1
|
Tu G, Jiang N, Chen W, Liu L, Hu M, Liao B. The neurobiological mechanisms underlying the effects of exercise interventions in autistic individuals. Rev Neurosci 2025; 36:27-51. [PMID: 39083671 DOI: 10.1515/revneuro-2024-0058] [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: 04/22/2024] [Accepted: 07/15/2024] [Indexed: 08/02/2024]
Abstract
Autism spectrum disorder is a pervasive and heterogeneous neurodevelopmental condition characterized by social communication difficulties and rigid, repetitive behaviors. Owing to the complex pathogenesis of autism, effective drugs for treating its core features are lacking. Nonpharmacological approaches, including education, social-communication, behavioral and psychological methods, and exercise interventions, play important roles in supporting the needs of autistic individuals. The advantages of exercise intervention, such as its low cost, easy implementation, and high acceptance, have garnered increasing attention. Exercise interventions can effectively improve the core features and co-occurring conditions of autism, but the underlying neurobiological mechanisms are unclear. Abnormal changes in the gut microbiome, neuroinflammation, neurogenesis, and synaptic plasticity may individually or interactively be responsible for atypical brain structure and connectivity, leading to specific autistic experiences and characteristics. Interestingly, exercise can affect these biological processes and reshape brain network connections, which may explain how exercise alleviates core features and co-occurring conditions in autistic individuals. In this review, we describe the definition, diagnostic approach, epidemiology, and current support strategies for autism; highlight the benefits of exercise interventions; and call for individualized programs for different subtypes of autistic individuals. Finally, the possible neurobiological mechanisms by which exercise improves autistic features are comprehensively summarized to inform the development of optimal exercise interventions and specific targets to meet the needs of autistic individuals.
Collapse
Affiliation(s)
- Genghong Tu
- Department of Sports Medicine, 47878 Guangzhou Sport University , Guangzhou, Guangdong, 510500, P.R. China
- Guangdong Provincial Key Laboratory of Physical Activity and Health Promotion, 47878 Scientific Research Center, Guangzhou Sport University , Guangzhou, Guangdong, 510500, P.R. China
| | - Nan Jiang
- Graduate School, 47878 Guangzhou Sport University , Guangzhou, Guangdong, 510500, P.R. China
| | - Weizhong Chen
- Graduate School, 47878 Guangzhou Sport University , Guangzhou, Guangdong, 510500, P.R. China
| | - Lining Liu
- Graduate School, 47878 Guangzhou Sport University , Guangzhou, Guangdong, 510500, P.R. China
| | - Min Hu
- Guangdong Provincial Key Laboratory of Physical Activity and Health Promotion, 47878 Scientific Research Center, Guangzhou Sport University , Guangzhou, Guangdong, 510500, P.R. China
| | - Bagen Liao
- Department of Sports Medicine, 47878 Guangzhou Sport University , Guangzhou, Guangdong, 510500, P.R. China
- Guangdong Provincial Key Laboratory of Physical Activity and Health Promotion, 47878 Scientific Research Center, Guangzhou Sport University , Guangzhou, Guangdong, 510500, P.R. China
| |
Collapse
|
2
|
Majumder P, Chatterjee B, Akter K, Ahsan A, Tan SJ, Huang CC, Chu JF, Shen CKJ. Molecular switch of the dendrite-to-spine transport of TDP-43/FMRP-bound neuronal mRNAs and its impairment in ASD. Cell Mol Biol Lett 2025; 30:6. [PMID: 39815169 PMCID: PMC11737055 DOI: 10.1186/s11658-024-00684-5] [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: 09/02/2024] [Accepted: 12/23/2024] [Indexed: 01/18/2025] Open
Abstract
BACKGROUND Regulation of messenger RNA (mRNA) transport and translation in neurons is essential for dendritic plasticity and learning/memory development. The trafficking of mRNAs along the hippocampal neuron dendrites remains translationally silent until they are selectively transported into the spines upon glutamate-induced receptor activation. However, the molecular mechanism(s) behind the spine entry of dendritic mRNAs under metabotropic glutamate receptor (mGluR)-mediated neuroactivation and long-term depression (LTD) as well as the fate of these mRNAs inside the spines are still elusive. METHOD Different molecular and imaging techniques, e.g., immunoprecipitation (IP), RNA-IP, Immunofluorescence (IF)/fluorescence in situ hybridization (FISH), live cell imaging, live cell tracking of RNA using beacon, and mouse model study are used to elucidate a novel mechanism regulating dendritic spine transport of mRNAs in mammalian neurons. RESULTS We demonstrate here that brief mGluR1 activation-mediated dephosphorylation of pFMRP (S499) results in the dissociation of FMRP from TDP-43 and handover of TDP-43/Rac1 mRNA complex from the dendritic transport track on microtubules to myosin V track on the spine actin filaments. Rac1 mRNA thus enters the spines for translational reactivation and increases the mature spine density. In contrast, during mGluR1-mediated neuronal LTD, FMRP (S499) remains phosphorylated and the TDP-43/Rac1 mRNA complex, being associated with kinesin 1-FMRP/cortactin/drebrin, enters the spines owing to Ca2+-dependent microtubule invasion into spines, but without translational reactivation. In a VPA-ASD mouse model, this regulation become anomalous. CONCLUSIONS This study, for the first time, highlights the importance of posttranslational modification of RBPs, such as the neurodevelopmental disease-related protein FMRP, as the molecular switch regulating the dendrite-to-spine transport of specific mRNAs under mGluR1-mediated neurotransmissions. The misregulation of this switch could contribute to the pathogenesis of FMRP-related neurodisorders including the autism spectrum disorder (ASD). It also could indicate a molecular connection between ASD and neurodegenerative disease-related protein TDP-43 and opens up a new perspective of research to elucidate TDP-43 proteinopathy among patients with ASD.
Collapse
Affiliation(s)
- Pritha Majumder
- PhD Program in Medical Neuroscience, Taipei Medical University, Taipei, Taiwan (R.O.C.).
- Institute of Molecular Medicine, College of Medicine, National Chen Kung University, Tainan, Taiwan (R.O.C.).
| | - Biswanath Chatterjee
- PhD Program in Medical Neuroscience, Taipei Medical University, Taipei, Taiwan (R.O.C.)
| | - Khadiza Akter
- PhD Program in Medical Neuroscience, Taipei Medical University, Taipei, Taiwan (R.O.C.)
| | - Asmar Ahsan
- PhD Program in Medical Neuroscience, Taipei Medical University, Taipei, Taiwan (R.O.C.)
| | - Su Jie Tan
- Institute of Molecular Medicine, College of Medicine, National Chen Kung University, Tainan, Taiwan (R.O.C.)
| | - Chi-Chen Huang
- PhD Program in Medical Neuroscience, Taipei Medical University, Taipei, Taiwan (R.O.C.)
| | - Jen-Fei Chu
- PhD Program in Medical Neuroscience, Taipei Medical University, Taipei, Taiwan (R.O.C.).
| | - Che-Kun James Shen
- PhD Program in Medical Neuroscience, Taipei Medical University, Taipei, Taiwan (R.O.C.).
- Institute of Molecular Biology, Academia Sinica, Nangang, Taipei, 115, Taiwan (R.O.C.).
| |
Collapse
|
3
|
Heuer SE, Bloss EB, Howell GR. Strategies to dissect microglia-synaptic interactions during aging and in Alzheimer's disease. Neuropharmacology 2024; 254:109987. [PMID: 38705570 DOI: 10.1016/j.neuropharm.2024.109987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 04/29/2024] [Accepted: 05/02/2024] [Indexed: 05/07/2024]
Abstract
Age is the largest risk factor for developing Alzheimer's disease (AD), a neurodegenerative disorder that causes a progressive and severe dementia. The underlying cause of cognitive deficits seen in AD is thought to be the disconnection of neural circuits that control memory and executive functions. Insight into the mechanisms by which AD diverges from normal aging will require identifying precisely which cellular events are driven by aging and which are impacted by AD-related pathologies. Since microglia, the brain-resident macrophages, are known to have critical roles in the formation and maintenance of neural circuits through synaptic pruning, they are well-positioned to modulate synaptic connectivity in circuits sensitive to aging or AD. In this review, we provide an overview of the current state of the field and on emerging technologies being employed to elucidate microglia-synaptic interactions in aging and AD. We also discuss the importance of leveraging genetic diversity to study how these interactions are shaped across more realistic contexts. We propose that these approaches will be essential to define specific aging- and disease-relevant trajectories for more personalized therapeutics aimed at reducing the effects of age or AD pathologies on the brain. This article is part of the Special Issue on "Microglia".
Collapse
Affiliation(s)
- Sarah E Heuer
- The Jackson Laboratory, Bar Harbor, ME, 04609, USA; Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, 02111, USA
| | - Erik B Bloss
- The Jackson Laboratory, Bar Harbor, ME, 04609, USA; Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, 02111, USA; Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME, 04469, USA.
| | - Gareth R Howell
- The Jackson Laboratory, Bar Harbor, ME, 04609, USA; Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, 02111, USA; Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME, 04469, USA.
| |
Collapse
|
4
|
Voglewede MM, Ozsen EN, Ivak N, Bernabucci M, Tang R, Sun M, Pang ZP, Zhang H. Loss of the polarity protein Par3 promotes dendritic spine neoteny and enhances learning and memory. iScience 2024; 27:110308. [PMID: 39045101 PMCID: PMC11263792 DOI: 10.1016/j.isci.2024.110308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 03/25/2024] [Accepted: 06/17/2024] [Indexed: 07/25/2024] Open
Abstract
The Par3 polarity protein is critical for subcellular compartmentalization in different developmental processes. Variants of PARD3, encoding PAR3, are associated with intelligence and neurodevelopmental disorders. However, the role of Par3 in glutamatergic synapse formation and cognitive functions in vivo remains unknown. Here, we show that forebrain-specific Par3 conditional knockout leads to increased long, thin dendritic spines in vivo. In addition, we observed a decrease in the amplitude of miniature excitatory postsynaptic currents. Surprisingly, loss of Par3 enhances hippocampal-dependent spatial learning and memory and repetitive behavior. Phosphoproteomic analysis revealed proteins regulating cytoskeletal dynamics are significantly dysregulated downstream of Par3. Mechanistically, we found Par3 deletion causes increased Rac1 activation and dysregulated microtubule dynamics through CAMSAP2. Together, our data reveal an unexpected role for Par3 as a molecular gatekeeper in regulating the pool of immature dendritic spines, a rate-limiting step of learning and memory, through modulating Rac1 activation and microtubule dynamics in vivo.
Collapse
Affiliation(s)
- Mikayla M. Voglewede
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Elif Naz Ozsen
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Noah Ivak
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Matteo Bernabucci
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
- The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Ruizhe Tang
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Miao Sun
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Zhiping P. Pang
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
- The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Huaye Zhang
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| |
Collapse
|
5
|
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
|
6
|
Zhuravskaya A, Yap K, Hamid F, Makeyev EV. Alternative splicing coupled to nonsense-mediated decay coordinates downregulation of non-neuronal genes in developing mouse neurons. Genome Biol 2024; 25:162. [PMID: 38902825 PMCID: PMC11188260 DOI: 10.1186/s13059-024-03305-8] [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: 07/08/2023] [Accepted: 06/07/2024] [Indexed: 06/22/2024] Open
Abstract
BACKGROUND The functional coupling between alternative pre-mRNA splicing (AS) and the mRNA quality control mechanism called nonsense-mediated decay (NMD) can modulate transcript abundance. Previous studies have identified several examples of such a regulation in developing neurons. However, the systems-level effects of AS-NMD in this context are poorly understood. RESULTS We developed an R package, factR2, which offers a comprehensive suite of AS-NMD analysis functions. Using this tool, we conducted a longitudinal analysis of gene expression in pluripotent stem cells undergoing induced neuronal differentiation. Our analysis uncovers hundreds of AS-NMD events with significant potential to regulate gene expression. Notably, this regulation is significantly overrepresented in specific functional groups of developmentally downregulated genes. Particularly strong association with gene downregulation is detected for alternative cassette exons stimulating NMD upon their inclusion into mature mRNA. By combining bioinformatic analyses with CRISPR/Cas9 genome editing and other experimental approaches we show that NMD-stimulating cassette exons regulated by the RNA-binding protein PTBP1 dampen the expression of their genes in developing neurons. We also provided evidence that the inclusion of NMD-stimulating cassette exons into mature mRNAs is temporally coordinated with NMD-independent gene repression mechanisms. CONCLUSIONS Our study provides an accessible workflow for the discovery and prioritization of AS-NMD targets. It further argues that the AS-NMD pathway plays a widespread role in developing neurons by facilitating the downregulation of functionally related non-neuronal genes.
