1
|
Lindamood HL, Liu TM, Read TA, Vitriol EA. Using ALS to understand profilin 1's diverse roles in cellular physiology. Cytoskeleton (Hoboken) 2025; 82:111-129. [PMID: 39056295 PMCID: PMC11762371 DOI: 10.1002/cm.21896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/03/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024]
Abstract
Profilin is an actin monomer-binding protein whose role in actin polymerization has been studied for nearly 50 years. While its principal biochemical features are now well understood, many questions remain about how profilin controls diverse processes within the cell. Dysregulation of profilin has been implicated in a broad range of human diseases, including neurodegeneration, inflammatory disorders, cardiac disease, and cancer. For example, mutations in the profilin 1 gene (PFN1) can cause amyotrophic lateral sclerosis (ALS), although the precise mechanisms that drive neurodegeneration remain unclear. While initial work suggested proteostasis and actin cytoskeleton defects as the main pathological pathways, multiple novel functions for PFN1 have since been discovered that may also contribute to ALS, including the regulation of nucleocytoplasmic transport, stress granules, mitochondria, and microtubules. Here, we will review these newly discovered roles for PFN1, speculate on their contribution to ALS, and discuss how defects in actin can contribute to these processes. By understanding profilin 1's involvement in ALS pathogenesis, we hope to gain insight into this functionally complex protein with significant influence over cellular physiology.
Collapse
Affiliation(s)
- Halli L Lindamood
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Tatiana M Liu
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Tracy-Ann Read
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Eric A Vitriol
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| |
Collapse
|
2
|
Kwon Y, Lee SJ, Shin YK, Choi JS, Park D, Shin JE. Loss of neuronal βPix isoforms impairs neuronal morphology in the hippocampus and causes behavioral defects. Anim Cells Syst (Seoul) 2025; 29:57-71. [PMID: 39802101 PMCID: PMC11722029 DOI: 10.1080/19768354.2024.2448999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 11/19/2024] [Accepted: 12/23/2024] [Indexed: 01/16/2025] Open
Abstract
βPix is a guanine nucleotide exchange factor for the Rac1 and Cdc42 small GTPases, which play important roles in dendritic spine morphogenesis by modulating actin cytoskeleton organization. The formation and plasticity of the dendritic spines are essential for normal brain function. Among the alternatively spliced βPix isoforms, βPix-b and βPix-d are expressed specifically in neurons. Our previous studies using cultured hippocampal neurons identified the roles of βPix-b and βPix-d in spine formation and neurite development, respectively. Here, we analyzed the in vivo role of the neuronal βPix isoforms in brain development and function by using βPix neuronal isoform knockout (βPix-NIKO) mice, in which the expression of the βPix-b and βPix-d isoforms is blocked, while the expression of the ubiquitous βPix-a isoform is maintained. Loss of the neuronal βPix isoforms leads to reduced activity of Rac1 and Cdc42, decreased dendritic complexity and spine density, and increased GluN2B and Ca2+/calmodulin-dependent protein kinase IIα expression in the hippocampus. The defects in neurite development, dendritic spine maturation, and synaptic density in cultured βPix-NIKO hippocampal neurons were rescued by the expression of βPix-b or βPix-d. In behavioral studies, βPix-NIKO mice exhibited robust deficits in novel object recognition and decreased anxiety levels. Our findings suggest that neuronal morphogenetic signaling by the neuronal βPix isoforms contributes to normal behaviors.
Collapse
Affiliation(s)
- Younghee Kwon
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Seung Joon Lee
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Yoon Kyung Shin
- Department of Molecular Neuroscience, College of Medicine, Dong-A University, Busan, Republic of Korea
| | - June-Seek Choi
- Department of Psychology, Korea University, Seoul, Republic of Korea
| | - Dongeun Park
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Jung Eun Shin
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
- Department of Molecular Neuroscience, College of Medicine, Dong-A University, Busan, Republic of Korea
- Department of Translational Biomedical Sciences, Graduate School of Dong-A University, Busan, Republic of Korea
| |
Collapse
|
3
|
Abstract
The brain has the powerful ability to transform experiences into anatomic maps and continuously integrate massive amounts of information to form new memories. The manner in which the brain performs these processes has been investigated extensively for decades. Emerging reports suggest that dendritic spines are the structural basis of information storage. The complex orchestration of functional and structural dynamics of dendritic spines is associated with learning and memory. Owing to advancements in techniques, more precise observations and manipulation enable the investigation of dendritic spines and provide clues to the challenging question of how memories reside in dendritic spines. In this review, we summarize the remarkable progress made in revealing the role of dendritic spines in fear memory and the techniques used in this field.
Collapse
Affiliation(s)
- Ja Eun Choi
- School of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Bong-Kiun Kaang
- School of Biological Sciences, Seoul National University, Seoul, South Korea
| |
Collapse
|
4
|
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
|
5
|
Yang J, Zhou S, Yang Z, Shi X, Liu H, Yang Z, Peng D, Ding Z, Ye S. Silencing of the T-type voltage-gated calcium channel α 1 subunit by fungus-mediated RNAi altered the structure of F-actin and caused defective behaviors in Ditylenchus destructor. Mol Biol Rep 2024; 51:673. [PMID: 38787479 DOI: 10.1007/s11033-024-09626-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 05/08/2024] [Indexed: 05/25/2024]
Abstract
BACKGROUND T-type calcium channels, characterized as low-voltage activated (LVA) calcium channels, play crucial physiological roles across a wide range of tissues, including both the neuronal and nonneuronal systems. Using in situ hybridization and RNA interference (RNAi) techniques in vitro, we previously identified the tissue distribution and physiological function of the T-type calcium channel α1 subunit (DdCα1G) in the plant-parasitic nematode Ditylenchus destructor. METHODS AND RESULTS To further characterize the functional role of DdCα1G, we employed a combination of immunohistochemistry and fungus-mediated RNAi and found that DdCα1G was clearly distributed in stylet-related tissue, oesophageal gland-related tissue, secretory-excretory duct-related tissue and male spicule-related tissue. Silencing DdCα1G led to impairments in the locomotion, feeding, reproductive ability and protein secretion of nematodes. To confirm the defects in behavior, we used phalloidin staining to examine muscle changes in DdCα1G-RNAi nematodes. Our observations demonstrated that defective behaviors are associated with related muscular atrophy. CONCLUSION Our findings provide a deeper understanding of the physiological functions of T-type calcium channels in plant-parasitic nematodes. The T-type calcium channel can be considered a promising target for sustainable nematode management practices.
