911 DOI: 10.1101/2024.07.23.601989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Fragile X syndrome (FX) is the most prevalent inheritable form of autism spectrum disorder (ASD), characterized by hypersensitivity, difficulty in habituating to new sensory stimuli, and intellectual disability. Individuals with FX often experience visual perception and learning deficits. Visual experience leads to the emergence of the familiarity-evoked theta band oscillations in the primary visual cortex (V1) and the lateromedial area (LM) of mice. These theta oscillations in V1 and LM are synchronized with each other, providing a mechanism of sensory multi-areal binding. However, how this multi-areal binding and the corresponding theta oscillations are altered in FX is not known. Using iDISCO whole brain clearing with light-sheet microscopy, we quantified immediate early gene Fos expression in V1 and LM, identifying deficits in experience-dependent neural activity in FX mice. We performed simultaneous in vivo recordings with silicon probes in V1 and LM of awake mice and channelrhodopsin-2-assisted circuit mapping (CRACM) in acute brain slices to examine the neural activity and strength of long-range synaptic connections between V1 and LM in both wildtype (WT) and Fmr1 knockout (KO) mice, the model of FX, before and after visual experience. Our findings reveal synchronized familiarity-evoked theta oscillations in V1 and LM, the increased strength of V1→LM functional and synaptic connections, which correlated with the corresponding changes of presynaptic short-term plasticity in WT mice. The LM oscillations were attenuated in FX mice and correlated with impaired functional and synaptic connectivity and short-term plasticity in the feedforward (FF) V1→LM and feedback (FB) LM→V1 pathways. Finally, using 4Pi single-molecule localization microscopy (SMLM) in thick brain tissue, we identified experience-dependent changes in the density and shape of dendritic spines in layer 5 pyramidal cells of WT mice, which correlated with the functional synaptic measurements. Interestingly, there was an increased dendritic spine density and length in naïve FX mice that failed to respond to experience. Our study provides the first comprehensive characterization of the role of visual experience in triggering inter-areal neural synchrony and shaping synaptic connectivity in WT and FX mice.
Collapse
|
39
|
Dickstein DL, Zhang R, Ru N, Vozenin MC, Perry BC, Wang J, Baulch J, Acharya MM, Limoli CL. Structural plasticity of pyramidal cell neurons measured after FLASH and conventional dose-rate irradiation. RESEARCH SQUARE 2024:rs.3.rs-4656938. [PMID: 39108471 PMCID: PMC11302692 DOI: 10.21203/rs.3.rs-4656938/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Evidence shows that ultra-high dose-rate FLASH-radiotherapy (FLASH-RT) protects against normal tissue complications and functional decrements in the irradiated brain. Past work has shown that radiation-induced cognitive impairment, neuroinflammation and reduced structural complexity of granule cell neurons were not observed to the same extent after FLASH-RT (> MGy/s) compared to conventional dose-rate (CONV, 0.1 Gy/s) delivery. To explore the sensitivity of different neuronal populations to cranial irradiation and dose-rate modulation, hippocampal CA1 and medial prefrontal cortex (PFC) pyramidal neurons were analyzed by electron and confocal microscopy. Neuron ultrastructural analyses by electron microscopy after 10 Gy FLASH- or CONV-RT exposures indicated that irradiation had little impact on dendritic complexity and synapse density in the CA1, but did increase length and head diameter of smaller non-perforated synapses. Similarly, irradiation caused no change in PFC prelimbic/infralimbic axospinous synapse density, but reductions in non-perforated synapse diameters. While irradiation resulted in thinner myelin sheaths compared to controls, none of these metrics were dose-rate sensitive. Analysis of fluorescently labeled CA1 neurons revealed no radiation-induced or dose-rate-dependent changes in overall dendritic complexity or spine density, in contrast to our past analysis of granule cell neurons. Super-resolution confocal microscopy following a clinical dosing paradigm (3×10Gy) showed significant reductions in excitatory vesicular glutamate transporter 1 and inhibitory vesicular GABA transporter puncta density within the CA1 that were largely dose-rate independent. Collectively, these data reveal that, compared to granule cell neurons, CA1 and mPFC neurons are more radioresistant irrespective of radiation dose-rate.