Collapse
Affiliation(s)
- Anna Zhuravskaya
- Centre for Developmental Neurobiology, King's College London, London, SE1 1UL, UK
| | - Karen Yap
- Centre for Developmental Neurobiology, King's College London, London, SE1 1UL, UK
| | - Fursham Hamid
- Centre for Developmental Neurobiology, King's College London, London, SE1 1UL, UK.
| | - Eugene V Makeyev
- Centre for Developmental Neurobiology, King's College London, London, SE1 1UL, UK.
| |
Collapse
|
7
|
Oevel K, Hohensee S, Kumar A, Rosas-Brugada I, Bartolini F, Soykan T, Haucke V. Rho GTPase signaling and mDia facilitate endocytosis via presynaptic actin. eLife 2024; 12:RP92755. [PMID: 38502163 PMCID: PMC10950329 DOI: 10.7554/elife.92755] [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: 03/20/2024] Open
Abstract
Neurotransmission at synapses is mediated by the fusion and subsequent endocytosis of synaptic vesicle membranes. Actin has been suggested to be required for presynaptic endocytosis but the mechanisms that control actin polymerization and its mode of action within presynaptic nerve terminals remain poorly understood. We combine optical recordings of presynaptic membrane dynamics and ultrastructural analysis with genetic and pharmacological manipulations to demonstrate that presynaptic endocytosis is controlled by actin regulatory diaphanous-related formins mDia1/3 and Rho family GTPase signaling in mouse hippocampal neurons. We show that impaired presynaptic actin assembly in the near absence of mDia1/3 and reduced RhoA activity is partly compensated by hyperactivation of Rac1. Inhibition of Rac1 signaling further aggravates impaired presynaptic endocytosis elicited by loss of mDia1/3. Our data suggest that interdependent mDia1/3-Rho and Rac1 signaling pathways cooperatively act to facilitate synaptic vesicle endocytosis by controlling presynaptic F-actin.
Collapse
Affiliation(s)
- Kristine Oevel
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP)BerlinGermany
| | - Svea Hohensee
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP)BerlinGermany
| | - Atul Kumar
- Department of Pathology and Cell Biology, Columbia University Medical CenterNew York CityUnited States
| | | | - Francesca Bartolini
- Department of Pathology and Cell Biology, Columbia University Medical CenterNew York CityUnited States
| | - Tolga Soykan
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP)BerlinGermany
| | - Volker Haucke
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP)BerlinGermany
- Faculty of Biology, Chemistry, Pharmacy, Freie Universität BerlinBerlinGermany
- NeuroCure Cluster of Excellence, Charité Universitätsmedizin BerlinBerlinGermany
| |
Collapse
|
8
|
Li D, Ai S, Huang C, Liu ZH, Wang HL. Icariin rescues developmental BPA exposure induced spatial memory deficits in rats. Toxicol Appl Pharmacol 2024; 482:116776. [PMID: 38043803 DOI: 10.1016/j.taap.2023.116776] [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: 06/18/2023] [Revised: 11/23/2023] [Accepted: 11/27/2023] [Indexed: 12/05/2023]
Abstract
Bisphenol A (BPA) has been implicated in cognitive impairment. Icariin is the main active ingredient extracted from Epimedium Herb with protective function of nervous system. However, the potential therapeutic effects of Icariin on spatial memory deficits induced by developmental BPA exposure in Sprague-Dawley rats have not been investigated. This study investigated the therapeutic effect of Icariin (10 mg/kg/day, from postnatal day (PND) 21 to PND 60 by gavage) on spatial memory deficits in rat induced by developmental BPA exposure (1 mg/kg/day, from embryonic to PND 60), demonstrating that Icariin can markedly improve spatial memory in BPA-exposed rat. Furthermore, intra-gastric administration of Icariin could attenuate abnormal hippocampal cell dispersion and loss, improved the dendritic spine density and Nissl bodies. Moreover, Icariin reversed BPA induced reduction of frequency of miniature excitatory postsynaptic currents(mEPSC) and decrease of Vesicular glutamate transporter 1(VGlut1). Collectively, Icariin could effectively rescue BPA-induced spatial memory impairment in male rats by preventing cell loss and reduction of dendritic spines in the hippocampus. In addition, we also found that VGlut1 is a critical target in the repair of BPA-induced spatial memory by Icariin. Thus, Icariin may be a promising therapeutic agent to attenuate BPA-induced spatial memory deficits.
Collapse
Affiliation(s)
- Danyang Li
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui Province 230009, China
| | - Shu Ai
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui Province 230009, China
| | - Chengqing Huang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui Province 230009, China
| | - Zhi-Hua Liu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui Province 230009, China.
| | - Hui-Li Wang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui Province 230009, China.
| |
Collapse
|
9
|
Mesa MH, Garcia GC, Hoerndli FJ, McCabe KJ, Rangamani P. Spine apparatus modulates Ca 2+ in spines through spatial localization of sources and sinks. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.22.558941. [PMID: 37790389 PMCID: PMC10542496 DOI: 10.1101/2023.09.22.558941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Dendritic spines are small protrusions on dendrites in neurons and serve as sites of postsynaptic activity. Some of these spines contain smooth endoplasmic reticulum (SER), and sometimes an even further specialized SER known as the spine apparatus (SA). In this work, we developed a stochastic spatial model to investigate the role of the SER and the SA in modulating Ca 2+ dynamics. Using this model, we investigated how ryanodine receptor (RyR) localization, spine membrane geometry, and SER geometry can impact Ca 2+ transients in the spine and in the dendrite. Our simulations found that RyR opening is dependent on where it is localized in the SER and on the SER geometry. In order to maximize Ca 2+ in the dendrites (for activating clusters of spines and spine-spine communication), a laminar SA was favorable with RyRs localized in the neck region, closer to the dendrite. We also found that the presence of the SER without the laminar structure, coupled with RyR localization at the head, leads to higher Ca 2+ presence in the spine. These predictions serve as design principles for understanding how spines with an ER can regulate Ca 2+ dynamics differently from spines without ER through a combination of geometry and receptor localization. Highlights 1RyR opening in dendritic spine ER is location dependent and spine geometry dependent. Ca 2+ buffers and SERCA can buffer against runaway potentiation of spines even when CICR is activated. RyRs located towards the ER neck allow for more Ca 2+ to reach the dendrites. RyRs located towards the spine head are favorable for increased Ca 2+ in spines.
Collapse
|
10
|
Voglewede MM, Ozsen EN, Ivak N, Bernabucci M, Sun M, Pang ZP, Zhang H. Loss of the polarity protein Par3 promotes dendritic spine neoteny and enhances learning and memory. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.30.555530. [PMID: 37693426 PMCID: PMC10491238 DOI: 10.1101/2023.08.30.555530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
The Par3 polarity protein is critical for subcellular compartmentalization in different developmental processes. Variants of PARD3 , which encodes PAR3, are associated with intelligence and neurodevelopmental disorders. However, the role of Par3 in glutamatergic synapse formation and cognitive functions in vivo remains unknown. Here, we show that forebrain conditional knockout of Par3 leads to an increase in long, thin dendritic spines without significantly impacting mushroom spines in vivo . In addition, we observed a decrease in the amplitude of miniature excitatory postsynaptic currents. Surprisingly, loss of Par3 in vivo enhances hippocampal- dependent spatial learning. Phosphoproteomic analysis revealed proteins regulating cytoskeletal dynamics are significantly dysregulated downstream of Par3. Mechanistically, we found Par3 deletion causes increased activation of the Rac1 pathway. Together, our data reveal an unexpected role for Par3 as a molecular gatekeeper in regulating the pool of immature dendritic spines, a rate-limiting step of learning and memory, through modulating Rac1 activation in vivo .
Collapse
|
11
|
Perry S, Han Y, Qiu C, Chien C, Goel P, Nishimura S, Sajnani M, Schmid A, Sigrist SJ, Dickman D. A glutamate receptor C-tail recruits CaMKII to suppress retrograde homeostatic signaling. Nat Commun 2022; 13:7656. [PMID: 36496500 PMCID: PMC9741633 DOI: 10.1038/s41467-022-35417-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 12/02/2022] [Indexed: 12/13/2022] Open
Abstract
Presynaptic homeostatic plasticity (PHP) adaptively enhances neurotransmitter release following diminished postsynaptic glutamate receptor (GluR) functionality to maintain synaptic strength. While much is known about PHP expression mechanisms, postsynaptic induction remains enigmatic. For over 20 years, diminished postsynaptic Ca2+ influx was hypothesized to reduce CaMKII activity and enable retrograde PHP signaling at the Drosophila neuromuscular junction. Here, we have interrogated inductive signaling and find that active CaMKII colocalizes with and requires the GluRIIA receptor subunit. Next, we generated Ca2+-impermeable GluRs to reveal that both CaMKII activity and PHP induction are Ca2+-insensitive. Rather, a GluRIIA C-tail domain is necessary and sufficient to recruit active CaMKII. Finally, chimeric receptors demonstrate that the GluRIIA tail constitutively occludes retrograde homeostatic signaling by stabilizing active CaMKII. Thus, the physical loss of the GluRIIA tail is sensed, rather than reduced Ca2+, to enable retrograde PHP signaling, highlighting a unique, Ca2+-independent control mechanism for CaMKII in gating homeostatic plasticity.
Collapse
Affiliation(s)
- Sarah Perry
- Department of Neurobiology, University of Southern California, Los Angeles, CA, USA
| | - Yifu Han
- Department of Neurobiology, University of Southern California, Los Angeles, CA, USA
| | - Chengjie Qiu
- Department of Neurobiology, University of Southern California, Los Angeles, CA, USA
| | - Chun Chien
- Department of Neurobiology, University of Southern California, Los Angeles, CA, USA
| | - Pragya Goel
- Department of Neurobiology, University of Southern California, Los Angeles, CA, USA
| | - Samantha Nishimura
- Department of Neurobiology, University of Southern California, Los Angeles, CA, USA
| | - Manisha Sajnani
- Department of Neurobiology, University of Southern California, Los Angeles, CA, USA
| | - Andreas Schmid
- Institute for Biology/Genetics, Freie Universität Berlin, Takustraße 6, 14195, Berlin, Germany
- Faculty of Life Sciences, Albstadt-Sigmaringen University, Sigmaringen, Germany
| | - Stephan J Sigrist
- Institute for Biology/Genetics, Freie Universität Berlin, Takustraße 6, 14195, Berlin, Germany
- NeuroCure Cluster of Excellence, Charité Universitätsmedizin, Charitéplatz 1, 10117, Berlin, Germany
| | - Dion Dickman
- Department of Neurobiology, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
12
|
Subcellular partitioning of protein kinase activity revealed by functional kinome profiling. Sci Rep 2022; 12:17300. [PMID: 36243751 PMCID: PMC9569338 DOI: 10.1038/s41598-022-21026-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 09/21/2022] [Indexed: 01/10/2023] Open
Abstract
Protein kinases and their substrates form signaling networks partitioned across subcellular compartments to facilitate critical biological processes. While the subcellular roles of many individual kinases have been elucidated, a comprehensive assessment of the synaptic subkinome is lacking. Further, most studies of kinases focus on transcript, protein, and/or phospho-protein expression levels, providing an indirect measure of protein kinase activity. Prior work suggests that gene expression levels are not a good predictor of protein function. Thus, we assessed global serine/threonine protein kinase activity profiles in synaptosomal, nuclear, and cytosolic fractions from rat frontal cortex homogenate using peptide arrays. Comparisons made between fractions demonstrated differences in overall protein kinase activity. Upstream kinase analysis revealed a list of cognate kinases that were enriched in the synaptosomal fraction compared to the nuclear fraction. We identified many kinases in the synaptic fraction previously implicated in this compartment, while also identifying other kinases with little or no evidence for synaptic localization. Our results show the feasibility of assessing subcellular fractions with peptide activity arrays, as well as suggesting compartment specific activity profiles associated with established and novel kinases.
Collapse
|
13
|
Voglewede MM, Zhang H. Polarity proteins: Shaping dendritic spines and memory. Dev Biol 2022; 488:68-73. [PMID: 35580729 PMCID: PMC9953585 DOI: 10.1016/j.ydbio.2022.05.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 05/09/2022] [Accepted: 05/10/2022] [Indexed: 02/01/2023]
Abstract
The morphogenesis and plasticity of dendritic spines are associated with synaptic strength, learning, and memory. Dendritic spines are highly compartmentalized structures, which makes proteins involved in cellular polarization and membrane compartmentalization likely candidates regulating their formation and maintenance. Indeed, recent studies suggest polarity proteins help form and maintain dendritic spines by compartmentalizing the spine neck and head. Here, we review emerging evidence that polarity proteins regulate dendritic spine plasticity and stability through the cytoskeleton, scaffolding molecules, and signaling molecules. We specifically analyze various polarity complexes known to contribute to different forms of cell polarization processes and examine the essential conceptual context linking these groups of polarity proteins to dendritic spine morphogenesis, plasticity, and cognitive functions.