Collapse
Affiliation(s)
- Jiahao Yang
- College of Plant Protection, Hunan Agricultural University, Changsha, Hunan, 410128, China
| | - Siyu Zhou
- College of Plant Protection, Hunan Agricultural University, Changsha, Hunan, 410128, China
| | - Ziqi Yang
- College of Plant Protection, Hunan Agricultural University, Changsha, Hunan, 410128, China
| | - Xuqi Shi
- College of Plant Protection, Hunan Agricultural University, Changsha, Hunan, 410128, China
| | - Haoran Liu
- College of Plant Protection, Hunan Agricultural University, Changsha, Hunan, 410128, China
| | - Zhuhong Yang
- College of Plant Protection, Hunan Agricultural University, Changsha, Hunan, 410128, China
- Hunan Provincial Engineering & Technology Research Center for Biopesticide and Formulation Processing, Changsha, Hunan, 410128, China
| | - Deliang Peng
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Zhong Ding
- College of Plant Protection, Hunan Agricultural University, Changsha, Hunan, 410128, China.
- Hunan Provincial Engineering & Technology Research Center for Biopesticide and Formulation Processing, Changsha, Hunan, 410128, China.
| | - Shan Ye
- College of Plant Protection, Hunan Agricultural University, Changsha, Hunan, 410128, China.
- Hunan Provincial Engineering & Technology Research Center for Biopesticide and Formulation Processing, Changsha, Hunan, 410128, China.
| |
Collapse
|
6
|
Yamazaki H, Koganezawa N, Yokoo H, Sekino Y, Shirao T. Super-resolution imaging reveals the relationship between CaMKIIβ and drebrin within dendritic spines. Neurosci Res 2024; 199:30-35. [PMID: 37659612 DOI: 10.1016/j.neures.2023.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/25/2023] [Accepted: 08/28/2023] [Indexed: 09/04/2023]
Abstract
Dendritic spines are unique postsynaptic structures that emerge from the dendrites of neurons. They undergo activity-dependent morphological changes known as structural plasticity. The changes involve actin cytoskeletal remodeling, which is regulated by actin-binding proteins. CaMKII is a crucial molecule in synaptic plasticity. Notably, CaMKIIβ subtype is known to bind to filamentous-actin and is closely involved in structural plasticity. We have shown that CaMKIIβ binds to drebrin, and is localized in spines as both drebrin-dependent and drebrin-independent pools. However, the nanoscale relationship between drebrin and CaMKIIβ within dendritic spines has not been clarified. In this study, we used stochastic optical reconstruction microscopy (STORM) to examine the detailed localization of these proteins. STORM imaging showed that CaMKIIβ co-localized with drebrin in the core region of spines, and localized in the submembrane region of spines without drebrin. Interestingly, the dissociation of CaMKIIβ and drebrin in the core region was induced by NMDA receptor activation. In drebrin knockdown neurons, CaMKIIβ was decreased in the core region but not in the submembrane region. Together it indicates that the clustering of CaMKIIβ in the spine core region is dependent on drebrin. These findings suggest that drebrin-dependent CaMKIIβ is in a standby state before its activation.
Collapse
Affiliation(s)
- Hiroyuki Yamazaki
- Faculty of Social Welfare, Gunma University of Health and Welfare, 191-1 Kawamagari-cho, Maebashi 371-0823, Japan; Department of Pharmacology, Gunma University Graduate School of Medicine, 3-39-22, Showa-machi, Maebashi 371-8511, Gunma, Japan.
| | - Noriko Koganezawa
- Department of Pharmacology, Gunma University Graduate School of Medicine, 3-39-22, Showa-machi, Maebashi 371-8511, Gunma, Japan
| | - Hideaki Yokoo
- Department of Human Pathology, Gunma University Graduate School of Medicine, 3-39-22, Showa-machi, Maebashi 371-8511, Gunma, Japan
| | - Yuko Sekino
- Department of Veterinary Pathophysiology and Animal Health, Graduate School of Agricultural and Life Sciences, the University of Tokyo, Bunkyo-ku, Tokyo 113-8657, Japan; Institute for Drug Discovery Innovation, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Tomoaki Shirao
- AlzMed, Inc, UT South building Entrepreneurs Lab, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-8485, Japan
| |
Collapse
|
7
|
Argunsah AÖ, Israely I. Homosynaptic plasticity induction causes heterosynaptic changes at the unstimulated neighbors in an induction pattern and location-specific manner. Front Cell Neurosci 2023; 17:1253446. [PMID: 37829671 PMCID: PMC10564986 DOI: 10.3389/fncel.2023.1253446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 08/24/2023] [Indexed: 10/14/2023] Open
Abstract
Dendritic spines are highly dynamic structures whose structural and functional fluctuations depend on multiple factors. Changes in synaptic strength are not limited to synapses directly involved in specific activity patterns. Unstimulated clusters of neighboring spines in and around the site of stimulation can also undergo alterations in strength. Usually, when plasticity is induced at single dendritic spines with glutamate uncaging, neighboring spines do not show any significant structural fluctuations. Here, using two-photon imaging and glutamate uncaging at single dendritic spines of hippocampal pyramidal neurons, we show that structural modifications at unstimulated neighboring spines occur and are a function of the temporal pattern of the plasticity-inducing stimulus. Further, the relative location of the unstimulated neighbors within the local dendritic segment correlates with the extent of heterosynaptic plasticity that is observed. These findings indicate that naturalistic patterns of activity at single spines can shape plasticity at nearby clusters of synapses, and may play a role in priming local inputs for further modifications.