Collapse
Affiliation(s)
| | | | - Ning Ru
- University of California, Irvine School of Medicine
| | | | | | - Juan Wang
- Uniformed Services University of Health Sciences
| | - Janet Baulch
- University of California, Irvine School of Medicine
| | | | | |
Collapse
|
40
|
Zhang J, Vaidya R, Chung HJ, Selvin PR. Automatic dendritic spine segmentation in widefield fluorescence images reveal synaptic nanostructures distribution with super-resolution imaging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.15.603616. [PMID: 39071361 PMCID: PMC11275708 DOI: 10.1101/2024.07.15.603616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Dendritic spines are the main sites for synaptic communication in neurons, and alterations in their density, size, and shapes occur in many brain disorders. Current spine segmentation methods perform poorly in conditions with low signal-to-noise and resolution, particularly in the widefield images of thick (10 μm) brain slices. Here, we combined two open-source machine-learning models to achieve automatic 3D spine segmentation in widefield diffraction-limited fluorescence images of neurons in thick brain slices. We validated the performance by comparison with manually segmented super-resolution images of spines reconstructed from direct stochastic optical reconstruction microscopy (dSTORM). Lastly, we show an application of our approach by combining spine segmentation from diffraction-limited images with dSTORM of synaptic protein PSD-95 in the same field-of-view. This allowed us to automatically analyze and quantify the nanoscale distribution of PSD-95 inside the spine. Importantly, we found the numbers, but not the average sizes, of synaptic nanomodules and nanodomains increase with spine size.
Collapse
Affiliation(s)
- Jiahao Zhang
- Dept. of Physics, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Rohit Vaidya
- Center for Biophysics and Quantitative Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Hee Jung Chung
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Paul R Selvin
- Dept. of Physics, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Center for Biophysics and Quantitative Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| |
Collapse
|
41
|
Leighton AH, Cheyne JE, Lohmann C. Clustered synapses develop in distinct dendritic domains in visual cortex before eye opening. eLife 2024; 12:RP93498. [PMID: 38990761 PMCID: PMC11239177 DOI: 10.7554/elife.93498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/13/2024] Open
Abstract
Synaptic inputs to cortical neurons are highly structured in adult sensory systems, such that neighboring synapses along dendrites are activated by similar stimuli. This organization of synaptic inputs, called synaptic clustering, is required for high-fidelity signal processing, and clustered synapses can already be observed before eye opening. However, how clustered inputs emerge during development is unknown. Here, we employed concurrent in vivo whole-cell patch-clamp and dendritic calcium imaging to map spontaneous synaptic inputs to dendrites of layer 2/3 neurons in the mouse primary visual cortex during the second postnatal week until eye opening. We found that the number of functional synapses and the frequency of transmission events increase several fold during this developmental period. At the beginning of the second postnatal week, synapses assemble specifically in confined dendritic segments, whereas other segments are devoid of synapses. By the end of the second postnatal week, just before eye opening, dendrites are almost entirely covered by domains of co-active synapses. Finally, co-activity with their neighbor synapses correlates with synaptic stabilization and potentiation. Thus, clustered synapses form in distinct functional domains presumably to equip dendrites with computational modules for high-capacity sensory processing when the eyes open.