Collapse
Affiliation(s)
| | - Huaye Zhang
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, 08854, USA.
| |
Collapse
|
14
|
Zhao F, Li B, Yang W, Ge T, Cui R. Brain-immune interaction mechanisms: Implications for cognitive dysfunction in psychiatric disorders. Cell Prolif 2022; 55:e13295. [PMID: 35860850 PMCID: PMC9528770 DOI: 10.1111/cpr.13295] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 04/28/2022] [Accepted: 06/07/2022] [Indexed: 12/15/2022] Open
Abstract
Objectives Cognitive dysfunction has been identified as a major symptom of a series of psychiatric disorders. Multidisciplinary studies have shown that cognitive dysfunction is monitored by a two‐way interaction between the neural and immune systems. However, the specific mechanisms of cognitive dysfunction in immune response and brain immune remain unclear. Materials and methods In this review, we summarized the relevant research to uncover our comprehension of the brain–immune interaction mechanisms underlying cognitive decline. Results The pathophysiological mechanisms of brain‐immune interactions in psychiatric‐based cognitive dysfunction involve several specific immune molecules and their associated signaling pathways, impairments in neural and synaptic plasticity, and the potential neuro‐immunological mechanism of stress. Conclusions Therefore, this review may provide a better theoretical basis for integrative therapeutic considerations for psychiatric disorders associated with cognitive dysfunction.
Collapse
Affiliation(s)
- Fangyi Zhao
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Bingjin Li
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Wei Yang
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Tongtong Ge
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Ranji Cui
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
15
|
Lim D, Kim D, Um JW, Ko J. Reassessing synaptic adhesion pathways. Trends Neurosci 2022; 45:517-528. [DOI: 10.1016/j.tins.2022.04.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 04/12/2022] [Accepted: 04/19/2022] [Indexed: 01/19/2023]
|
16
|
Yao M, Meng M, Yang X, Wang S, Zhang H, Zhang F, Shi L, Zhang Y, Zhang X, Xu Z. POSH regulates assembly of the NMDAR/PSD-95/Shank complex and synaptic function. Cell Rep 2022; 39:110642. [PMID: 35385725 DOI: 10.1016/j.celrep.2022.110642] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/01/2022] [Accepted: 03/16/2022] [Indexed: 11/03/2022] Open
Abstract
Mutation or disruption of the Shank/ProSAP family of genes is a high risk factor for autism spectrum disorders (ASDs) and intellectual disability. N-methyl-D-aspartate glutamate receptor (NMDAR) dysfunction contributes to the development of autism-like behaviors. However, the molecular mechanism of Shank-mediated NMDAR modulation is still not clear. Here, we show that the scaffold protein plenty of SH3s (POSH) directly interacts with two other scaffold proteins, PSD95 and SHANK2/3, at excitatory synapses. In POSH conditional knockout (cKO) mice, normal synaptic clustering of NMDAR/PSD-95/SHANK complex is disrupted, accompanied by abnormal dendritic spine development and glutamatergic transmission in hippocampal neurons. POSH cKO mice display profound autism-like behaviors, including impairments in social interactions, social communication, repetitive behaviors, and deficits in learning and memory. Thus, POSH clusters at the postsynaptic density (PSD) with PSD-95 and SHANK2/3 and plays important roles in the signaling mechanisms of the NMDAR/PSD-95/POSH/SHANK complex as well as in spine development and brain function.
Collapse
Affiliation(s)
- Minghui Yao
- State Key Laboratory of Molecular Developmental Biology, CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100101, China.
| | - Meizhen Meng
- State Key Laboratory of Cognitive Neuroscience and Learning, IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China
| | - Xiyu Yang
- State Key Laboratory of Cognitive Neuroscience and Learning, IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China
| | - Shuo Wang
- State Key Laboratory of Molecular Developmental Biology, CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100101, China
| | - Hongsheng Zhang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, China
| | - Feng Zhang
- State Key Laboratory of Molecular Developmental Biology, CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100101, China
| | - Lei Shi
- State Key Laboratory of Molecular Developmental Biology, CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yongqing Zhang
- State Key Laboratory of Molecular Developmental Biology, CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100101, China
| | - Xiaohui Zhang
- State Key Laboratory of Cognitive Neuroscience and Learning, IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China.
| | - Zhiheng Xu
- State Key Laboratory of Molecular Developmental Biology, CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100101, China; Parkinson's Disease Center, Beijing Institute for Brain Disorders, Beijing 100101, China.
| |
Collapse
|
17
|
Tazerart S, Blanchard MG, Miranda-Rottmann S, Mitchell DE, Navea Pina B, Thomas CI, Kamasawa N, Araya R. Selective activation of BK channels in small-headed dendritic spines suppresses excitatory postsynaptic potentials. J Physiol 2022; 600:2165-2187. [PMID: 35194785 DOI: 10.1113/jp282303] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 02/14/2022] [Indexed: 12/22/2022] Open
Abstract
Dendritic spines are the main receptacles of excitatory information in the brain. Their particular morphology, with a small head connected to the dendrite by a slender neck, has inspired theoretical and experimental work to understand how these structural features affect the processing, storage and integration of synaptic inputs in pyramidal neurons (PNs). The activation of glutamate receptors in spines triggers a large voltage change as well as calcium signals at the spine head. Thus, voltage-gated and calcium-activated potassium channels located in the spine head likely play a key role in synaptic transmission. Here we study the presence and function of large conductance calcium-activated potassium (BK) channels in spines from layer 5 PNs. We found that BK channels are localized to dendrites and spines regardless of their size, but their activity can only be detected in spines with small head volumes (≤0.09 μm3 ), which reduces the amplitude of two-photon uncaging excitatory postsynaptic potentials recorded at the soma. In addition, we found that calcium signals in spines with small head volumes are significantly larger than those observed in spines with larger head volumes. In accordance with our experimental data, numerical simulations predict that synaptic inputs impinging onto spines with small head volumes generate voltage responses and calcium signals within the spine head itself that are significantly larger than those observed in spines with larger head volumes, which are sufficient to activate spine BK channels. These results show that BK channels are selectively activated in small-headed spines, suggesting a new level of dendritic spine-mediated regulation of synaptic processing, integration and plasticity in cortical PNs. KEY POINTS: BK channels are expressed in the visual cortex and layer 5 pyramidal neuron somata, dendrites and spines regardless of their size. BK channels are selectively activated in small-headed spines (≤0.09 μm3 ), which reduces the amplitude of two-photon (2P) uncaging excitatory postsynaptic potentials (EPSPs) recorded at the soma. Two-photon imaging revealed that intracellular calcium responses in the head of 2P-activated spines are significantly larger in small-headed spines (≤0.09 μm3 ) than in spines with larger head volumes. In accordance with our experimental data, numerical simulations showed that synaptic inputs impinging onto spines with small head volumes (≤0.09 μm3 ) generate voltage responses and calcium signals within the spine head itself that are significantly larger than those observed in spines with larger head volumes, sufficient to activate spine BK channels and suppress EPSPs.
Collapse
Affiliation(s)
- Sabrina Tazerart
- Département de Neurosciences, Université de Montréal, Montréal, Canada.,The CHU Sainte-Justine Research Center, Montréal, Canada
| | - Maxime G Blanchard
- Département de Neurosciences, Université de Montréal, Montréal, Canada.,The CHU Sainte-Justine Research Center, Montréal, Canada
| | - Soledad Miranda-Rottmann
- Département de Neurosciences, Université de Montréal, Montréal, Canada.,The CHU Sainte-Justine Research Center, Montréal, Canada
| | - Diana E Mitchell
- Département de Neurosciences, Université de Montréal, Montréal, Canada.,The CHU Sainte-Justine Research Center, Montréal, Canada
| | - Bruno Navea Pina
- Département de Neurosciences, Université de Montréal, Montréal, Canada.,The CHU Sainte-Justine Research Center, Montréal, Canada
| | - Connon I Thomas
- The Imaging Center and Electron Microscopy Core Facility, Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
| | - Naomi Kamasawa
- The Imaging Center and Electron Microscopy Core Facility, Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
| | - Roberto Araya
- Département de Neurosciences, Université de Montréal, Montréal, Canada.,The CHU Sainte-Justine Research Center, Montréal, Canada
| |
Collapse
|
18
|
Tsubo T. Analysis of the mechanism of synaptic integration focusing on the charge held in the spine. Biophys Physicobiol 2022; 18:290-304. [PMID: 35004103 PMCID: PMC8685514 DOI: 10.2142/biophysico.bppb-v18.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 11/30/2021] [Indexed: 12/01/2022] Open
Abstract
Successful synaptic integration is said to require that multiple excitatory postsynaptic potentials (EPSPs) occur almost simultaneously over a short period of time, so that they overlap and increase. However, if brain function is based on a chain of successful synaptic integrations, then constraints on the spacing of multiple EPSP generation must be released to allow for a higher probability of successful synaptic integration. This paper demonstrates that Ca2+ ions retained in spines after EPSP generation polarize spine neck fluid and dendritic fluid as a dielectric medium, that polarization is transmitted through dendrites to the cell body (soma), that polarization is enhanced by the addition of polarization from each spine, and that I propose that synaptic integration is successful when the membrane potential, as determined by the enhanced polarization and membrane capacitance, reaches the threshold of voltage-gated Na+ channels. Furthermore, the approach taken in this study suggests that a single neuron can integrate synapses for many combinations of synaptic inputs, that successful synaptic integration depends on spine neck capacitance and spine head size, and that spines farther from the soma are able to contribute to successful synaptic integration, and led to the elucidation of a number of important issues, including the fact that inhibitory post-synapses on dendrites suppress s effectively synaptic integration.
Collapse
Affiliation(s)
- Takayoshi Tsubo
- Brain Basic Function Laboratory, Hachioji, Tokyo 192-0914, Japan
| |
Collapse
|
19
|
Kourosh-Arami M, Hosseini N, Komaki A. Brain is modulated by neuronal plasticity during postnatal development. J Physiol Sci 2021; 71:34. [PMID: 34789147 PMCID: PMC10716960 DOI: 10.1186/s12576-021-00819-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 10/27/2021] [Indexed: 11/10/2022]
Abstract
Neuroplasticity is referred to the ability of the nervous system to change its structure or functions as a result of former stimuli. It is a plausible mechanism underlying a dynamic brain through adaptation processes of neural structure and activity patterns. Nevertheless, it is still unclear how the plastic neural systems achieve and maintain their equilibrium. Additionally, the alterations of balanced brain dynamics under different plasticity rules have not been explored either. Therefore, the present article primarily aims to review recent research studies regarding homosynaptic and heterosynaptic neuroplasticity characterized by the manipulation of excitatory and inhibitory synaptic inputs. Moreover, it attempts to understand different mechanisms related to the main forms of synaptic plasticity at the excitatory and inhibitory synapses during the brain development processes. Hence, this study comprised surveying those articles published since 1988 and available through PubMed, Google Scholar and science direct databases on a keyword-based search paradigm. All in all, the study results presented extensive and corroborative pieces of evidence for the main types of plasticity, including the long-term potentiation (LTP) and long-term depression (LTD) of the excitatory and inhibitory postsynaptic potentials (EPSPs and IPSPs).
Collapse
Affiliation(s)
- Masoumeh Kourosh-Arami
- Department of Neuroscience, School of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Nasrin Hosseini
- Neuroscience Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Alireza Komaki
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
20
|
Niño SA, Vázquez-Hernández N, Arevalo-Villalobos J, Chi-Ahumada E, Martín-Amaya-Barajas FL, Díaz-Cintra S, Martel-Gallegos G, González-Burgos I, Jiménez-Capdeville ME. Cortical Synaptic Reorganization Under Chronic Arsenic Exposure. Neurotox Res 2021; 39:1970-1980. [PMID: 34533753 DOI: 10.1007/s12640-021-00409-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 08/02/2021] [Accepted: 08/30/2021] [Indexed: 12/24/2022]
Abstract
There is solid epidemiological evidence that arsenic exposure leads to cognitive impairment, while experimental work supports the hypothesis that it also contributes to neurodegeneration. Energy deficit, oxidative stress, demyelination, and defective neurotransmission are demonstrated arsenic effects, but it remains unclear whether synaptic structure is also affected. Employing both a triple-transgenic Alzheimer's disease model and Wistar rats, the cortical microstructure and synapses were analyzed under chronic arsenic exposure. Male animals were studied at 2 and 4 months of age, after exposure to 3 ppm sodium arsenite in drinking water during gestation, lactation, and postnatal development. Through nuclear magnetic resonance, diffusion-weighted images were acquired and anisotropy (integrity; FA) and apparent diffusion coefficient (dispersion degree; ADC) metrics were derived. Postsynaptic density protein and synaptophysin were analyzed by means of immunoblot and immunohistochemistry, while dendritic spine density and morphology of cortical pyramidal neurons were quantified after Golgi staining. A structural reorganization of the cortex was evidenced through high-ADC and low-FA values in the exposed group. Similar changes in synaptic protein levels in the 2 models suggest a decreased synaptic connectivity at 4 months of age. An abnormal dendritic arborization was observed at 4 months of age, after increased spine density at 2 months. These findings demonstrate alterations of cortical synaptic connectivity and microstructure associated to arsenic exposure appearing in young rodents and adults, and these subtle and non-adaptive plastic changes in dendritic spines and in synaptic markers may further progress to the degeneration observed at older ages.