Collapse
Affiliation(s)
- Ali Özgür Argunsah
- Laboratory of Neuronal Circuit Assembly, Brain Research Institute (HiFo), University of Zurich, Zurich, Switzerland
- Department of Molecular Biology and Genetics, Faculty of Engineering and Natural Sciences, Kadir Has University, Istanbul, Türkiye
| | - Inbal Israely
- Department of Physiology and Biophysics, University of Washington School of Medicine, Seattle, WA, United States
| |
Collapse
|
8
|
Ma Q, Ruan H, Dai H, Yao WD. USP48/USP31 Is a Nuclear Deubiquitinase that Potently Regulates Synapse Remodeling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.19.558317. [PMID: 37781625 PMCID: PMC10541093 DOI: 10.1101/2023.09.19.558317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/03/2023]
Abstract
Deubiquitinases present locally at synapses regulate synaptic development, function, and plasticity. It remains largely unknown, however, whether deubiquitinases localized outside of the synapse control synapse remodeling. Here we identify ubiquitin specific protease 48 (USP48; formerly USP31) as a nuclear deubiquitinase mediating robust synapse removal. USP48 is expressed primarily during the first postnatal week in the rodent brain and is virtually restricted to nuclei, mediated by a conserved, 13-amino acid nuclear localization signal. When exogenously expressed, USP48, in a deubiquitinase and nuclear localization-dependent manner, induces striking filopodia elaboration, marked spine loss, and significantly reduced synaptic protein clustering in vitro, and erases ~70% of functional synapses in vivo. USP48 interacts with the transcription factor NF-κB, deubiquitinates NF-κB subunit p65 and promotes its stability and activation, and up-regulates NF-κB target genes known to inhibit synaptogenesis. Depleting NF-κB prevents USP48-dependent spine pruning. These findings identify a novel nucleus-enriched deubiquitinase that plays critical roles in synapse remodeling.
Collapse
Affiliation(s)
- Qi Ma
- Departments of Psychiatry and Neuroscience, State University of New York, Upstate Medical University, Syracuse, NY 13210
| | - Hongyu Ruan
- Departments of Psychiatry and Neuroscience, State University of New York, Upstate Medical University, Syracuse, NY 13210
| | - Huihui Dai
- Departments of Psychiatry and Neuroscience, State University of New York, Upstate Medical University, Syracuse, NY 13210
| | - Wei-Dong Yao
- Departments of Psychiatry and Neuroscience, State University of New York, Upstate Medical University, Syracuse, NY 13210
| |
Collapse
|
9
|
Dysregulated Signaling at Postsynaptic Density: A Systematic Review and Translational Appraisal for the Pathophysiology, Clinics, and Antipsychotics' Treatment of Schizophrenia. Cells 2023; 12:cells12040574. [PMID: 36831241 PMCID: PMC9954794 DOI: 10.3390/cells12040574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 02/07/2023] [Accepted: 02/08/2023] [Indexed: 02/12/2023] Open
Abstract
Emerging evidence from genomics, post-mortem, and preclinical studies point to a potential dysregulation of molecular signaling at postsynaptic density (PSD) in schizophrenia pathophysiology. The PSD that identifies the archetypal asymmetric synapse is a structure of approximately 300 nm in diameter, localized behind the neuronal membrane in the glutamatergic synapse, and constituted by more than 1000 proteins, including receptors, adaptors, kinases, and scaffold proteins. Furthermore, using FASS (fluorescence-activated synaptosome sorting) techniques, glutamatergic synaptosomes were isolated at around 70 nm, where the receptors anchored to the PSD proteins can diffuse laterally along the PSD and were stabilized by scaffold proteins in nanodomains of 50-80 nm at a distance of 20-40 nm creating "nanocolumns" within the synaptic button. In this context, PSD was envisioned as a multimodal hub integrating multiple signaling-related intracellular functions. Dysfunctions of glutamate signaling have been postulated in schizophrenia, starting from the glutamate receptor's interaction with scaffolding proteins involved in the N-methyl-D-aspartate receptor (NMDAR). Despite the emerging role of PSD proteins in behavioral disorders, there is currently no systematic review that integrates preclinical and clinical findings addressing dysregulated PSD signaling and translational implications for antipsychotic treatment in the aberrant postsynaptic function context. Here we reviewed a critical appraisal of the role of dysregulated PSD proteins signaling in the pathophysiology of schizophrenia, discussing how antipsychotics may affect PSD structures and synaptic plasticity in brain regions relevant to psychosis.
Collapse
|
10
|
KASAI H. Unraveling the mysteries of dendritic spine dynamics: Five key principles shaping memory and cognition. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2023; 99:254-305. [PMID: 37821392 PMCID: PMC10749395 DOI: 10.2183/pjab.99.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Accepted: 07/11/2023] [Indexed: 10/13/2023]
Abstract
Recent research extends our understanding of brain processes beyond just action potentials and chemical transmissions within neural circuits, emphasizing the mechanical forces generated by excitatory synapses on dendritic spines to modulate presynaptic function. From in vivo and in vitro studies, we outline five central principles of synaptic mechanics in brain function: P1: Stability - Underpinning the integral relationship between the structure and function of the spine synapses. P2: Extrinsic dynamics - Highlighting synapse-selective structural plasticity which plays a crucial role in Hebbian associative learning, distinct from pathway-selective long-term potentiation (LTP) and depression (LTD). P3: Neuromodulation - Analyzing the role of G-protein-coupled receptors, particularly dopamine receptors, in time-sensitive modulation of associative learning frameworks such as Pavlovian classical conditioning and Thorndike's reinforcement learning (RL). P4: Instability - Addressing the intrinsic dynamics crucial to memory management during continual learning, spotlighting their role in "spine dysgenesis" associated with mental disorders. P5: Mechanics - Exploring how synaptic mechanics influence both sides of synapses to establish structural traces of short- and long-term memory, thereby aiding the integration of mental functions. We also delve into the historical background and foresee impending challenges.
Collapse
Affiliation(s)
- Haruo KASAI
- International Research Center for Neurointelligence (WPI-IRCN), UTIAS, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Laboratory of Structural Physiology, Center for Disease Biology and Integrative Medicine, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
11
|
Heck N, Santos MD. Dendritic Spines in Learning and Memory: From First Discoveries to Current Insights. ADVANCES IN NEUROBIOLOGY 2023; 34:311-348. [PMID: 37962799 DOI: 10.1007/978-3-031-36159-3_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
The central nervous system is composed of neural ensembles, and their activity patterns are neural correlates of cognitive functions. Those ensembles are networks of neurons connected to each other by synapses. Most neurons integrate synaptic signal through a remarkable subcellular structure called spine. Dendritic spines are protrusions whose diverse shapes make them appear as a specific neuronal compartment, and they have been the focus of studies for more than a century. Soon after their first description by Ramón y Cajal, it has been hypothesized that spine morphological changes could modify neuronal connectivity and sustain cognitive abilities. Later studies demonstrated that changes in spine density and morphology occurred in experience-dependent plasticity during development, and in clinical cases of mental retardation. This gave ground for the assumption that dendritic spines are the particular locus of cerebral plasticity. With the discovery of synaptic long-term potentiation, a research program emerged with the aim to establish whether dendritic spine plasticity could explain learning and memory. The development of live imaging methods revealed on the one hand that dendritic spine remodeling is compatible with learning process and, on the other hand, that their long-term stability is compatible with lifelong memories. Furthermore, the study of the mechanisms of spine growth and maintenance shed new light on the rules of plasticity. In behavioral paradigms of memory, spine formation or elimination and morphological changes were found to correlate with learning. In a last critical step, recent experiments have provided evidence that dendritic spines play a causal role in learning and memory.