Collapse
Affiliation(s)
- Alexandra H Leighton
- Department of Synapse and Network Development, Netherlands Institute for NeuroscienceAmsterdamNetherlands
| | - Juliette E Cheyne
- Department of Synapse and Network Development, Netherlands Institute for NeuroscienceAmsterdamNetherlands
| | - Christian Lohmann
- Department of Synapse and Network Development, Netherlands Institute for NeuroscienceAmsterdamNetherlands
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, VU University AmsterdamAmsterdamNetherlands
| |
Collapse
|
42
|
Currey L, Mitchell B, Al-Khalily M, McElnea SJ, Kozulin P, Harkins D, Pelenyi A, Fenlon L, Suarez R, Kurniawan ND, Burne TH, Harris L, Thor S, Piper M. Polycomb repressive complex 2 is critical for mouse cortical glutamatergic neuron development. Cereb Cortex 2024; 34:bhae268. [PMID: 38960704 PMCID: PMC11221884 DOI: 10.1093/cercor/bhae268] [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: 11/21/2023] [Revised: 06/11/2024] [Accepted: 06/13/2024] [Indexed: 07/05/2024] Open
Abstract
The Polycomb Repressive Complex 2 (PRC2) regulates corticogenesis, yet the consequences of mutations to this epigenetic modifier in the mature brain are poorly defined. Importantly, PRC2 core genes are haploinsufficient and causative of several human neurodevelopmental disorders. To address the role of PRC2 in mature cortical structure and function, we conditionally deleted the PRC2 gene Eed from the developing mouse dorsal telencephalon. Adult homozygotes displayed smaller forebrain structures. Single-nucleus transcriptomics revealed that glutamatergic neurons were particularly affected, exhibiting dysregulated gene expression profiles, accompanied by aberrations in neuronal morphology and connectivity. Remarkably, homozygous mice performed well on challenging cognitive tasks. In contrast, while heterozygous mice did not exhibit clear anatomical or behavioral differences, they displayed dysregulation of neuronal genes and altered neuronal morphology that was strikingly different from homozygous phenotypes. Collectively, these data reveal how alterations to PRC2 function shape the mature brain and reveal a dose-specific role for PRC2 in determining glutamatergic neuron identity.
Collapse
Affiliation(s)
- Laura Currey
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Benjamin Mitchell
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Majd Al-Khalily
- Centre for Advanced Imaging, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, QLD 4072, Australia
| | - Sarah-Jayne McElnea
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Peter Kozulin
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Danyon Harkins
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Alexandra Pelenyi
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Laura Fenlon
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Rodrigo Suarez
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Nyoman D Kurniawan
- Centre for Advanced Imaging, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, QLD 4072, Australia
| | - Thomas H Burne
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
- Queensland Centre for Mental Health Research, The Park Centre for Mental Health, Wacol, QLD 4076, Australia
| | - Lachlan Harris
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
- Cancer Neuroscience Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia
| | - Stefan Thor
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Michael Piper
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| |
Collapse
|
43
|
Lee CT, Bell M, Bonilla-Quintana M, Rangamani P. Biophysical Modeling of Synaptic Plasticity. Annu Rev Biophys 2024; 53:397-426. [PMID: 38382115 DOI: 10.1146/annurev-biophys-072123-124954] [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: 02/23/2024]
Abstract
Dendritic spines are small, bulbous compartments that function as postsynaptic sites and undergo intense biochemical and biophysical activity. The role of the myriad signaling pathways that are implicated in synaptic plasticity is well studied. A recent abundance of quantitative experimental data has made the events associated with synaptic plasticity amenable to quantitative biophysical modeling. Spines are also fascinating biophysical computational units because spine geometry, signal transduction, and mechanics work in a complex feedback loop to tune synaptic plasticity. In this sense, ideas from modeling cell motility can inspire us to develop multiscale approaches for predictive modeling of synaptic plasticity. In this article, we review the key steps in postsynaptic plasticity with a specific focus on the impact of spine geometry on signaling, cytoskeleton rearrangement, and membrane mechanics. We summarize the main experimental observations and highlight how theory and computation can aid our understanding of these complex processes.