Collapse
Affiliation(s)
- Sandra A Niño
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| | - Nallely Vázquez-Hernández
- División de Neurociencias, Centro de Investigación Biomédica de Occidente, IMSS. Guadalajara, Jalisco, Mexico
| | - Jaime Arevalo-Villalobos
- Facultad de Agronomía y Veterinaria, Universidad Autónoma de San Luis Potosí, Soledad de Graciano Sánchez, San Luis Potosí, Mexico
| | - Erika Chi-Ahumada
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| | | | - Sofía Díaz-Cintra
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Mexico
| | - Guadalupe Martel-Gallegos
- Laboratorio de Biomedicina, Unidad Académica Multidisciplinaria Zona Media, Universidad Autónoma de San Luis Potosí, Rio Verde, San Luis Potosí, Mexico
| | - Ignacio González-Burgos
- División de Neurociencias, Centro de Investigación Biomédica de Occidente, IMSS. Guadalajara, Jalisco, Mexico
| | - María E Jiménez-Capdeville
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico.
| |
Collapse
|
21
|
Xu D, Yang P, Yang ZJ, Li QG, Ouyang YT, Yu T, Shangguan JH, Wan YY, Jiang LP, Qu XH, Han XJ. Blockage of Drp1 phosphorylation at Ser579 protects neurons against Aβ 1‑42‑induced degeneration. Mol Med Rep 2021; 24:657. [PMID: 34278489 PMCID: PMC8299198 DOI: 10.3892/mmr.2021.12296] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 06/24/2021] [Indexed: 01/13/2023] Open
Abstract
Alzheimer's disease (AD), one of the most common types of chronic neurodegenerative diseases, is pathologically characterized by the formation of amyloid β (Aβ) peptide-containing plaques and neurofibrillary tangles. Among Aβ peptides, Aβ1–42 induces neuronal toxicity and neurodegeneration. In our previous studies, Cdk5 was found to regulate Aβ1–42-induced mitochondrial fission via the phosphorylation of dynamin-related protein 1 (Drp1) at Ser579. However, whether blockage of Drp1 phosphorylation at Ser579 protects neurons against Aβ1–42-induced degeneration remains to be elucidated. Thus, the aim the present study was to examine the effect of mutant Drp1-S579A on neurodegeneration and its underlying mechanism. First, the phosphorylation-defect (phospho-defect) mutant, Lenti-Drp1-S579A was constructed. Phospho-defect Drp1-S579A expression was detected in primary cultures of mouse cortical neurons infected with Lenti-Drp1-S579A using western blotting and it was found to successfully attenuate the phosphorylation of endogenous Drp1 at Ser579. In primary neuronal cultures, the neuronal processes were evaluated under microscopy. Treatment with 10 µM Aβ1–42 significantly decreased dendritic density and length, spine outgrowth and synapse number. As expected, infection of neurons with Lenti-Drp1-S579A efficiently alleviated the inhibitory effect of Aβ1–42 on neurite outgrowth and synapse density. In addition, infection with Lenti-Drp1-S579A abolished the cleavage of caspase-3 and apoptosis in neurons exposed to Aβ1–42. Thus, the current data demonstrated that blockage of Drp1 phosphorylation at Ser579 may be an effective strategy to protect neurons against Aβ1–42-induced degeneration and apoptosis. These findings underline the therapeutic potential of targeting Drp1 in the treatment of AD.
Collapse
Affiliation(s)
- Dan Xu
- Institute of Geriatrics, Affiliated People's Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Ping Yang
- Department of Neurology, Affiliated People's Hospital of Nanchang University, Jiangxi 330006, P.R. China
| | - Zhang-Jian Yang
- Department of Pharmacology, School of Pharmaceutical Science, Nanchang University, Jiangxi 330006, P.R. China
| | - Qiu-Gen Li
- Institute of Geriatrics, Affiliated People's Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Ye-Tong Ouyang
- Department of Neurology, Affiliated People's Hospital of Nanchang University, Jiangxi 330006, P.R. China
| | - Ting Yu
- Department of Neurology, Affiliated People's Hospital of Nanchang University, Jiangxi 330006, P.R. China
| | - Jian-Hui Shangguan
- Department of Neurology, Affiliated People's Hospital of Nanchang University, Jiangxi 330006, P.R. China
| | - Yu-Ying Wan
- Department of Intra‑Hospital Infection Management, The Second Affiliated Hospital of Nanchang University, Jiangxi 330006, P.R. China
| | - Li-Ping Jiang
- Department of Pharmacology, School of Pharmaceutical Science, Nanchang University, Jiangxi 330006, P.R. China
| | - Xin-Hui Qu
- Institute of Geriatrics, Affiliated People's Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Xiao-Jian Han
- Institute of Geriatrics, Affiliated People's Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
22
|
Vallejo D, Lindsay CB, González-Billault C, Inestrosa NC. Wnt5a modulates dendritic spine dynamics through the regulation of Cofilin via small Rho GTPase activity in hippocampal neurons. J Neurochem 2021; 158:673-693. [PMID: 34107066 DOI: 10.1111/jnc.15448] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 06/03/2021] [Accepted: 06/07/2021] [Indexed: 01/21/2023]
Abstract
Dendritic spines are small, actin-rich protrusions that act as the receiving sites of most excitatory inputs in the central nervous system. The remodeling of the synapse architecture is mediated by actin cytoskeleton dynamics, a process precisely regulated by the small Rho GTPase family. Wnt ligands exert their presynaptic and postsynaptic effects during formation and consolidation of the synaptic structure. Specifically, Wnt5a has been identified as an indispensable synaptogenic factor for the regulation and organization of the postsynaptic side; however, the molecular mechanisms through which Wnt5a induces morphological changes resulting from actin cytoskeleton dynamics within dendritic spines remain unclear. In this work, we employ primary rat hippocampal cultures and HT22 murine hippocampal neuronal cell models, molecular and pharmacological tools, and fluorescence microscopy (laser confocal and epifluorescence) to define the Wnt5a-induced molecular signaling involved in postsynaptic remodeling mediated via the regulation of the small Rho GTPase family. We report that Wnt5a differentially regulates the phosphorylation of Cofilin in neurons through both Ras-related C3 botulinum toxin substrate 1 and cell division cycle 42 depending on the subcellular compartment and the extracellular calcium levels. Additionally, we demonstrate that Wnt5a increases the density of dendritic spines and promotes their maturation via Ras-related C3 botulinum toxin substrate 1. Accordingly, we find that Wnt5a requires the combined activation of small Rho GTPases to increase the levels of filamentous actin, thus promoting the stability of actin filaments. Altogether, these results provide evidence for a new mechanism by which Wnt5a may target actin dynamics, thereby regulating the subsequent morphological changes in dendritic spine architecture.
Collapse
Affiliation(s)
- Daniela Vallejo
- Centro de Envejecimiento y Regeneración (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Carolina B Lindsay
- Centro de Envejecimiento y Regeneración (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Christian González-Billault
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile.,Geroscience Center for Brain Health and Metabolism (GERO), Santiago, Chile.,The Buck Institute for Research on Aging, Novato, CA, USA
| | - Nibaldo C Inestrosa
- Centro de Envejecimiento y Regeneración (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile
| |
Collapse
|
23
|
Alimohamadi H, Bell MK, Halpain S, Rangamani P. Mechanical Principles Governing the Shapes of Dendritic Spines. Front Physiol 2021; 12:657074. [PMID: 34220531 PMCID: PMC8242199 DOI: 10.3389/fphys.2021.657074] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 05/13/2021] [Indexed: 02/04/2023] Open
Abstract
Dendritic spines are small, bulbous protrusions along the dendrites of neurons and are sites of excitatory postsynaptic activity. The morphology of spines has been implicated in their function in synaptic plasticity and their shapes have been well-characterized, but the potential mechanics underlying their shape development and maintenance have not yet been fully understood. In this work, we explore the mechanical principles that could underlie specific shapes using a minimal biophysical model of membrane-actin interactions. Using this model, we first identify the possible force regimes that give rise to the classic spine shapes-stubby, filopodia, thin, and mushroom-shaped spines. We also use this model to investigate how the spine neck might be stabilized using periodic rings of actin or associated proteins. Finally, we use this model to predict that the cooperation between force generation and ring structures can regulate the energy landscape of spine shapes across a wide range of tensions. Thus, our study provides insights into how mechanical aspects of actin-mediated force generation and tension can play critical roles in spine shape maintenance.
Collapse
Affiliation(s)
- Haleh Alimohamadi
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA, United States
| | - Miriam K. Bell
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA, United States
| | - Shelley Halpain
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, United States
- Section of Neurobiology, Division of Biological Sciences, University of California, San Diego, La Jolla, CA, United States
| | - Padmini Rangamani
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
24
|
Semaphorin3F Drives Dendritic Spine Pruning Through Rho-GTPase Signaling. Mol Neurobiol 2021; 58:3817-3834. [PMID: 33856648 DOI: 10.1007/s12035-021-02373-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 03/24/2021] [Indexed: 12/18/2022]
Abstract
Dendritic spines of cortical pyramidal neurons are initially overproduced then remodeled substantially in the adolescent brain to achieve appropriate excitatory balance in mature circuits. Here we investigated the molecular mechanism of developmental spine pruning by Semaphorin 3F (Sema3F) and its holoreceptor complex, which consists of immunoglobulin-class adhesion molecule NrCAM, Neuropilin-2 (Npn2), and PlexinA3 (PlexA3) signaling subunits. Structure-function studies of the NrCAM-Npn2 interface showed that NrCAM stabilizes binding between Npn2 and PlexA3 necessary for Sema3F-induced spine pruning. Using a mouse neuronal culture system, we identified a dual signaling pathway for Sema3F-induced pruning, which involves activation of Tiam1-Rac1-PAK1-3 -LIMK1/2-Cofilin1 and RhoA-ROCK1/2-Myosin II in dendritic spines. Inhibitors of actin remodeling impaired spine collapse in the cortical neurons. Elucidation of these pathways expands our understanding of critical events that sculpt neuronal networks and may provide insight into how interruptions to these pathways could lead to spine dysgenesis in diseases such as autism, bipolar disorder, and schizophrenia.
Collapse
|
25
|
Zhu Y, Uytiepo M, Bushong E, Haberl M, Beutter E, Scheiwe F, Zhang W, Chang L, Luu D, Chui B, Ellisman M, Maximov A. Nanoscale 3D EM reconstructions reveal intrinsic mechanisms of structural diversity of chemical synapses. Cell Rep 2021; 35:108953. [PMID: 33826888 PMCID: PMC8354523 DOI: 10.1016/j.celrep.2021.108953] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 01/21/2021] [Accepted: 03/15/2021] [Indexed: 10/25/2022] Open
Abstract
Chemical synapses of shared cellular origins have remarkably heterogeneous structures, but how this diversity is generated is unclear. Here, we use three-dimensional (3D) electron microscopy and artificial intelligence algorithms for image processing to reconstruct functional excitatory microcircuits in the mouse hippocampus and microcircuits in which neurotransmitter signaling is permanently suppressed with genetic tools throughout the lifespan. These nanoscale analyses reveal that experience is dispensable for morphogenesis of synapses with different geometric shapes and contents of membrane organelles and that arrangement of morphologically distinct connections in local networks is stochastic. Moreover, loss of activity increases the variability in sizes of opposed pre- and postsynaptic structures without disrupting their alignments, suggesting that inherently variable weights of naive connections become progressively matched with repetitive use. These results demonstrate that mechanisms for the structural diversity of neuronal synapses are intrinsic and provide insights into how circuits essential for memory storage assemble and integrate information.