Collapse
Affiliation(s)
- Nicolas Heck
- Laboratory Neurosciences Paris Seine, Sorbonne Université, Paris, France.
| | - Marc Dos Santos
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
12
|
Petanjek Z, Banovac I, Sedmak D, Hladnik A. Dendritic Spines: Synaptogenesis and Synaptic Pruning for the Developmental Organization of Brain Circuits. ADVANCES IN NEUROBIOLOGY 2023; 34:143-221. [PMID: 37962796 DOI: 10.1007/978-3-031-36159-3_4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Synaptic overproduction and elimination is a regular developmental event in the mammalian brain. In the cerebral cortex, synaptic overproduction is almost exclusively correlated with glutamatergic synapses located on dendritic spines. Therefore, analysis of changes in spine density on different parts of the dendritic tree in identified classes of principal neurons could provide insight into developmental reorganization of specific microcircuits.The activity-dependent stabilization and selective elimination of the initially overproduced synapses is a major mechanism for generating diversity of neural connections beyond their genetic determination. The largest number of overproduced synapses was found in the monkey and human cerebral cortex. The highest (exceeding adult values by two- to threefold) and most protracted overproduction (up to third decade of life) was described for associative layer IIIC pyramidal neurons in the human dorsolateral prefrontal cortex.Therefore, the highest proportion and extraordinarily extended phase of synaptic spine overproduction is a hallmark of neural circuitry in human higher-order associative areas. This indicates that microcircuits processing the most complex human cognitive functions have the highest level of developmental plasticity. This finding is the backbone for understanding the effect of environmental impact on the development of the most complex, human-specific cognitive and emotional capacities, and on the late onset of human-specific neuropsychiatric disorders, such as autism and schizophrenia.
Collapse
Affiliation(s)
- Zdravko Petanjek
- Department of Anatomy and Clinical Anatomy, School of Medicine, University of Zagreb, Zagreb, Croatia.
- Department of Neuroscience, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia.
- Center of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine, University of Zagreb, Zagreb, Croatia.
| | - Ivan Banovac
- Department of Anatomy and Clinical Anatomy, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Neuroscience, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Center of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Dora Sedmak
- Department of Anatomy and Clinical Anatomy, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Neuroscience, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Center of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Ana Hladnik
- Department of Anatomy and Clinical Anatomy, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Neuroscience, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Center of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine, University of Zagreb, Zagreb, Croatia
| |
Collapse
|
13
|
Roy A, Sharma S, Nag TC, Katyal J, Gupta YK, Jain S. Cognitive Dysfunction and Anxiety Resulting from Synaptic Downscaling, Hippocampal Atrophy, and Ventricular Enlargement with Intracerebroventricular Streptozotocin Injection in Male Wistar Rats. Neurotox Res 2022; 40:2179-2202. [PMID: 36069980 DOI: 10.1007/s12640-022-00563-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 08/04/2022] [Accepted: 08/18/2022] [Indexed: 12/31/2022]
Abstract
Insulin-resistant brain state is proposed to be the early sign of Alzheimer's disease (AD), which can be studied in the intracerebroventricular streptozotocin (ICV-STZ) rodent model. ICV-STZ is reported to induce sporadic AD with the majority of the disease hallmarks as phenotype. On the other hand, available experimental evidence has used varying doses of STZ (< 1 to 3 mg/kg) and studied its effect for different study durations, ranging from 14 to 270 days. Though these studies suggest 3 mg/kg of ICV-STZ to be the optimum dose for progressive pathogenesis, the reason for such is elusive. Here, we sought to investigate the mechanism of action of 3 mg/kg ICV-STZ on cognitive and non-cognitive aspects at a follow-up interval of 2 weeks for 2 months. On the 60th day, we examined the layer thickness, cell density, ventricular volume, spine density, protein expression related to brain metabolism, and mitochondrial function by histological examination. The findings suggest a progressive loss of a spatial, episodic, and avoidance memory with an increase in anxiety in a span of 2 months. Furthermore, hippocampal neurodegeneration, ventricular enlargement, diffused amyloid plaque deposition, loss of spine in the dentate gyrus, and imbalance in energy homeostasis were found on the 60th day post-injection. Interestingly, AD rats showed a uniform fraction of time spent in four quadrants of the water maze with a change in strategy when they were exposed to height. Our findings reveal that ICV-STZ injection at a dose of 3 mg/kg can cause cognitive and neuropsychiatric abnormalities due to structural loss both at the neuronal as well as the synaptic level, which is tightly associated with the change in neuronal metabolism.
Collapse
Affiliation(s)
- Avishek Roy
- Department of Physiology, All India Institute of Medical Sciences, New Delhi, India. .,UMR-5297, Interdisciplinary Institute of Neurosciences, University of Bordeaux, Bordeaux, France.
| | - Sakshi Sharma
- School of Interdisciplinary Research, Indian Institute of Technology, Delhi, India
| | - Tapas Chandra Nag
- Department of Anatomy, All India Institute of Medical Sciences, Delhi, India
| | - Jatinder Katyal
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, India
| | | | - Suman Jain
- Department of Physiology, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
14
|
GLAST versus GFAP as astroglial marker for the subcellular study of cannabinoid CB 1 receptors in astrocytes. Histochem Cell Biol 2022; 158:561-569. [PMID: 35852615 DOI: 10.1007/s00418-022-02139-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/06/2022] [Indexed: 12/13/2022]
Abstract
The cannabinoid CB1 receptor-mediated functions in astrocytes are highly dependent on the CB1 receptor distribution in these glial cells relative to neuronal sites, particularly at the nearby synapses under normal or pathological conditions. However, the portrait of the CB1 receptor distribution in astroglial compartments remains uncompleted because of the scarce CB1 receptor expression in these cells and the limited identification of astrocytes. The glial fibrillary acidic protein (GFAP) is commonly used as astroglial marker. However, because GFAP is a cytoskeleton protein mostly restricted to the astroglial cell bodies and their main branches, it seems not ideal for the localization of CB1 receptor distribution in astrocytes. Therefore, alternative markers to decipher the actual astroglial CB1 receptors are required. In this work, we have compared the glutamate aspartate transporter (GLAST) versus GFAP for the CB1 receptor localization in astrocytes. We found by immunoelectron microscopy that GLAST reveals almost three-fold astroglial area and four-fold astroglial membranes compared to GFAP. In addition, this better visualization of astrocytes was associated with the detection of 12% of the total CB1 receptor labeling in GLAST-positive astrocytes.