Collapse
Affiliation(s)
- Christopher T Lee
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, California, USA;
| | - Miriam Bell
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, California, USA;
| | - Mayte Bonilla-Quintana
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, California, USA;
| | - Padmini Rangamani
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, California, USA;
| |
Collapse
|
44
|
Ge Q, Zhou S, Porras J, Fu P, Wang T, Du J, Li K. SARS-CoV-2 neurotropism-induced anxiety/depression-like behaviors require Microglia activation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.02.560570. [PMID: 37873397 PMCID: PMC10592887 DOI: 10.1101/2023.10.02.560570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has been associated with a wide range of "long COVID" neurological symptoms. However, the mechanisms governing SARS-CoV-2 neurotropism and its effects on long-term behavioral changes remain poorly understood. Using a highly virulent mouse-adapted SARS-CoV-2 strain, denoted as SARS2-N501Y MA30 , we demonstrated that intranasal inoculation of SARS2-N501Y MA30 results in viral dissemination to multiple brain regions, including the amygdala and hippocampus. Behavioral assays indicated a marked elevation in anxiety- and depression-like behaviors post infection. A comparative analysis of RNA expression profiles disclosed alterations in the post-infected brains. Additionally, we observed dendritic spine remodeling on neurons within the amygdala after infection. Infection with SARS2-N501Y MA30 was associated with microglial activation and a subsequent increase in microglia-dependent neuronal activity in the amygdala. Pharmacological inhibition of microglial activity subsequent to viral spike inoculation mitigates microglia-dependent neuronal hyperactivity. Transcriptomic analysis of infected brains revealed the upregulation of inflammatory and cytokine-related pathways, implicating microglia-driven neuroinflammation in the pathogenesis of neuronal hyperactivity and behavioral abnormality. Overall, these data provide critical insights into the neurological consequences of SARS-CoV-2 infection and underscore microglia as a potential therapeutic target for ameliorating virus-induced neurobehavioral abnormalities.
Collapse
|
45
|
Li XY, Zhang SY, Hong YZ, Chen ZG, Long Y, Yuan DH, Zhao JJ, Tang SS, Wang H, Hong H. TGR5-mediated lateral hypothalamus-dCA3-dorsolateral septum circuit regulates depressive-like behavior in male mice. Neuron 2024; 112:1795-1814.e10. [PMID: 38518778 DOI: 10.1016/j.neuron.2024.02.019] [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: 07/17/2023] [Revised: 01/30/2024] [Accepted: 02/28/2024] [Indexed: 03/24/2024]
Abstract
Although bile acids play a notable role in depression, the pathological significance of the bile acid TGR5 membrane-type receptor in this disorder remains elusive. Using depression models of chronic social defeat stress and chronic restraint stress in male mice, we found that TGR5 in the lateral hypothalamic area (LHA) predominantly decreased in GABAergic neurons, the excitability of which increased in depressive-like mice. Upregulation of TGR5 or inhibition of GABAergic excitability in LHA markedly alleviated depressive-like behavior, whereas down-regulation of TGR5 or enhancement of GABAergic excitability facilitated stress-induced depressive-like behavior. TGR5 also bidirectionally regulated excitability of LHA GABAergic neurons via extracellular regulated protein kinases-dependent Kv4.2 channels. Notably, LHA GABAergic neurons specifically innervated dorsal CA3 (dCA3) CaMKIIα neurons for mediation of depressive-like behavior. LHA GABAergic TGR5 exerted antidepressant-like effects by disinhibiting dCA3 CaMKIIα neurons projecting to the dorsolateral septum (DLS). These findings advance our understanding of TGR5 and the LHAGABA→dCA3CaMKIIα→DLSGABA circuit for the development of potential therapeutic strategies in depression.