Collapse
Affiliation(s)
- Yongchuan Zhu
- Department of Neuroscience, The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Marco Uytiepo
- Department of Neuroscience, The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Eric Bushong
- National Center for Microscopy and Imaging Research, University of California, San Diego, CA 92037, USA; Department of Neurosciences, University of California, San Diego, School of Medicine, La Jolla, CA 92037, USA
| | - Matthias Haberl
- National Center for Microscopy and Imaging Research, University of California, San Diego, CA 92037, USA; Department of Neurosciences, University of California, San Diego, School of Medicine, La Jolla, CA 92037, USA
| | - Elizabeth Beutter
- Department of Neuroscience, The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Frederieke Scheiwe
- Department of Neuroscience, The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Weiheng Zhang
- Department of Neuroscience, The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Lyanne Chang
- Department of Neuroscience, The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Danielle Luu
- Department of Neuroscience, The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Brandon Chui
- Department of Neuroscience, The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Mark Ellisman
- National Center for Microscopy and Imaging Research, University of California, San Diego, CA 92037, USA; Department of Neurosciences, University of California, San Diego, School of Medicine, La Jolla, CA 92037, USA.
| | - Anton Maximov
- Department of Neuroscience, The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
26
|
ADAP1/Centaurin-α1 Negatively Regulates Dendritic Spine Function and Memory Formation in the Hippocampus. eNeuro 2021; 8:ENEURO.0111-20.2020. [PMID: 33139322 PMCID: PMC7808333 DOI: 10.1523/eneuro.0111-20.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 09/25/2020] [Accepted: 10/01/2020] [Indexed: 12/27/2022] Open
Abstract
ADAP1/Centaurin-α1 (CentA1) functions as an Arf6 GTPase-activating protein highly enriched in the brain. Previous studies demonstrated the involvement of CentA1 in brain function as a regulator of dendritic differentiation and a potential mediator of Alzheimer’s disease (AD) pathogenesis. To better understand the neurobiological functions of CentA1 signaling in the brain, we developed Centa1 knock-out (KO) mice. The KO animals showed neither brain development nor synaptic ultrastructure deficits in the hippocampus. However, they exhibited significantly higher density and enhanced structural plasticity of dendritic spines in the CA1 region of the hippocampus compared with non-transgenic (NTG) littermates. Moreover, the deletion of Centa1 improved performance in the object-in-place (OIP) spatial memory task. These results suggest that CentA1 functions as a negative regulator of spine density and plasticity, and of hippocampus-dependent memory formation. Thus, CentA1 and its downstream signaling may serve as a potential therapeutic target to prevent memory decline associated with aging and brain disorders.
Collapse
|
27
|
Xie X, Mahmood SR, Gjorgjieva T, Percipalle P. Emerging roles of cytoskeletal proteins in regulating gene expression and genome organization during differentiation. Nucleus 2020; 11:53-65. [PMID: 32212905 PMCID: PMC7289583 DOI: 10.1080/19491034.2020.1742066] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
In the eukaryotic cell nucleus, cytoskeletal proteins are emerging as essential players in nuclear function. In particular, actin regulates chromatin as part of ATP-dependent chromatin remodeling complexes, it modulates transcription and it is incorporated into nascent ribonucleoprotein complexes, accompanying them from the site of transcription to polyribosomes. The nuclear actin pool is undistinguishable from the cytoplasmic one in terms of its ability to undergo polymerization and it has also been implicated in the dynamics of chromatin, regulating heterochromatin segregation at the nuclear lamina and maintaining heterochromatin levels in the nuclear interiors. One of the next frontiers is, therefore, to determine a possible involvement of nuclear actin in the functional architecture of the cell nucleus by regulating the hierarchical organization of chromatin and, thus, genome organization. Here, we discuss the repertoire of these potential actin functions and how they are likely to play a role in the context of cellular differentiation.
Collapse
Affiliation(s)
- Xin Xie
- Science Division, Biology Program, New York University Abu Dhabi (NYUAD), Abu Dhabi, United Arab Emirates
| | - S Raza Mahmood
- Science Division, Biology Program, New York University Abu Dhabi (NYUAD), Abu Dhabi, United Arab Emirates.,Department of Biology, New York University, New York, NY, USA
| | - Tamara Gjorgjieva
- Science Division, Biology Program, New York University Abu Dhabi (NYUAD), Abu Dhabi, United Arab Emirates
| | - Piergiorgio Percipalle
- Science Division, Biology Program, New York University Abu Dhabi (NYUAD), Abu Dhabi, United Arab Emirates.,Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| |
Collapse
|
28
|
Yap K, Drakew A, Smilovic D, Rietsche M, Paul MH, Vuksic M, Del Turco D, Deller T. The actin-modulating protein synaptopodin mediates long-term survival of dendritic spines. eLife 2020; 9:e62944. [PMID: 33275099 PMCID: PMC7717903 DOI: 10.7554/elife.62944] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 11/14/2020] [Indexed: 12/15/2022] Open
Abstract
Large spines are stable and important for memory trace formation. The majority of large spines also contains synaptopodin (SP), an actin-modulating and plasticity-related protein. Since SP stabilizes F-actin, we speculated that the presence of SP within large spines could explain their long lifetime. Indeed, using 2-photon time-lapse imaging of SP-transgenic granule cells in mouse organotypic tissue cultures we found that spines containing SP survived considerably longer than spines of equal size without SP. Of note, SP-positive (SP+) spines that underwent pruning first lost SP before disappearing. Whereas the survival time courses of SP+ spines followed conditional two-stage decay functions, SP-negative (SP-) spines and all spines of SP-deficient animals showed single-phase exponential decays. This was also the case following afferent denervation. These results implicate SP as a major regulator of long-term spine stability: SP clusters stabilize spines, and the presence of SP indicates spines of high stability.
Collapse
Affiliation(s)
- Kenrick Yap
- Institute of Clinical Neuroanatomy, Dr. Senckenberg Anatomy, Neuroscience Center, Goethe University FrankfurtFrankfurtGermany
| | - Alexander Drakew
- Institute of Clinical Neuroanatomy, Dr. Senckenberg Anatomy, Neuroscience Center, Goethe University FrankfurtFrankfurtGermany
| | - Dinko Smilovic
- Institute of Clinical Neuroanatomy, Dr. Senckenberg Anatomy, Neuroscience Center, Goethe University FrankfurtFrankfurtGermany
- Croatian Institute for Brain Research, School of Medicine, University of ZagrebZagrebCroatia
| | - Michael Rietsche
- Institute of Clinical Neuroanatomy, Dr. Senckenberg Anatomy, Neuroscience Center, Goethe University FrankfurtFrankfurtGermany
| | - Mandy H Paul
- Institute of Clinical Neuroanatomy, Dr. Senckenberg Anatomy, Neuroscience Center, Goethe University FrankfurtFrankfurtGermany
| | - Mario Vuksic
- Institute of Clinical Neuroanatomy, Dr. Senckenberg Anatomy, Neuroscience Center, Goethe University FrankfurtFrankfurtGermany
- Croatian Institute for Brain Research, School of Medicine, University of ZagrebZagrebCroatia
| | - Domenico Del Turco
- Institute of Clinical Neuroanatomy, Dr. Senckenberg Anatomy, Neuroscience Center, Goethe University FrankfurtFrankfurtGermany
| | - Thomas Deller
- Institute of Clinical Neuroanatomy, Dr. Senckenberg Anatomy, Neuroscience Center, Goethe University FrankfurtFrankfurtGermany
| |
Collapse
|
29
|
Antipsychotic Drugs Reverse MK801-Inhibited Cell Migration and F-actin Condensation by Modulating the Rho Signaling Pathway in B35 Cells. Behav Neurol 2020. [DOI: 10.1155/2020/4163274] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Background and Aim. MK801-induced psychotic symptoms and also the Ras homolog family member A (RhoA) expression and cell division control protein 42 (cdc42) mRNA modulation in the rat brain have been investigated. Antipsychotic drugs (APDs) have been reported to induce Rho GDP-dissociation inhibitor (RhoGDI) pathway regulation related to cytoskeleton reorganization in neuronal cells. It will be necessary to clarify the effects of APDs on MK801-induced RhoGDI signaling regulation in neuronal cells. Methods. B35 neuronal cells were treated with MK801 for 7 days then treated with MK801 in combination with haloperidol or clozapine for a further 7 days. Cell migration, F-actin condensation, and RhoGDI signaling regulation were examined to investigate the regulatory effects of MK801, haloperidol, and clozapine in B35 neuronal cells. Results. MK801 reduced B35 cell migration, whereas both haloperidol and clozapine reversed the reduction in cell migration induced by MK801. Haloperidol and clozapine restored F-actin condensation after it was diminished by MK801 in B35 cell nuclei. MK801 increased the RhoGDI1 and RhoA expression, which was diminished by the addition of haloperidol and clozapine. MK801 reduced the CDC42 expression, which was restored by haloperidol and clozapine. MK801 reduced the Rho-associated coiled-coil containing protein kinase 1 (ROCK1), profilin1 (PFN1), and neuronal Wiskott–Aldrich Syndrome protein (N-WASP) expression, which was further reduced by haloperidol and clozapine. MK801 also increased the phosphorylated myosin light chain 2 (p-MLC2), postsynaptic density protein 95 (PSD-95), and c-jun expression, which was decreased by haloperidol and clozapine. p21 (RAC1-) activated kinase 1 (PAK1) expression was not affected by MK801.
Collapse
|
30
|
Zagrebelsky M, Tacke C, Korte M. BDNF signaling during the lifetime of dendritic spines. Cell Tissue Res 2020; 382:185-199. [PMID: 32537724 PMCID: PMC7529616 DOI: 10.1007/s00441-020-03226-5] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 04/27/2020] [Indexed: 12/13/2022]
Abstract
Dendritic spines are tiny membrane specialization forming the postsynaptic part of most excitatory synapses. They have been suggested to play a crucial role in regulating synaptic transmission during development and in adult learning processes. Changes in their number, size, and shape are correlated with processes of structural synaptic plasticity and learning and memory and also with neurodegenerative diseases, when spines are lost. Thus, their alterations can correlate with neuronal homeostasis, but also with dysfunction in several neurological disorders characterized by cognitive impairment. Therefore, it is important to understand how different stages in the life of a dendritic spine, including formation, maturation, and plasticity, are strictly regulated. In this context, brain-derived neurotrophic factor (BDNF), belonging to the NGF-neurotrophin family, is among the most intensively investigated molecule. This review would like to report the current knowledge regarding the role of BDNF in regulating dendritic spine number, structure, and plasticity concentrating especially on its signaling via its two often functionally antagonistic receptors, TrkB and p75NTR. In addition, we point out a series of open points in which, while the role of BDNF signaling is extremely likely conclusive, evidence is still missing.
Collapse
Affiliation(s)
- Marta Zagrebelsky
- Division of Cellular Neurobiology, Zoological Institute, TU Braunschweig, Spielmannstr 7, 38106, Braunschweig, Germany.
- Helmholtz Centre for Infection Research, AG NIND, Inhoffenstr. 7, D-38124, Braunschweig, Germany.
| | - Charlotte Tacke
- Division of Cellular Neurobiology, Zoological Institute, TU Braunschweig, Spielmannstr 7, 38106, Braunschweig, Germany
| | - Martin Korte
- Division of Cellular Neurobiology, Zoological Institute, TU Braunschweig, Spielmannstr 7, 38106, Braunschweig, Germany.
- Helmholtz Centre for Infection Research, AG NIND, Inhoffenstr. 7, D-38124, Braunschweig, Germany.
| |
Collapse
|
31
|
Frey S, Schieweck R, Forné I, Imhof A, Straub T, Popper B, Kiebler MA. Physical Activity Dynamically Regulates the Hippocampal Proteome along the Dorso-Ventral Axis. Int J Mol Sci 2020; 21:E3501. [PMID: 32429128 PMCID: PMC7278950 DOI: 10.3390/ijms21103501] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 05/13/2020] [Accepted: 05/14/2020] [Indexed: 01/19/2023] Open
Abstract
The hippocampus is central for higher cognition and emotions. In patients suffering from neuropsychiatric or neurodegenerative diseases, hippocampal signaling is altered causing cognitive defects. Thus, therapeutic approaches aim at improving cognition by targeting the hippocampus. Enhanced physical activity (EPA) improves cognition in rodents and humans. A systematic screen, however, for expression changes in the hippocampus along the dorso-ventral axis is missing, which is a prerequisite for understanding molecular mechanisms. Here, we exploited label free mass spectrometry to detect proteomic changes in the hippocampus of male mice upon voluntary wheel running. To identify regional differences, we examined dorsal and ventral CA1, CA3 and dentate gyrus hippocampal subregions. We found metabolic enzymes and actin binding proteins, such as RhoA, being upregulated in the hippocampus upon EPA suggesting a coordination between metabolism and cytoskeleton remodeling; two pathways essential for synaptic plasticity. Strikingly, dorsal and ventral hippocampal subregions respond differentially to EPA. Together, our results provide new insight into proteomic adaptations driven by physical activity in mice. In addition, our results suggest that dorsal and ventral hippocampus, as well as hippocampal subregions themselves, contribute differently to this process. Our study therefore provides an important resource for studying hippocampal subregion diversity in response to EPA.