Collapse
|
15
|
Phldb2 is essential for regulating hippocampal dendritic spine morphology through drebrin in an adult-type isoform-specific manner. Neurosci Res 2022; 185:1-10. [PMID: 36162735 DOI: 10.1016/j.neures.2022.09.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 09/16/2022] [Accepted: 09/21/2022] [Indexed: 11/24/2022]
Abstract
Morphologically dynamic dendritic spines are the major sites of neuronal plasticity in the brain; however, the molecular mechanisms underlying their morphological dynamics have not been fully elucidated. Phldb2 is a protein that contains two predicted coiled-coil domains and the pleckstrin homology domain, whose binding is highly sensitive to PIP3. We have previously demonstrated that Phldb2 regulates synaptic plasticity, glutamate receptor trafficking, and PSD-95 turnover. Drebrin is one of the most abundant neuron-specific F-actin-binding proteins that are pivotal for synaptic morphology and plasticity. We observed that Phldb2 bound to drebrin A (adult-type drebrin), but not to drebrin E (embryonic-type drebrin). In the absence of Phldb2, the subcellular localization of drebrin A in the hippocampal spines and its distribution in the hippocampus were altered. Immature spines, such as the filopodium type, increased relatively in the CA1 regions of the hippocampus, whereas mushroom spines, a typical mature type, decreased in Phldb2-/- mice. Phldb2 suppressed the formation of an abnormal filopodium structure induced by drebrin A overexpression. Taken together, these findings demonstrate that Phldb2 is pivotal for dendritic spine morphology and possibly for synaptic plasticity in mature animals by regulating drebrin A localization.
Collapse
|
16
|
Mariano A, Bovio CL, Criscuolo V, Santoro F. Bioinspired micro- and nano-structured neural interfaces. NANOTECHNOLOGY 2022; 33:492501. [PMID: 35947922 DOI: 10.1088/1361-6528/ac8881] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 08/10/2022] [Indexed: 06/15/2023]
Abstract
The development of a functional nervous system requires neurons to interact with and promptly respond to a wealth of biochemical, mechanical and topographical cues found in the neural extracellular matrix (ECM). Among these, ECM topographical cues have been found to strongly influence neuronal function and behavior. Here, we discuss how the blueprint of the architectural organization of the brain ECM has been tremendously useful as a source of inspiration to design biomimetic substrates to enhance neural interfaces and dictate neuronal behavior at the cell-material interface. In particular, we focus on different strategies to recapitulate cell-ECM and cell-cell interactions. In order to mimic cell-ECM interactions, we introduce roughness as a first approach to provide informative topographical biomimetic cues to neurons. We then examine 3D scaffolds and hydrogels, as softer 3D platforms for neural interfaces. Moreover, we will discuss how anisotropic features such as grooves and fibers, recapitulating both ECM fibrils and axonal tracts, may provide recognizable paths and tracks that neuron can follow as they develop and establish functional connections. Finally, we show how isotropic topographical cues, recapitulating shapes, and geometries of filopodia- and mushroom-like dendritic spines, have been instrumental to better reproduce neuron-neuron interactions for applications in bioelectronics and neural repair strategies. The high complexity of the brain architecture makes the quest for the fabrication of create more biologically relevant biomimetic architectures in continuous and fast development. Here, we discuss how recent advancements in two-photon polymerization and remotely reconfigurable dynamic interfaces are paving the way towards to a new class of smart biointerfaces forin vitroapplications spanning from neural tissue engineering as well as neural repair strategies.
Collapse
Affiliation(s)
- Anna Mariano
- Tissue Electronics, Istituto Italiano di Tecnologia, I-80125 Naples, Italy
| | - Claudia Latte Bovio
- Tissue Electronics, Istituto Italiano di Tecnologia, I-80125 Naples, Italy
- Dipartimento di Chimica, Materiali e Produzione Industriale, Università di Napoli Federico II, I-80125, Naples, Italy
| | - Valeria Criscuolo
- Faculty of Electrical Engineering and IT, RWTH Aachen, D-52074, Germany
| | - Francesca Santoro
- Tissue Electronics, Istituto Italiano di Tecnologia, I-80125 Naples, Italy
- Faculty of Electrical Engineering and IT, RWTH Aachen, D-52074, Germany
- Institute for Biological Information Processing-Bioelectronics, Forschungszentrum Juelich, D-52428, Germany
| |
Collapse
|
17
|
Qi C, Luo LD, Feng I, Ma S. Molecular mechanisms of synaptogenesis. Front Synaptic Neurosci 2022; 14:939793. [PMID: 36176941 PMCID: PMC9513053 DOI: 10.3389/fnsyn.2022.939793] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 07/27/2022] [Indexed: 11/29/2022] Open
Abstract
Synapses are the basic units for information processing and storage in the nervous system. It is only when the synaptic connection is established, that it becomes meaningful to discuss the structure and function of a circuit. In humans, our unparalleled cognitive abilities are correlated with an increase in the number of synapses. Additionally, genes involved in synaptogenesis are also frequently associated with neurological or psychiatric disorders, suggesting a relationship between synaptogenesis and brain physiology and pathology. Thus, understanding the molecular mechanisms of synaptogenesis is the key to the mystery of circuit assembly and neural computation. Furthermore, it would provide therapeutic insights for the treatment of neurological and psychiatric disorders. Multiple molecular events must be precisely coordinated to generate a synapse. To understand the molecular mechanisms underlying synaptogenesis, we need to know the molecular components of synapses, how these molecular components are held together, and how the molecular networks are refined in response to neural activity to generate new synapses. Thanks to the intensive investigations in this field, our understanding of the process of synaptogenesis has progressed significantly. Here, we will review the molecular mechanisms of synaptogenesis by going over the studies on the identification of molecular components in synapses and their functions in synaptogenesis, how cell adhesion molecules connect these synaptic molecules together, and how neural activity mobilizes these molecules to generate new synapses. Finally, we will summarize the human-specific regulatory mechanisms in synaptogenesis and results from human genetics studies on synaptogenesis and brain disorders.