Collapse
Affiliation(s)
- Xu-Yi Li
- College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Shi-Ya Zhang
- College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yi-Zhou Hong
- Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Zhi-Gang Chen
- College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yan Long
- College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Dan-Hua Yuan
- College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Jia-Jia Zhao
- College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Su-Su Tang
- College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| | - Hao Wang
- Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine/Nanhu Brain-Computer Interface Institute, Hangzhou 310013, China.
| | - Hao Hong
- College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
46
|
Deng Y, Liang X, Li Y, Jiang L, Wang J, Tang J, Li J, Xie Y, Xiao K, Zhu P, Guo Y, Luo Y, Tang Y. PGC-1α in the hippocampus mediates depressive-like and stress-coping behaviours and regulates excitatory synapses in the dentate gyrus in mice. Neuropharmacology 2024; 250:109908. [PMID: 38492883 DOI: 10.1016/j.neuropharm.2024.109908] [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: 09/17/2023] [Revised: 02/13/2024] [Accepted: 03/10/2024] [Indexed: 03/18/2024]
Abstract
Decreased hippocampal synaptic plasticity is an important pathological change in stress-related mood disorders, including major depressive disorder. However, the underlying mechanism is unclear. PGC-1α, a transcriptional coactivator, is a key factor in synaptic plasticity. We investigated the relationships between changes in hippocampal PGC-1α expression and depressive-like and stress-coping behaviours, and whether they are related to hippocampal synapses. Adeno-associated virus was used to alter hippocampal PGC-1α expression in male C57BL/6 mice. The sucrose preference test and forced swimming test were used to assess their depressive-like and stress-coping behaviours, respectively. Immunohistochemistry and stereology were used to calculate the total number of excitatory synapses in each hippocampal subregion (the cornu ammonis (CA) 1, CA3, and dentate gyrus). Immunofluorescence was used to visualize the changes in dendritic structure. Western blotting was used to detect the expression of hippocampal PGC-1α and mitochondrial-associated proteins, such as UCP2, NRF1 and mtTFAs. Our results showed that mice with downregulated PGC-1α expression in the hippocampus exhibited depressive-like and passive stress-coping behaviours, while mice with upregulated PGC-1α in the hippocampus exhibited increased stress-coping behaviours. Moreover, the downregulation of hippocampal PGC-1α expression resulted in a decrease in the number of excitatory synapses in the DG and in the protein expression of UCP2 in the hippocampus. Alternatively, upregulation of hippocampal PGC-1α yielded the opposite results. This suggests that hippocampal PGC-1α is involved in regulating depressive-like and stress-coping behaviours and modulating the number of excitatory synapses in the DG. This provides new insight for the development of antidepressants.
Collapse
Affiliation(s)
- Yuhui Deng
- Department of Histology and Embryology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China
| | - Xin Liang
- Department of Pathology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China
| | - Yue Li
- Department of Histology and Embryology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China
| | - Lin Jiang
- Lab Teaching and Management Center, Chongqing Medical University, Chongqing, 400016, PR China
| | - Jin Wang
- Department of Histology and Embryology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China
| | - Jing Tang
- Department of Histology and Embryology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China
| | - Jing Li
- Department of Histology and Embryology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China
| | - Yuhan Xie
- Department of Histology and Embryology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China
| | - Kai Xiao
- Department of Histology and Embryology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China
| | - Peilin Zhu
- Department of Histology and Embryology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China
| | - Yijing Guo
- Department of Histology and Embryology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China
| | - Yanmin Luo
- Department of Physiology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China.
| | - Yong Tang
- Department of Histology and Embryology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China.
| |
Collapse
|
47
|
Kaizuka T, Takumi T. Alteration of synaptic protein composition during developmental synapse maturation. Eur J Neurosci 2024; 59:2894-2914. [PMID: 38571321 DOI: 10.1111/ejn.16304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 01/02/2024] [Accepted: 02/07/2024] [Indexed: 04/05/2024]
Abstract
The postsynaptic density (PSD) is a collection of specialized proteins assembled beneath the postsynaptic membrane of dendritic spines. The PSD proteome comprises ~1000 proteins, including neurotransmitter receptors, scaffolding proteins and signalling enzymes. Many of these proteins have essential roles in synaptic function and plasticity. During brain development, changes are observed in synapse density and in the stability and shape of spines, reflecting the underlying molecular maturation of synapses. Synaptic protein composition changes in terms of protein abundance and the assembly of protein complexes, supercomplexes and the physical organization of the PSD. Here, we summarize the developmental alterations of postsynaptic protein composition during synapse maturation. We describe major PSD proteins involved in postsynaptic signalling that regulates synaptic plasticity and discuss the effect of altered expression of these proteins during development. We consider the abnormality of synaptic profiles and synaptic protein composition in the brain in neurodevelopmental disorders such as autism spectrum disorders. We also explain differences in synapse development between rodents and primates in terms of synaptic profiles and protein composition. Finally, we introduce recent findings related to synaptic diversity and nanoarchitecture and discuss their impact on future research. Synaptic protein composition can be considered a major determinant and marker of synapse maturation in normality and disease.