Collapse
Affiliation(s)
- Surina Frey
- Department for Cell Biology & Anatomy, Biomedical Center (BMC), Medical Faculty, Ludwig-Maximilians- University, 82152 Planegg-Martinsried, Germany; (S.F.); (R.S.)
| | - Rico Schieweck
- Department for Cell Biology & Anatomy, Biomedical Center (BMC), Medical Faculty, Ludwig-Maximilians- University, 82152 Planegg-Martinsried, Germany; (S.F.); (R.S.)
| | - Ignasi Forné
- Department for Molecular Biology (protein analysis unit), Biomedical Center (BMC), Ludwig-Maximilians-University, 82152 Planegg-Martinsried, Germany; (I.F.); (A.I.)
| | - Axel Imhof
- Department for Molecular Biology (protein analysis unit), Biomedical Center (BMC), Ludwig-Maximilians-University, 82152 Planegg-Martinsried, Germany; (I.F.); (A.I.)
| | - Tobias Straub
- Department for Molecular Biology (Core facility bioinformatics), Biomedical Center (BMC), Ludwig-Maximilians-University, 82152 Planegg-Martinsried, Germany;
| | - Bastian Popper
- Department for Cell Biology & Anatomy, Biomedical Center (BMC), Medical Faculty, Ludwig-Maximilians- University, 82152 Planegg-Martinsried, Germany; (S.F.); (R.S.)
- Core Facility Animal Models, Biomedical Center (BMC), Medical Faculty, Ludwig-Maximilians-University, 82152 Munich, Germany
- Institute of Pathology, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Michael A. Kiebler
- Department for Cell Biology & Anatomy, Biomedical Center (BMC), Medical Faculty, Ludwig-Maximilians- University, 82152 Planegg-Martinsried, Germany; (S.F.); (R.S.)
| |
Collapse
|
32
|
Mikuni T, Uchigashima M. Methodological approaches to understand the molecular mechanism of structural plasticity of dendritic spines. Eur J Neurosci 2020; 54:6902-6911. [PMID: 32248570 DOI: 10.1111/ejn.14734] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 03/23/2020] [Accepted: 03/25/2020] [Indexed: 12/16/2022]
Abstract
Dendritic spines are tiny protrusions emanating from the neuronal dendrites, typically housing single excitatory postsynapses. Structural plasticity of dendritic spines is considered to be essential for synaptic functional plasticity and also reorganization of neural circuits during learning and memory. Structural plasticity of spines is mediated by complex biochemical signaling with various spatial and temporal scales. A variety of methods based on pharmacological, genetic, molecular, imaging and optical approaches has been developed and applied to dissect the complex signal transduction pathways. In this review, we overview both conventional and new methodological approaches to identify, monitor and manipulate key molecules for structural plasticity of dendritic spines, ultimately aiming to understand the molecular mechanism of learning and memory in behaving animals.
Collapse
Affiliation(s)
- Takayasu Mikuni
- Department of Cellular Neuropathology, Brain Research Institute, Niigata University, Niigata, Japan.,Japan Science and Technology Agency, PRESTO, Kawaguchi, Japan
| | - Motokazu Uchigashima
- Department of Cellular Neuropathology, Brain Research Institute, Niigata University, Niigata, Japan
| |
Collapse
|
33
|
Birefringence Changes of Dendrites in Mouse Hippocampal Slices Revealed with Polarizing Microscopy. Biophys J 2020; 118:2366-2384. [PMID: 32294480 DOI: 10.1016/j.bpj.2020.03.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 02/20/2020] [Accepted: 03/06/2020] [Indexed: 11/23/2022] Open
Abstract
Intrinsic optical signal (IOS) imaging has been widely used to map the patterns of brain activity in vivo in a label-free manner. Traditional IOS refers to changes in light transmission, absorption, reflectance, and scattering of the brain tissue. Here, we use polarized light for IOS imaging to monitor structural changes of cellular and subcellular architectures due to their neuronal activity in isolated brain slices. To reveal fast spatiotemporal changes of subcellular structures associated with neuronal activity, we developed the instantaneous polarized light microscope (PolScope), which allows us to observe birefringence changes in neuronal cells and tissues while stimulating neuronal activity. The instantaneous PolScope records changes in transmission, birefringence, and slow axis orientation in tissue at a high spatial and temporal resolution using a single camera exposure. These capabilities enabled us to correlate polarization-sensitive IOS with traditional IOS on the same preparations. We detected reproducible spatiotemporal changes in both IOSs at the stratum radiatum in mouse hippocampal slices evoked by electrical stimulation at Schaffer collaterals. Upon stimulation, changes in traditional IOS signals were broadly uniform across the area, whereas birefringence imaging revealed local variations not seen in traditional IOS. Locations with high resting birefringence produced larger stimulation-evoked birefringence changes than those produced at low resting birefringence. Local application of glutamate to the synaptic region in CA1 induced an increase in both transmittance and birefringence signals. Blocking synaptic transmission with inhibitors CNQX (for AMPA-type glutamate receptor) and D-APV (for NMDA-type glutamate receptor) reduced the peak amplitude of the optical signals. Changes in both IOSs were enhanced by an inhibitor of the membranous glutamate transporter, DL-TBOA. Our results indicate that the detection of activity-induced structural changes of the subcellular architecture in dendrites is possible in a label-free manner.
Collapse
|
34
|
Cui Y, Cao K, Lin H, Cui S, Shen C, Wen W, Mo H, Dong Z, Bai S, Yang L, Shi Y, Zhang R. Early-Life Stress Induces Depression-Like Behavior and Synaptic-Plasticity Changes in a Maternal Separation Rat Model: Gender Difference and Metabolomics Study. Front Pharmacol 2020; 11:102. [PMID: 32174832 PMCID: PMC7055479 DOI: 10.3389/fphar.2020.00102] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 01/28/2020] [Indexed: 12/18/2022] Open
Abstract
More than 300 million people suffer from depressive disorders globally. People under early-life stress (ELS) are reportedly vulnerable to depression in their adulthood, and synaptic plasticity can be the molecular mechanism underlying such depression. Herein, we simulated ELS by using a maternal separation (MS) model and evaluated the behavior of Sprague-Dawley (SD) rats in adulthood through behavioral examination, including sucrose preference, forced swimming, and open-field tests. The behavior tests showed that SD rats in the MS group were more susceptible to depression- and anxiety-like behaviors than did the non-MS (NMS) group. Nissl staining analysis indicated a significant reduction in the number of neurons at the prefrontal cortex and hippocampus, including the CA1, CA2, CA3, and DG regions of SD rats in the MS group. Immunohistochemistry results showed that the percentages of synaptophysin-positive area in the prefrontal cortex and hippocampus (including the CA1, CA2, CA3, and DG regions) slice of the MS group significantly decreased compared with those of the NMS group. Western blot analysis was used to assess synaptic-plasticity protein markers, including postsynaptic density 95, synaptophysin, and growth-associated binding protein 43 protein expression in the cortex and hippocampus. Results showed that the expression levels of these three proteins in the MS group were significantly lower than those in the NMS group. LC-MS/MS analysis revealed no significant differences in the peak areas of sex hormones and their metabolites, including estradiol, testosterone, androstenedione, estrone, estriol, and 5β-dihydrotestosterone. Through the application of nontargeted metabolomics to the overall analysis of differential metabolites, pathway-enrichment results showed the importance of arginine and proline metabolism; pantothenate and CoA biosyntheses; glutathione metabolism; and the phenylalanine, tyrosine, and tryptophan biosynthesis pathways. In summary, the MS model caused adult SD rats to be susceptible to depression, which may regulate synaptic plasticity through arginine and proline metabolism; pantothenate and CoA biosyntheses; glutathione metabolism; and phenylalanine, tyrosine, and tryptophan biosyntheses.
Collapse
Affiliation(s)
- Yongfei Cui
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Kerun Cao
- School of Fundamental Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Huiyuan Lin
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Sainan Cui
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chongkun Shen
- School of Fundamental Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wenhao Wen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Haixin Mo
- School of Fundamental Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhaoyang Dong
- School of Nursing, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shasha Bai
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lei Yang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yafei Shi
- School of Fundamental Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Rong Zhang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
35
|
Tu X, Yasuda R, Colgan LA. Rac1 is a downstream effector of PKCα in structural synaptic plasticity. Sci Rep 2020; 10:1777. [PMID: 32019972 PMCID: PMC7000694 DOI: 10.1038/s41598-020-58610-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Accepted: 01/17/2020] [Indexed: 11/21/2022] Open
Abstract
Structural and functional plasticity of dendritic spines is the basis of animal learning. The rapid remodeling of actin cytoskeleton is associated with spine enlargement and shrinkage, which are essential for structural plasticity. The calcium-dependent protein kinase C isoform, PKCα, has been suggested to be critical for this actin-dependent plasticity. However, mechanisms linking PKCα and structural plasticity of spines are unknown. Here, we examine the spatiotemporal activation of actin regulators, including small GTPases Rac1, Cdc42 and Ras, in the presence or absence of PKCα during single-spine structural plasticity. Removal of PKCα expression in the postsynapse attenuated Rac1 activation during structural plasticity without affecting Ras or Cdc42 activity. Moreover, disruption of a PDZ binding domain within PKCα led to impaired Rac1 activation and deficits in structural spine remodeling. These results demonstrate that PKCα positively regulates the activation of Rac1 during structural plasticity.
Collapse
Affiliation(s)
- Xun Tu
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
- International Max Planck Research School for Brain and Behavior, Jupiter, FL, USA
- FAU/Max Planck Florida Institute Joint Graduate Program in Integrative Biology and Neuroscience, Florida Atlantic University, Boca Raton, FL, USA
| | - Ryohei Yasuda
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA.
- International Max Planck Research School for Brain and Behavior, Jupiter, FL, USA.
- FAU/Max Planck Florida Institute Joint Graduate Program in Integrative Biology and Neuroscience, Florida Atlantic University, Boca Raton, FL, USA.
| | - Lesley A Colgan
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA.
| |
Collapse
|
36
|
Gutierrez DA, Vargas LM, Chandia-Cristi A, de la Fuente C, Leal N, Alvarez AR. c-Abl Deficiency Provides Synaptic Resiliency Against Aβ-Oligomers. Front Cell Neurosci 2019; 13:526. [PMID: 31849613 PMCID: PMC6902026 DOI: 10.3389/fncel.2019.00526] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 11/11/2019] [Indexed: 12/13/2022] Open
Abstract
Spine pathology has been implicated in the early onset of Alzheimer’s disease (AD), where Aβ-Oligomers (AβOs) cause synaptic dysfunction and loss. Previously, we described that pharmacological inhibition of c-Abl prevents AβOs-induced synaptic alterations. Hence, this kinase seems to be a key element in AD progression. Here, we studied the role of c-Abl on dendritic spine morphological changes induced by AβOs using c-Abl null neurons (c-Abl-KO). First, we characterized the effect of c-Abl deficiency on dendritic spine density and found that its absence increases dendritic spine density. While AβOs-treatment reduces the spine number in both wild-type (WT) and c-Abl-KO neurons, AβOs-driven spine density loss was not affected by c-Abl. We then characterized AβOs-induced morphological changes in dendritic spines of c-Abl-KO neurons. AβOs induced a decrease in the number of mushroom spines in c-Abl-KO neurons while preserving the populations of immature stubby, thin, and filopodia spines. Furthermore, synaptic contacts evaluated by PSD95/Piccolo clustering and cell viability were preserved in AβOs-exposed c-Abl-KO neurons. In conclusion, our results indicate that in the presence of AβOs c-Abl participates in synaptic contact removal, increasing susceptibility to AβOs damage. Its deficiency increases the immature spine population reducing AβOs-induced synapse elimination. Therefore, c-Abl signaling could be a relevant actor in the early stages of AD.
Collapse
Affiliation(s)
- Daniela A Gutierrez
- Cell Signaling Laboratory, Faculty of Biological Science, Department of Cell and Molecular Biology, Center for Aging and Regeneration (CARE), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Lina M Vargas
- Cell Signaling Laboratory, Faculty of Biological Science, Department of Cell and Molecular Biology, Center for Aging and Regeneration (CARE), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - América Chandia-Cristi
- Cell Signaling Laboratory, Faculty of Biological Science, Department of Cell and Molecular Biology, Center for Aging and Regeneration (CARE), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Catalina de la Fuente
- Cell Signaling Laboratory, Faculty of Biological Science, Department of Cell and Molecular Biology, Center for Aging and Regeneration (CARE), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Nancy Leal
- Cell Signaling Laboratory, Faculty of Biological Science, Department of Cell and Molecular Biology, Center for Aging and Regeneration (CARE), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alejandra R Alvarez
- Cell Signaling Laboratory, Faculty of Biological Science, Department of Cell and Molecular Biology, Center for Aging and Regeneration (CARE), Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
37
|
Nishiyama J. Plasticity of dendritic spines: Molecular function and dysfunction in neurodevelopmental disorders. Psychiatry Clin Neurosci 2019; 73:541-550. [PMID: 31215705 DOI: 10.1111/pcn.12899] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Revised: 06/04/2019] [Accepted: 06/06/2019] [Indexed: 02/06/2023]
Abstract
Dendritic spines are tiny postsynaptic protrusions from a dendrite that receive most of the excitatory synaptic input in the brain. Functional and structural changes in dendritic spines are critical for synaptic plasticity, a cellular model of learning and memory. Conversely, altered spine morphology and plasticity are common hallmarks of human neurodevelopmental disorders, such as intellectual disability and autism. The advances in molecular and optical techniques have allowed for exploration of dynamic changes in structure and signal transduction at single-spine resolution, providing significant insights into the molecular regulation underlying spine structural plasticity. Here, I review recent findings on: how synaptic stimulation leads to diverse forms of spine structural plasticity; how the associated biochemical signals are initiated and transmitted into neuronal compartments; and how disruption of single genes associated with neurodevelopmental disorders can lead to abnormal spine structure in human and mouse brains. In particular, I discuss the functions of the Ras superfamily of small GTPases in spatiotemporal regulation of the actin cytoskeleton and protein synthesis in dendritic spines. Multiple lines of evidence implicate disrupted Ras signaling pathways in the spine structural abnormalities observed in neurodevelopmental disorders. Both deficient and excessive Ras activities lead to disrupted spine structure and deficits in learning and memory. Dysregulation of spine Ras signaling, therefore, may play a key role in the pathogenesis of multiple neurodevelopmental disorders with distinct etiologies.