Collapse
Affiliation(s)
- Cai Qi
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, United States
- *Correspondence: Cai Qi,
| | - Li-Da Luo
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, United States
- Department of Cellular and Molecular Physiology, Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT, United States
| | - Irena Feng
- Boston University School of Medicine, Boston, MA, United States
| | - Shaojie Ma
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, United States
| |
Collapse
|
18
|
Kim YJ. Activity-induced synaptic structural modifications by Akt. Biochem Biophys Res Commun 2022; 621:94-100. [PMID: 35820284 DOI: 10.1016/j.bbrc.2022.06.093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 06/28/2022] [Indexed: 11/17/2022]
Abstract
The activity-dependent regulation of synaptic structures plays a key role in synaptic development and plasticity; however, the signaling mechanisms involved remain largely unknown. The serine/threonine protein kinase Akt, a downstream effector of phosphoinositide 3-kinase (PI3K), plays a pivotal role in a wide range of physiological functions. We focused on the importance of Akt in rapid synaptic structural changes after stimulation at the Drosophila neuromuscular junction, a well-studied model synapse. Compared with wild-type larvae, akt mutants showed significantly reduced muscle size and an increased number of boutons per area, suggesting that Akt is required for proper pre- and postsynaptic growth. In addition, the level of cysteine string protein (CSP) was significantly increased, and its distribution was different in akt mutants. After high K+ single stimulation, the CSP level of akt mutant NMJs increased dramatically compared with that of wild-type NMJs. Interestingly, ghost boutons without postsynaptic specialization were found in akt mutant NMJs, and the number of these boutons was significantly increased by patterned stimulation. In contrast, the postsynaptic change in the subsynaptic reticulum (SSR) in the akt mutant occurred independent of stimulation. These results suggest that Akt functions in both pre- and postsynaptic growth and differentiation, and in particular, presynaptic action occurs in an activity-dependent manner.
Collapse
Affiliation(s)
- Yoon-Jung Kim
- Department of Physiology and Neuroscience, Dental Research Institute, Seoul National University School of Dentistry, Seoul, 03080, South Korea.
| |
Collapse
|
19
|
Mohanan AG, Gunasekaran S, Jacob RS, Omkumar RV. Role of Ca2+/Calmodulin-Dependent Protein Kinase Type II in Mediating Function and Dysfunction at Glutamatergic Synapses. Front Mol Neurosci 2022; 15:855752. [PMID: 35795689 PMCID: PMC9252440 DOI: 10.3389/fnmol.2022.855752] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 03/21/2022] [Indexed: 01/25/2023] Open
Abstract
Glutamatergic synapses harbor abundant amounts of the multifunctional Ca2+/calmodulin-dependent protein kinase type II (CaMKII). Both in the postsynaptic density as well as in the cytosolic compartment of postsynaptic terminals, CaMKII plays major roles. In addition to its Ca2+-stimulated kinase activity, it can also bind to a variety of membrane proteins at the synapse and thus exert spatially restricted activity. The abundance of CaMKII in glutamatergic synapse is akin to scaffolding proteins although its prominent function still appears to be that of a kinase. The multimeric structure of CaMKII also confers several functional capabilities on the enzyme. The versatility of the enzyme has prompted hypotheses proposing several roles for the enzyme such as Ca2+ signal transduction, memory molecule function and scaffolding. The article will review the multiple roles played by CaMKII in glutamatergic synapses and how they are affected in disease conditions.
Collapse
Affiliation(s)
- Archana G. Mohanan
- Neurobiology Division, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - Sowmya Gunasekaran
- Neurobiology Division, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
- Research Scholar, Manipal Academy of Higher Education, Manipal, India
| | - Reena Sarah Jacob
- Neurobiology Division, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
- Research Scholar, Manipal Academy of Higher Education, Manipal, India
| | - R. V. Omkumar
- Neurobiology Division, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
- *Correspondence: R. V. Omkumar,
| |
Collapse
|
20
|
Rudolf MA, Andreeva A, Kim CE, DeNovio ACJ, Koshar AN, Baker W, Cartagena-Rivera AX, Corwin JT. Stiffening of Circumferential F-Actin Bands Correlates With Regenerative Failure and May Act as a Biomechanical Brake in the Mammalian Inner Ear. Front Cell Neurosci 2022; 16:859882. [PMID: 35602553 PMCID: PMC9114303 DOI: 10.3389/fncel.2022.859882] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 04/06/2022] [Indexed: 11/13/2022] Open
Abstract
The loss of inner ear hair cells causes permanent hearing and balance deficits in humans and other mammals, but non-mammals recover after supporting cells (SCs) divide and replace hair cells. The proliferative capacity of mammalian SCs declines as exceptionally thick circumferential F-actin bands develop at their adherens junctions. We hypothesized that the reinforced junctions were limiting regenerative responses of mammalian SCs by impeding changes in cell shape and epithelial tension. Using micropipette aspiration and atomic force microscopy, we measured mechanical properties of utricles from mice and chickens. Our data show that the epithelial surface of the mouse utricle stiffens significantly during postnatal maturation. This stiffening correlates with and is dependent on the postnatal accumulation of F-actin and the cross-linker Alpha-Actinin-4 at SC-SC junctions. In chicken utricles, where SCs lack junctional reinforcement, the epithelial surface remains compliant. There, SCs undergo oriented cell divisions and their apical surfaces progressively elongate throughout development, consistent with anisotropic intraepithelial tension. In chicken utricles, inhibition of actomyosin contractility led to drastic SC shape change and epithelial buckling, but neither occurred in mouse utricles. These findings suggest that species differences in the capacity for hair cell regeneration may be attributable in part to the differences in the stiffness and contractility of the actin cytoskeletal elements that reinforce adherens junctions and participate in regulation of the cell cycle.