Collapse
Affiliation(s)
- Takeshi Kaizuka
- Department of Physiology and Cell Biology, Kobe University School of Medicine, Kobe, Japan
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Toru Takumi
- Department of Physiology and Cell Biology, Kobe University School of Medicine, Kobe, Japan
- RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| |
Collapse
|
48
|
Yu L, Zeng F, Fan M, Zhang K, Duan J, Tan Y, Liao P, Wen J, Wang C, Wang M, Yuan J, Pang X, Huang Y, Zhang Y, Li JD, Zhang Z, Hu Z. PCDH17 restricts dendritic spine morphogenesis by regulating ROCK2-dependent control of the actin cytoskeleton, modulating emotional behavior. Zool Res 2024; 45:535-550. [PMID: 38747058 PMCID: PMC11188600 DOI: 10.24272/j.issn.2095-8137.2024.055] [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: 02/26/2024] [Accepted: 05/10/2024] [Indexed: 06/05/2024] Open
Abstract
Proper regulation of synapse formation and elimination is critical for establishing mature neuronal circuits and maintaining brain function. Synaptic abnormalities, such as defects in the density and morphology of postsynaptic dendritic spines, underlie the pathology of various neuropsychiatric disorders. Protocadherin 17 (PCDH17) is associated with major mood disorders, including bipolar disorder and depression. However, the molecular mechanisms by which PCDH17 regulates spine number, morphology, and behavior remain elusive. In this study, we found that PCDH17 functions at postsynaptic sites, restricting the number and size of dendritic spines in excitatory neurons. Selective overexpression of PCDH17 in the ventral hippocampal CA1 results in spine loss and anxiety- and depression-like behaviors in mice. Mechanistically, PCDH17 interacts with actin-relevant proteins and regulates actin filament (F-actin) organization. Specifically, PCDH17 binds to ROCK2, increasing its expression and subsequently enhancing the activity of downstream targets such as LIMK1 and the phosphorylation of cofilin serine-3 (Ser3). Inhibition of ROCK2 activity with belumosudil (KD025) ameliorates the defective F-actin organization and spine structure induced by PCDH17 overexpression, suggesting that ROCK2 mediates the effects of PCDH17 on F-actin content and spine development. Hence, these findings reveal a novel mechanism by which PCDH17 regulates synapse development and behavior, providing pathological insights into the neurobiological basis of mood disorders.