Collapse
Affiliation(s)
- Jun Nishiyama
- Program in Neuroscience and Behavioral Disorders, Duke-National University of Singapore Medical School, Singapore
| |
Collapse
|
38
|
Kulik YD, Watson DJ, Cao G, Kuwajima M, Harris KM. Structural plasticity of dendritic secretory compartments during LTP-induced synaptogenesis. eLife 2019; 8:e46356. [PMID: 31433297 PMCID: PMC6728136 DOI: 10.7554/elife.46356] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Accepted: 08/20/2019] [Indexed: 12/30/2022] Open
Abstract
Long-term potentiation (LTP), an increase in synaptic efficacy following high-frequency stimulation, is widely considered a mechanism of learning. LTP involves local remodeling of dendritic spines and synapses. Smooth endoplasmic reticulum (SER) and endosomal compartments could provide local stores of membrane and proteins, bypassing the distant Golgi apparatus. To test this hypothesis, effects of LTP were compared to control stimulation in rat hippocampal area CA1 at postnatal day 15 (P15). By two hours, small spines lacking SER increased after LTP, whereas large spines did not change in frequency, size, or SER content. Total SER volume decreased after LTP consistent with transfer of membrane to the added spines. Shaft SER remained more abundant in spiny than aspiny dendritic regions, apparently supporting the added spines. Recycling endosomes were elevated specifically in small spines after LTP. These findings suggest local secretory trafficking contributes to LTP-induced synaptogenesis and primes the new spines for future plasticity.
Collapse
Affiliation(s)
- Yelena D Kulik
- Center for Learning and Memory, Department of NeuroscienceThe University of Texas at AustinAustinUnited States
| | - Deborah J Watson
- Center for Learning and Memory, Department of NeuroscienceThe University of Texas at AustinAustinUnited States
| | - Guan Cao
- Center for Learning and Memory, Department of NeuroscienceThe University of Texas at AustinAustinUnited States
| | - Masaaki Kuwajima
- Center for Learning and Memory, Department of NeuroscienceThe University of Texas at AustinAustinUnited States
| | - Kristen M Harris
- Center for Learning and Memory, Department of NeuroscienceThe University of Texas at AustinAustinUnited States
| |
Collapse
|
39
|
Mitchell DE, Martineau É, Tazerart S, Araya R. Probing Single Synapses via the Photolytic Release of Neurotransmitters. Front Synaptic Neurosci 2019; 11:19. [PMID: 31354469 PMCID: PMC6640007 DOI: 10.3389/fnsyn.2019.00019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 06/24/2019] [Indexed: 11/13/2022] Open
Abstract
The development of two-photon microscopy has revolutionized our understanding of how synapses are formed and how they transform synaptic inputs in dendritic spines-tiny protrusions that cover the dendrites of pyramidal neurons that receive most excitatory synaptic information in the brain. These discoveries have led us to better comprehend the neuronal computations that take place at the level of dendritic spines as well as within neuronal circuits with unprecedented resolution. Here, we describe a method that uses a two-photon (2P) microscope and 2P uncaging of caged neurotransmitters for the activation of single and multiple spines in the dendrites of cortical pyramidal neurons. In addition, we propose a cost-effective description of the components necessary for the construction of a one laser source-2P microscope capable of nearly simultaneous 2P uncaging of neurotransmitters and 2P calcium imaging of the activated spines and nearby dendrites. We provide a brief overview on how the use of these techniques have helped researchers in the last 15 years unravel the function of spines in: (a) information processing; (b) storage; and (c) integration of excitatory synaptic inputs.
Collapse
Affiliation(s)
- Diana E. Mitchell
- Department of Neurosciences, Faculty of Medicine, University of Montreal, Montreal, QC, Canada
- The CHU Sainte-Justine Research Center, Montreal, QC, Canada
| | - Éric Martineau
- Department of Neurosciences, Faculty of Medicine, University of Montreal, Montreal, QC, Canada
- The CHU Sainte-Justine Research Center, Montreal, QC, Canada
| | - Sabrina Tazerart
- Department of Neurosciences, Faculty of Medicine, University of Montreal, Montreal, QC, Canada
- The CHU Sainte-Justine Research Center, Montreal, QC, Canada
| | - Roberto Araya
- Department of Neurosciences, Faculty of Medicine, University of Montreal, Montreal, QC, Canada
- The CHU Sainte-Justine Research Center, Montreal, QC, Canada
| |
Collapse
|
40
|
Kim T, Tanaka-Yamamoto K. Postsynaptic Stability and Variability Described by a Stochastic Model of Endosomal Trafficking. Front Cell Neurosci 2019; 13:72. [PMID: 30863286 PMCID: PMC6399135 DOI: 10.3389/fncel.2019.00072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 02/13/2019] [Indexed: 12/04/2022] Open
Abstract
Neurons undergo dynamic processes of constitutive AMPA-type glutamate receptor (AMPAR) trafficking, such as the insertion and internalization of AMPARs by exocytosis and endocytosis, while stably maintaining synaptic efficacy. Studies using advanced imaging techniques have suggested that the frequency of these constitutive trafficking processes, as well as the number of AMPARs that are involved in a particular event highly fluctuate. In addition, mechanisms that trigger some forms of synaptic plasticity have been shown to include not only these processes but also additional fluctuating processes, such as the sorting of AMPARs to late endosomes (LEs). Thus, the regulation of postsynaptic AMPARs by the endosomal trafficking system appears to have superficially conflicting properties between the stability or organized control of plasticity and highly fluctuating or stochastic processes. However, it is not clear how the endosomal trafficking system reconciles and utilizes such conflicting properties. Although deterministic models have been effective to describe the stable maintenance of synaptic AMPAR numbers by constitutive recycling, as well as the involvement of endosomal trafficking in synaptic plasticity, they do not take stochasticity into account. In this study, we introduced the stochasticity into the model of each crucial machinery of the endosomal trafficking system. The specific questions we solved by our improved model are whether stability is accomplished even with a combination of fluctuating processes, and how overall variability occurs while controlling long-term synaptic depression (LTD). Our new stochastic model indeed demonstrated the stable regulation of postsynaptic AMPAR numbers at the basal state and during LTD maintenance, despite fast fluctuations in AMPAR numbers as well as high variability in the time course and amounts of LTD. In addition, our analysis suggested that the high variability arising from this stochasticity is beneficial for reproducing the relatively constant timing of LE sorting for LTD. We therefore propose that the coexistence of stability and stochasticity in the endosomal trafficking system is suitable for stable synaptic transmission and the reliable induction of synaptic plasticity, with variable properties that have been observed experimentally.
Collapse
Affiliation(s)
- Taegon Kim
- Center for Functional Connectomics, Korea Institute of Science and Technology (KIST), Seoul, South Korea
| | - Keiko Tanaka-Yamamoto
- Center for Functional Connectomics, Korea Institute of Science and Technology (KIST), Seoul, South Korea.,Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul, South Korea
| |
Collapse
|
41
|
Urban P, Rezaei V, Bokota G, Denkiewicz M, Basu S, Plewczyński D. Dendritic Spines Taxonomy: The Functional and Structural Classification • Time-Dependent Probabilistic Model of Neuronal Activation. J Comput Biol 2019; 26:322-335. [PMID: 30810368 DOI: 10.1089/cmb.2018.0155] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Categorizing spines into four subpopulations, stubby, mushroom, thin, or filopodia, is one of the common approaches in morphological analysis. Most cellular models describing synaptic plasticity, long-term potentiation (LTP), and long-term depression associate synaptic strength with either spine enlargement or spine shrinkage. Unfortunately, although we have a lot of available software with automatic spine segmentation and feature extraction methods, at present none of them allows for automatic and unbiased distinction between dendritic spine subpopulations, or for the detailed computational models of spine behavior. Therefore, we propose structural classification based on two different mathematical approaches: unsupervised construction of spine shape taxonomy based on arbitrary features (SpineTool) and supervised classification exploiting convolution kernels theory (2dSpAn). We compared two populations of spines in a form of static and dynamic data sets gathered at three time points. The dynamic data contain two sets of spines: the active set and the control set. The first population was stimulated with LTP, and the other population in its resting state was used as a control population. We propose one equation describing the distribution of variables that best fits all dendritic spine parameters.
Collapse
Affiliation(s)
- Paulina Urban
- 1 Center of New Technologies, University of Warsaw, Warsaw, Poland.,2 College of Inter-Faculty Individual Studies in Mathematics and Natural Sciences, University of Warsaw, Warsaw, Poland
| | - Vahid Rezaei
- 3 Department of Statistics, Faculty of Mathematics and Computer Sciences, Allameh Tabataba'i University, Tehran, Iran.,4 School of Biological Sciences, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran
| | - Grzegorz Bokota
- 1 Center of New Technologies, University of Warsaw, Warsaw, Poland.,5 Faculty of Mathematics, Informatics and Mechanics, University of Warsaw, Warsaw, Poland
| | - Michał Denkiewicz
- 1 Center of New Technologies, University of Warsaw, Warsaw, Poland.,2 College of Inter-Faculty Individual Studies in Mathematics and Natural Sciences, University of Warsaw, Warsaw, Poland
| | - Subhadip Basu
- 6 Department of Computer Science and Engineering, Jadavpur University, Kolkata, India
| | - Dariusz Plewczyński
- 1 Center of New Technologies, University of Warsaw, Warsaw, Poland.,7 Faculty of Mathematics and Information Science, Warsaw University of Technology, Warsaw, Poland
| |
Collapse
|
42
|
Ji J, Moquin A, Bertorelle F, KY Chang P, Antoine R, Luo J, McKinney RA, Maysinger D. Organotypic and primary neural cultures as models to assess effects of different gold nanostructures on glia and neurons. Nanotoxicology 2019; 13:285-304. [DOI: 10.1080/17435390.2018.1543468] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Jeff Ji
- Department of Pharmacology & Therapeutics, McGill University, Montreal, Canada
| | - Alexandre Moquin
- Department of Pharmacology & Therapeutics, McGill University, Montreal, Canada
| | - Franck Bertorelle
- CNRS, Institut Lumière Matière, Université Lyon Université Claude Bernard Lyon 1, Lyon, France
| | - Philip KY Chang
- Department of Pharmacology & Therapeutics, McGill University, Montreal, Canada
| | - Rodolphe Antoine
- CNRS, Institut Lumière Matière, Université Lyon Université Claude Bernard Lyon 1, Lyon, France
| | - Julia Luo
- Department of Pharmacology & Therapeutics, McGill University, Montreal, Canada
| | - R. Anne McKinney
- Department of Pharmacology & Therapeutics, McGill University, Montreal, Canada
| | - Dusica Maysinger
- Department of Pharmacology & Therapeutics, McGill University, Montreal, Canada
| |
Collapse
|
43
|
Ketamine and selective activation of parvalbumin interneurons inhibit stress-induced dendritic spine elimination. Transl Psychiatry 2018; 8:272. [PMID: 30531859 PMCID: PMC6288154 DOI: 10.1038/s41398-018-0321-5] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 11/12/2018] [Accepted: 11/13/2018] [Indexed: 12/13/2022] Open
Abstract
Stress is a major risk factor for the onset of many psychiatric diseases. In rodent models, chronic stress induces depression and impairs excitatory neurotransmission. However, little is known about the effect of stress on synaptic circuitry during the development of behavioral symptoms. Using two-photon transcranial imaging, we studied the effect of repeated restraint stress on dendritic spine plasticity in the frontal cortex in vivo. We found that restraint stress induced dendritic spine loss by decreasing the rate of spine formation and increasing the rate of spine elimination. The N-methyl-D-aspartate receptor antagonist ketamine inhibited stress-induced spine loss mainly by protecting mushroom spines from elimination. Ketamine also induced re-formation of spines in close proximity to previously stress-eliminated spines. Electrophysiological and in vivo imaging experiments showed that ketamine enhanced activity of parvalbumin (PV) interneurons under stress and counterbalanced the stress-induced net loss of PV axonal boutons. In addition, selective chemogenetic excitation of PV interneurons mimicked the protective effects of ketamine on dendritic spines against stress. Collectively, our data provide new insights on the effects of ketamine on synaptic circuitry under stress and a possible mechanism to counteract stress-induced synaptic impairments through PV interneuron activation.