Collapse
Affiliation(s)
- Mark A. Rudolf
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Anna Andreeva
- School of Sciences and Humanities, Nazarbayev University, Nur-Sultan, Kazakhstan
| | - Christina E. Kim
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Anthony C.-J. DeNovio
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Antoan N. Koshar
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Wendy Baker
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Alexander X. Cartagena-Rivera
- Section on Mechanobiology, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, United States
| | - Jeffrey T. Corwin
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, United States
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA, United States
| |
Collapse
|
21
|
Bonilla-Quintana M, Rangamani P. Can biophysical models of dendritic spines be used to explore synaptic changes associated with addiction? Phys Biol 2022; 19. [PMID: 35508164 DOI: 10.1088/1478-3975/ac6cbe] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 05/04/2022] [Indexed: 11/11/2022]
Abstract
Effective treatments that prevent or reduce drug relapse vulnerability should be developed to relieve the high burden of drug addiction on society. This will only be possible by enhancing the understanding of the molecular mechanisms underlying the neurobiology of addiction. Recent experimental data have shown that dendritic spines, small protrusions from the dendrites that receive excitatory input, of spiny neurons in the nucleus accumbens exhibit morphological changes during drug exposure and withdrawal. Moreover, these changes relate to the characteristic drug-seeking behavior of addiction. However, due to the complexity of the dendritic spines, we do not yet fully understand the processes underlying their structural changes in response to different inputs. We propose that biophysical models can enhance the current understanding of these processes by incorporating different, and sometimes, discrepant experimental data to identify the shared underlying mechanisms and generate experimentally testable hypotheses. This review aims to give an up-to-date report on biophysical models of dendritic spines, focusing on those models that describe their shape changes, which are well-known to relate to learning and memory. Moreover, it examines how these models can enhance our understanding of the effect of the drugs and the synaptic changes during withdrawal, as well as during neurodegenerative disease progression such as Alzheimer's disease.
Collapse
Affiliation(s)
- Mayte Bonilla-Quintana
- Mechanical Aerospace Engineering, University of California San Diego, 9500 Gilman Drive, La Jolla, California, 92093-0021, UNITED STATES
| | - Padmini Rangamani
- Mechanical Aerospace Engineering, University of California San Diego, 9500 Gilman Drive, La Jolla, California, 92093-0021, UNITED STATES
| |
Collapse
|
22
|
Control of Synapse Structure and Function by Actin and Its Regulators. Cells 2022; 11:cells11040603. [PMID: 35203254 PMCID: PMC8869895 DOI: 10.3390/cells11040603] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 01/30/2022] [Accepted: 02/06/2022] [Indexed: 02/07/2023] Open
Abstract
Neurons transmit and receive information at specialized junctions called synapses. Excitatory synapses form at the junction between a presynaptic axon terminal and a postsynaptic dendritic spine. Supporting the shape and function of these junctions is a complex network of actin filaments and its regulators. Advances in microscopic techniques have enabled studies of the organization of actin at synapses and its dynamic regulation. In addition to highlighting recent advances in the field, we will provide a brief historical perspective of the understanding of synaptic actin at the synapse. We will also highlight key neuronal functions regulated by actin, including organization of proteins in the pre- and post- synaptic compartments and endocytosis of ion channels. We review the evidence that synapses contain distinct actin pools that differ in their localization and dynamic behaviors and discuss key functions for these actin pools. Finally, whole exome sequencing of humans with neurodevelopmental and psychiatric disorders has identified synaptic actin regulators as key disease risk genes. We briefly summarize how genetic variants in these genes impact neurotransmission via their impact on synaptic actin.
Collapse
|
23
|
Trujillo-Estrada L, Vanderklish PW, Nguyen MMT, Kuang RR, Nguyen C, Huynh E, da Cunha C, Javonillo DI, Forner S, Martini AC, Sarraf ST, Simmon VF, Baglietto-Vargas D, LaFerla FM. SPG302 Reverses Synaptic and Cognitive Deficits Without Altering Amyloid or Tau Pathology in a Transgenic Model of Alzheimer's Disease. Neurotherapeutics 2021; 18:2468-2483. [PMID: 34738197 PMCID: PMC8804111 DOI: 10.1007/s13311-021-01143-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/11/2021] [Indexed: 12/04/2022] Open
Abstract
Alzheimer's disease (AD) is conceptualized as a synaptic failure disorder in which loss of glutamatergic synapses is a major driver of cognitive decline. Thus, novel therapeutic strategies aimed at regenerating synapses may represent a promising approach to mitigate cognitive deficits in AD patients. At present, no disease-modifying drugs exist for AD, and approved therapies are palliative at best, lacking in the ability to reverse the synaptic failure. Here, we tested the efficacy of a novel synaptogenic small molecule, SPG302 - a 3rd-generation benzothiazole derivative that increases the density of axospinous glutamatergic synapses - in 3xTg-AD mice. Daily dosing of 3xTg-AD mice with SPG302 at 3 and 30 mg/kg (i.p.) for 4 weeks restored hippocampal synaptic density and improved cognitive function in hippocampal-dependent tasks. Mushroom and stubby spine profiles were increased by SPG302, and associated with enhanced expression of key postsynaptic proteins - including postsynaptic density protein 95 (PSD95), drebrin, and amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) - and increased colocalization of PSD95 with synaptophysin. Notably, SPG302 proved efficacious in this model without modifying Aβ and tau pathology. Thus, our study provides preclinical support for the idea that compounds capable of restoring synaptic density offer a viable strategy to reverse cognitive decline in AD.
Collapse
Affiliation(s)
- Laura Trujillo-Estrada
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA
- Departamento Biología Celular, Genetica y Fisiologia, Instituto de Investigacion Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, Malaga, Spain
- Centro de Investigacion Biomedica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Peter W Vanderklish
- Spinogenix Inc, 10210 Campus Point Drive, Suite 150, San Diego, CA, 92121, USA.
| | - Marie Minh Thu Nguyen
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA
| | - Run Rong Kuang
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA
| | - Caroline Nguyen
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA
| | - Eric Huynh
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA
| | - Celia da Cunha
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA
| | - Dominic Ibarra Javonillo
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA
| | - Stefania Forner
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA
| | - Alessandra C Martini
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA
| | - Stella T Sarraf
- Spinogenix Inc, 10210 Campus Point Drive, Suite 150, San Diego, CA, 92121, USA
| | - Vincent F Simmon
- Spinogenix Inc, 10210 Campus Point Drive, Suite 150, San Diego, CA, 92121, USA.
| | - David Baglietto-Vargas
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA.
- Department of Neurobiology and Behavior, University of California, Irvine, CA, 92697-1450, USA.