Collapse
Affiliation(s)
- Laidong Yu
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan 410008, China
| | - Fangfang Zeng
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan 410008, China
| | - Mengshu Fan
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan 410008, China
| | - Kexuan Zhang
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan 410008, China
| | - Jingjing Duan
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Hunan Provincial Clinical Research Center for Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yalu Tan
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Hunan Provincial Clinical Research Center for Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Panlin Liao
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Hunan Provincial Clinical Research Center for Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Jin Wen
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan 410008, China
| | - Chenyu Wang
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Meilin Wang
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Jialong Yuan
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan 410008, China
| | - Xinxin Pang
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan 410008, China
| | - Yan Huang
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Hunan Provincial Clinical Research Center for Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yangzhou Zhang
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Hunan Provincial Clinical Research Center for Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Jia-Da Li
- Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan 410008, China
- Hunan Key Laboratory of Animal Models for Human Diseases, School of Life Sciences, Central South University, Changsha, Hunan 410008, China
- MOE Key Lab of Rare Pediatric Diseases, School of Life Sciences, Central South University, Changsha, Hunan 410008, China. E-mail:
| | - Zhuohua Zhang
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan 410008, China
- MOE Key Lab of Rare Pediatric Diseases, School of Life Sciences, Central South University, Changsha, Hunan 410008, China
- Department of Neurosciences, University of South China Medical School, Hengyang, Hunan 421001, China. E-mail:
| | - Zhonghua Hu
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Hunan Provincial Clinical Research Center for Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- MOE Key Lab of Rare Pediatric Diseases, School of Life Sciences, Central South University, Changsha, Hunan 410008, China. E-mail:
| |
Collapse
|
49
|
Zong R, Zhang X, Dong X, Liu G, Zhang J, Gao Y, Zhang Z, Ma Y, Gao H, Gamper N. Genetic deletion of zinc transporter ZnT 3 induces progressive cognitive deficits in mice by impairing dendritic spine plasticity and glucose metabolism. Front Mol Neurosci 2024; 17:1375925. [PMID: 38807922 PMCID: PMC11130425 DOI: 10.3389/fnmol.2024.1375925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 04/22/2024] [Indexed: 05/30/2024] Open
Abstract
Zinc transporter 3 (ZnT3) is abundantly expressed in the brain, residing in synaptic vesicles, where it plays important roles in controlling the luminal zinc levels. In this study, we found that ZnT3 knockout in mice decreased zinc levels in the hippocampus and cortex, and was associated with progressive cognitive impairments, assessed at 2, 6, and 9-month of age. The results of Golgi-Cox staining demonstrated that ZnT3 deficiency was associated with an increase in dendritic complexity and a decrease in the density of mature dendritic spines, indicating potential synaptic plasticity deficit. Since ZnT3 deficiency was previously linked to glucose metabolism abnormalities, we tested the expression levels of genes related to insulin signaling pathway in the hippocampus and cortex. We found that the Expression of glucose transporters, GLUT3, GLUT4, and the insulin receptor in the whole tissue and synaptosome fraction of the hippocampus of the ZnT3 knockout mice were significantly reduced, as compared to wild-type controls. Expression of AKT (A serine/threonine protein kinase) and insulin-induced AKT phosphorylation was also reduced in the hippocampus of ZnT3 knockout mice. We hypothesize that the ZnT3 deficiency and reduced brain zinc levels may cause cognitive impairment by negatively affecting glycose metabolism via decreased expression of key components of insulin signaling, as well as via changes in synaptic plasticity. These finding may provide new therapeutic target for treatments of neurodegenerative disorders.
Collapse
Affiliation(s)
- Rui Zong
- Department of Pharmacology, Center for Innovative Drug Research and Evaluation, Institute of Medical Science and Health, The Hebei Collaboration Innovation Center for Mechanism, Diagnosis and Treatment of Neurological and Psychiatric Disease, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xiaoding Zhang
- Department of Pharmacology, Center for Innovative Drug Research and Evaluation, Institute of Medical Science and Health, The Hebei Collaboration Innovation Center for Mechanism, Diagnosis and Treatment of Neurological and Psychiatric Disease, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xiaohui Dong