Collapse
|
44
|
Neuhofer D, Kalivas P. Metaplasticity at the addicted tetrapartite synapse: A common denominator of drug induced adaptations and potential treatment target for addiction. Neurobiol Learn Mem 2018; 154:97-111. [PMID: 29428364 PMCID: PMC6112115 DOI: 10.1016/j.nlm.2018.02.007] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 01/26/2018] [Accepted: 02/07/2018] [Indexed: 11/22/2022]
Abstract
In light of the current worldwide addiction epidemic, the need for successful therapies is more urgent than ever. Although we made substantial progress in our basic understanding of addiction, reliable therapies are lacking. Since 40-60% of patients treated for substance use disorder return to active substance use within a year following treatment discharge, alleviating the vulnerability to relapse is regarded as the most promising avenue for addiction therapy. Preclinical addiction research often focuses on maladaptive synaptic plasticity within the reward pathway. However, drug induced neuroadaptations do not only lead to a strengthening of distinct drug associated cues and drug conditioned behaviors, but also seem to increase plasticity thresholds for environmental stimuli that are not associated with the drug. This form of higher order plasticity, or synaptic metaplasticity, is not expressed as a change in the efficacy of synaptic transmission but as a change in the direction or degree of plasticity induced by a distinct stimulation pattern. Experimental addiction research has demonstrated metaplasticity after exposure to multiple classes of addictive drugs. In this review we will focus on the concept of synaptic metaplasticity in the context of preclinical addiction research. We will take a closer look at the tetrapartite glutamatergic synapse and outline forms of metaplasticity that have been described at the addicted synapse. Finally we will discuss the different potential avenues for pharmacotherapies that target glutamatergic synaptic plasticity and metaplasticity. Here we will argue that aberrant metaplasticity renders the reward seeking circuitry more rigid and hence less able to adapt to changing environmental contingencies. An understanding of the molecular mechanisms that underlie this metaplasticity is crucial for the development of new strategies for addiction therapy. The correction of drug-induced metaplasticity could be used to support behavioral and pharmacotherapies for the treatment of addiction.
Collapse
Affiliation(s)
- Daniela Neuhofer
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC 29425, United States.
| | - Peter Kalivas
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC 29425, United States
| |
Collapse
|
45
|
Nakahata Y, Yasuda R. Plasticity of Spine Structure: Local Signaling, Translation and Cytoskeletal Reorganization. Front Synaptic Neurosci 2018; 10:29. [PMID: 30210329 PMCID: PMC6123351 DOI: 10.3389/fnsyn.2018.00029] [Citation(s) in RCA: 142] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 08/07/2018] [Indexed: 12/31/2022] Open
Abstract
Dendritic spines are small protrusive structures on dendritic surfaces, and function as postsynaptic compartments for excitatory synapses. Plasticity of spine structure is associated with many forms of long-term neuronal plasticity, learning and memory. Inside these small dendritic compartments, biochemical states and protein-protein interactions are dynamically modulated by synaptic activity, leading to the regulation of protein synthesis and reorganization of cytoskeletal architecture. This in turn causes plasticity of structure and function of the spine. Technical advances in monitoring molecular behaviors in single dendritic spines have revealed that each signaling pathway is differently regulated across multiple spatiotemporal domains. The spatial pattern of signaling activity expands from a single spine to the nearby dendritic area, dendritic branch and the nucleus, regulating different cellular events at each spatial scale. Temporally, biochemical events are typically triggered by short Ca2+ pulses (~10–100 ms). However, these signals can then trigger activation of downstream protein cascades that can last from milliseconds to hours. Recent imaging studies provide many insights into the biochemical processes governing signaling events of molecular assemblies at different spatial localizations. Here, we highlight recent findings of signaling dynamics during synaptic plasticity and discuss their roles in long-term structural plasticity of dendritic spines.
Collapse
Affiliation(s)
- Yoshihisa Nakahata
- Neuronal Signal Transduction, Max Planck Florida Institute for Neuroscience (MPFI), Jupiter, FL, United States
| | - Ryohei Yasuda
- Neuronal Signal Transduction, Max Planck Florida Institute for Neuroscience (MPFI), Jupiter, FL, United States
| |
Collapse
|
46
|
Sancho L, Bloodgood BL. Functional Distinctions between Spine and Dendritic Synapses Made onto Parvalbumin-Positive Interneurons in Mouse Cortex. Cell Rep 2018; 24:2075-2087. [DOI: 10.1016/j.celrep.2018.07.070] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 06/15/2018] [Accepted: 07/19/2018] [Indexed: 11/29/2022] Open
|
47
|
Omar MH, Kerrisk Campbell M, Xiao X, Zhong Q, Brunken WJ, Miner JH, Greer CA, Koleske AJ. CNS Neurons Deposit Laminin α5 to Stabilize Synapses. Cell Rep 2018; 21:1281-1292. [PMID: 29091766 PMCID: PMC5776391 DOI: 10.1016/j.celrep.2017.10.028] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 09/21/2017] [Accepted: 10/08/2017] [Indexed: 11/28/2022] Open
Abstract
Synapses in the developing brain are structurally dynamic but become stable by early adulthood. We demonstrate here that an α5-subunit-containing laminin stabilizes synapses during this developmental transition. Hippocampal neurons deposit laminin α5 at synapses during adolescence as connections stabilize. Disruption of laminin α5 in neurons causes dramatic fluctuations in dendritic spine head size that can be rescued by exogenous α5-containing laminin. Conditional deletion of laminin α5 in vivo increases dendritic spine size and leads to an age-dependent loss of synapses accompanied by behavioral defects. Remaining synapses have larger postsynaptic densities and enhanced neurotransmission. Finally, we provide evidence that laminin α5 acts through an integrin α3β1-Abl2 kinase-p190RhoGAP signaling cascade and partners with laminin β2 to regulate dendritic spine density and behavior. Together, our results identify laminin α5 as a stabilizer of dendritic spines and synapses in the brain and elucidate key cellular and molecular mechanisms by which it acts.
Collapse
Affiliation(s)
- Mitchell H Omar
- Interdepartmental Neuroscience Program, Yale University, New Haven, CT 06510, USA; Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06510, USA
| | - Meghan Kerrisk Campbell
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06510, USA
| | - Xiao Xiao
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06510, USA
| | - Qiaonan Zhong
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT 06510, USA
| | - William J Brunken
- Department of Ophthalmology, Upstate Medical University, Syracuse, NY 13202, USA
| | - Jeffrey H Miner
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Charles A Greer
- Interdepartmental Neuroscience Program, Yale University, New Haven, CT 06510, USA; Department of Neuroscience, Yale University, New Haven, CT 06510, USA; Department of Neurosurgery, Yale University, New Haven, CT 06510, USA
| | - Anthony J Koleske
- Interdepartmental Neuroscience Program, Yale University, New Haven, CT 06510, USA; Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06510, USA; Department of Neuroscience, Yale University, New Haven, CT 06510, USA.
| |
Collapse
|
48
|
Postnatal Ablation of Synaptic Retinoic Acid Signaling Impairs Cortical Information Processing and Sensory Discrimination in Mice. J Neurosci 2018; 38:5277-5288. [PMID: 29760176 DOI: 10.1523/jneurosci.3028-17.2018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 04/28/2018] [Accepted: 05/03/2018] [Indexed: 12/28/2022] Open
Abstract
Retinoic acid (RA) and its receptors (RARs) are well established essential transcriptional regulators during embryonic development. Recent findings in cultured neurons identified an independent and critical post-transcriptional role of RA and RARα in the homeostatic regulation of excitatory and inhibitory synaptic transmission in mature neurons. However, the functional relevance of synaptic RA signaling in vivo has not been established. Here, using somatosensory cortex as a model system and the RARα conditional knock-out mouse as a tool, we applied multiple genetic manipulations to delete RARα postnatally in specific populations of cortical neurons, and asked whether synaptic RA signaling observed in cultured neurons is involved in cortical information processing in vivo Indeed, conditional ablation of RARα in mice via a CaMKIIα-Cre or a layer 5-Cre driver line or via somatosensory cortex-specific viral expression of Cre-recombinase impaired whisker-dependent texture discrimination, suggesting a critical requirement of RARα expression in L5 pyramidal neurons of somatosensory cortex for normal tactile sensory processing. Transcranial two-photon imaging revealed a significant increase in dendritic spine elimination on apical dendrites of somatosensory cortical layer 5 pyramidal neurons in these mice. Interestingly, the enhancement of spine elimination is whisker experience-dependent as whisker trimming rescued the spine elimination phenotype. Additionally, experiencing an enriched environment improved texture discrimination in RARα-deficient mice and reduced excessive spine pruning. Thus, RA signaling is essential for normal experience-dependent cortical circuit remodeling and sensory processing.SIGNIFICANCE STATEMENT The importance of synaptic RA signaling has been demonstrated in in vitro studies. However, whether RA signaling mediated by RARα contributes to neural circuit functions in vivo remains largely unknown. In this study, using a RARα conditional knock-out mouse, we performed multiple regional/cell-type-specific manipulation of RARα expression in the postnatal brain, and show that RARα signaling contributes to normal whisker-dependent texture discrimination as well as regulating spine dynamics of apical dendrites from layer (L5) pyramidal neurons in S1. Deletion of RARα in excitatory neurons in the forebrain induces elevated spine elimination and impaired sensory discrimination. Our study provides novel insights into the role of RARα signaling in cortical processing and experience-dependent spine maturation.
Collapse
|
49
|
Abstract
Dynamic modification of synaptic connectivity in response to sensory experience is a vital step in the refinement of brain circuits as they are established during development and modified during learning. In addition to the well-established role for new spine growth and stabilization in the experience-dependent plasticity of neural circuits, dendritic spine elimination has been linked to improvements in learning, and dysregulation of spine elimination has been associated with intellectual disability and behavioral impairment. Proper brain function requires a tightly regulated balance between spine formation and spine elimination. Although most studies have focused on the mechanisms of spine formation, considerable progress has been made recently in delineating the neural activity patterns and downstream molecular mechanisms that drive dendritic spine elimination. Here, we review the current state of knowledge concerning the signaling pathways that drive dendritic spine shrinkage and elimination in the cerebral cortex and we discuss their implication in neuropsychiatric and neurodegenerative disease.
Collapse
Affiliation(s)
- Ivar S Stein
- 1 Center for Neuroscience, University of California, Davis, CA, USA
| | - Karen Zito
- 1 Center for Neuroscience, University of California, Davis, CA, USA
| |
Collapse
|
50
|
Banghart MR, He XJ, Sabatini BL. A Caged Enkephalin Optimized for Simultaneously Probing Mu and Delta Opioid Receptors. ACS Chem Neurosci 2018; 9:684-690. [PMID: 29266926 PMCID: PMC5906201 DOI: 10.1021/acschemneuro.7b00485] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Physiological responses to the opioid neuropeptide enkephalin often involve both mu and delta opioid receptors. To facilitate quantitative studies into opioid signaling, we previously developed a caged [Leu5]-enkephalin that responds to ultraviolet irradiation, but its residual activity at delta receptors confounds experiments that involve both receptors. To reduce residual activity, we evaluated side-chain, N-terminus, and backbone caging sites and further incorporated the dimethoxy-nitrobenzyl moiety to improve sensitivity to ultraviolet light-emitting diodes (LEDs). Residual activity was characterized using an in vitro functional assay, and the power dependence and kinetics of the uncaging response to 355 nm laser irradiation were assayed using electrophysiological recordings of mu opioid receptor-mediated potassium currents in brain slices of rat locus coeruleus. These experiments identified N-MNVOC-LE as an optimal compound. Using ultraviolet LED illumination to photoactivate N-MNVOC-LE in the CA1 region of hippocampus, we found that enkephalin engages both mu and delta opioid receptors to suppress inhibitory synaptic transmission.
Collapse
Affiliation(s)
- Matthew R. Banghart
- Division of Biological Sciences, Section on Neurobiology, University of California San Diego, La Jolla, California 92093, United States
- Howard Hughes Medical Institute, Department of Neurobiology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Xinyi J. He
- Division of Biological Sciences, Section on Neurobiology, University of California San Diego, La Jolla, California 92093, United States
| | - Bernardo L. Sabatini
- Howard Hughes Medical Institute, Department of Neurobiology, Harvard Medical School, Boston, Massachusetts 02115, United States
| |
Collapse
|