- Department of Pharmacology, Center for Innovative Drug Research and Evaluation, Institute of Medical Science and Health, The Hebei Collaboration Innovation Center for Mechanism, Diagnosis and Treatment of Neurological and Psychiatric Disease, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Guan Liu
- Department of Pharmacology, Center for Innovative Drug Research and Evaluation, Institute of Medical Science and Health, The Hebei Collaboration Innovation Center for Mechanism, Diagnosis and Treatment of Neurological and Psychiatric Disease, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jieyao Zhang
- Department of Pharmacology, Center for Innovative Drug Research and Evaluation, Institute of Medical Science and Health, The Hebei Collaboration Innovation Center for Mechanism, Diagnosis and Treatment of Neurological and Psychiatric Disease, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yiting Gao
- Department of Pharmacology, Center for Innovative Drug Research and Evaluation, Institute of Medical Science and Health, The Hebei Collaboration Innovation Center for Mechanism, Diagnosis and Treatment of Neurological and Psychiatric Disease, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Zhongyang Zhang
- Department of Pharmacology, Center for Innovative Drug Research and Evaluation, Institute of Medical Science and Health, The Hebei Collaboration Innovation Center for Mechanism, Diagnosis and Treatment of Neurological and Psychiatric Disease, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yiming Ma
- Department of Pharmacology, Center for Innovative Drug Research and Evaluation, Institute of Medical Science and Health, The Hebei Collaboration Innovation Center for Mechanism, Diagnosis and Treatment of Neurological and Psychiatric Disease, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Haixia Gao
- Department of Pharmacology, Center for Innovative Drug Research and Evaluation, Institute of Medical Science and Health, The Hebei Collaboration Innovation Center for Mechanism, Diagnosis and Treatment of Neurological and Psychiatric Disease, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Nikita Gamper
- Department of Pharmacology, Center for Innovative Drug Research and Evaluation, Institute of Medical Science and Health, The Hebei Collaboration Innovation Center for Mechanism, Diagnosis and Treatment of Neurological and Psychiatric Disease, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei, China
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
50
|
Odierna GL, Vucic S, Dyer M, Dickson T, Woodhouse A, Blizzard C. How do we get from hyperexcitability to excitotoxicity in amyotrophic lateral sclerosis? Brain 2024; 147:1610-1621. [PMID: 38408864 PMCID: PMC11068114 DOI: 10.1093/brain/awae039] [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: 07/17/2023] [Revised: 11/15/2023] [Accepted: 12/10/2023] [Indexed: 02/28/2024] Open
Abstract
Amyotrophic lateral sclerosis is a devastating neurodegenerative disease that, at present, has no effective cure. Evidence of increased circulating glutamate and hyperexcitability of the motor cortex in patients with amyotrophic lateral sclerosis have provided an empirical support base for the 'dying forward' excitotoxicity hypothesis. The hypothesis postulates that increased activation of upper motor neurons spreads pathology to lower motor neurons in the spinal cord in the form of excessive glutamate release, which triggers excitotoxic processes. Many clinical trials have focused on therapies that target excitotoxicity via dampening neuronal activation, but not all are effective. As such, there is a growing tension between the rising tide of evidence for the 'dying forward' excitotoxicity hypothesis and the failure of therapies that target neuronal activation. One possible solution to these contradictory outcomes is that our interpretation of the current evidence requires revision in the context of appreciating the complexity of the nervous system and the limitations of the neurobiological assays we use to study it. In this review we provide an evaluation of evidence relevant to the 'dying forward' excitotoxicity hypothesis and by doing so, identify key gaps in our knowledge that need to be addressed. We hope to provide a road map from hyperexcitability to excitotoxicity so that we can better develop therapies for patients suffering from amyotrophic lateral sclerosis. We conclude that studies of upper motor neuron activity and their synaptic output will play a decisive role in the future of amyotrophic lateral sclerosis therapy.
Collapse
Affiliation(s)
- G Lorenzo Odierna
- Tasmanian School of Medicine, University of Tasmania, Hobart, TAS 7000, Australia
| | - Steve Vucic
- Brain and Nerve Research Center, The University of Sydney, Sydney 2050, Australia
| | - Marcus Dyer
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia
- Department of Pharmaceutical and Pharmacological Sciences, Center for Neurosciences, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium
| | - Tracey Dickson
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia
| | - Adele Woodhouse
- The Wicking Dementia Centre, University of Tasmania, Hobart, TAS 7000, Australia
| | - Catherine Blizzard
- Tasmanian School of Medicine, University of Tasmania, Hobart, TAS 7000, Australia
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia
| |
Collapse
|