- Departamento Biología Celular, Genetica y Fisiologia, Instituto de Investigacion Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, Malaga, Spain.
- Centro de Investigacion Biomedica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
| | - Frank M LaFerla
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA.
- Department of Neurobiology and Behavior, University of California, Irvine, CA, 92697-1450, USA.
| |
Collapse
|
24
|
Presynaptic AMPA Receptors in Health and Disease. Cells 2021; 10:cells10092260. [PMID: 34571906 PMCID: PMC8470629 DOI: 10.3390/cells10092260] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/23/2021] [Accepted: 08/26/2021] [Indexed: 01/04/2023] Open
Abstract
AMPA receptors (AMPARs) are ionotropic glutamate receptors that play a major role in excitatory neurotransmission. AMPARs are located at both presynaptic and postsynaptic plasma membranes. A huge number of studies investigated the role of postsynaptic AMPARs in the normal and abnormal functioning of the mammalian central nervous system (CNS). These studies highlighted that changes in the functional properties or abundance of postsynaptic AMPARs are major mechanisms underlying synaptic plasticity phenomena, providing molecular explanations for the processes of learning and memory. Conversely, the role of AMPARs at presynaptic terminals is as yet poorly clarified. Accruing evidence demonstrates that presynaptic AMPARs can modulate the release of various neurotransmitters. Recent studies also suggest that presynaptic AMPARs may possess double ionotropic-metabotropic features and that they are involved in the local regulation of actin dynamics in both dendritic and axonal compartments. In addition, evidence suggests a key role of presynaptic AMPARs in axonal pathology, in regulation of pain transmission and in the physiology of the auditory system. Thus, it appears that presynaptic AMPARs play an important modulatory role in nerve terminal activity, making them attractive as novel pharmacological targets for a variety of pathological conditions.
Collapse
|
25
|
Cornelius J, Rottner K, Korte M, Michaelsen-Preusse K. Cortactin Contributes to Activity-Dependent Modulation of Spine Actin Dynamics and Spatial Memory Formation. Cells 2021; 10:cells10071835. [PMID: 34360003 PMCID: PMC8303107 DOI: 10.3390/cells10071835] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/13/2021] [Accepted: 07/16/2021] [Indexed: 01/21/2023] Open
Abstract
Postsynaptic structures on excitatory neurons, dendritic spines, are actin-rich. It is well known that actin-binding proteins regulate actin dynamics and by this means orchestrate structural plasticity during the development of the brain, as well as synaptic plasticity mediating learning and memory processes. The actin-binding protein cortactin is localized to pre- and postsynaptic structures and translocates in a stimulus-dependent manner between spines and the dendritic compartment, thereby indicating a crucial role for synaptic plasticity and neuronal function. While it is known that cortactin directly binds F-actin, the Arp2/3 complex important for actin nucleation and branching as well as other factors involved in synaptic plasticity processes, its precise role in modulating actin remodeling in neurons needs to be deciphered. In this study, we characterized the general neuronal function of cortactin in knockout mice. Interestingly, we found that the loss of cortactin leads to deficits in hippocampus-dependent spatial memory formation. This impairment is correlated with a prominent dysregulation of functional and structural plasticity. Additional evidence shows impaired long-term potentiation in cortactin knockout mice together with a complete absence of structural spine plasticity. These phenotypes might at least in part be explained by alterations in the activity-dependent modulation of synaptic actin in cortactin-deficient neurons.
Collapse
Affiliation(s)
- Jonas Cornelius
- Division of Cellular Neurobiology, Zoological Institute, TU Braunschweig, 38106 Braunschweig, Germany; (J.C.); (M.K.)
| | - Klemens Rottner
- Research Group Molecular Cell Biology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany;
- Division of Molecular Cell Biology, Zoological Institute, TU Braunschweig, 38106 Braunschweig, Germany
| | - Martin Korte
- Division of Cellular Neurobiology, Zoological Institute, TU Braunschweig, 38106 Braunschweig, Germany; (J.C.); (M.K.)
- Research Group Neuroinflammation and Neurodegeneration, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Kristin Michaelsen-Preusse
- Division of Cellular Neurobiology, Zoological Institute, TU Braunschweig, 38106 Braunschweig, Germany; (J.C.); (M.K.)
- Correspondence:
| |
Collapse
|
26
|
Vukovic N, Hansen B, Lund TE, Jespersen S, Shtyrov Y. Rapid microstructural plasticity in the cortical semantic network following a short language learning session. PLoS Biol 2021; 19:e3001290. [PMID: 34125828 PMCID: PMC8202930 DOI: 10.1371/journal.pbio.3001290] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 05/17/2021] [Indexed: 01/22/2023] Open
Abstract
Despite the clear importance of language in our life, our vital ability to quickly and effectively learn new words and meanings is neurobiologically poorly understood. Conventional knowledge maintains that language learning—especially in adulthood—is slow and laborious. Furthermore, its structural basis remains unclear. Even though behavioural manifestations of learning are evident near instantly, previous neuroimaging work across a range of semantic categories has largely studied neural changes associated with months or years of practice. Here, we address rapid neuroanatomical plasticity accompanying new lexicon acquisition, specifically focussing on the learning of action-related language, which has been linked to the brain’s motor systems. Our results show that it is possible to measure and to externally modulate (using transcranial magnetic stimulation (TMS) of motor cortex) cortical microanatomic reorganisation after mere minutes of new word learning. Learning-induced microstructural changes, as measured by diffusion kurtosis imaging (DKI) and machine learning-based analysis, were evident in prefrontal, temporal, and parietal neocortical sites, likely reflecting integrative lexico-semantic processing and formation of new memory circuits immediately during the learning tasks. These results suggest a structural basis for the rapid neocortical word encoding mechanism and reveal the causally interactive relationship of modal and associative brain regions in supporting learning and word acquisition. This combined neuroimaging and brain stimulation study reveals rapid and distributed microstructural plasticity after a single immersive language learning session, demonstrating the causal relevance of the motor cortex in encoding the meaning of novel action words.
Collapse
Affiliation(s)
- Nikola Vukovic
- Department of Psychiatry, University of California San Francisco, San Francisco, United States of America
- * E-mail:
| | - Brian Hansen
- Center of Functionally Integrative Neuroscience, Aarhus University, Aarhus, Denmark
| | | | - Sune Jespersen
- Center of Functionally Integrative Neuroscience, Aarhus University, Aarhus, Denmark
- Department of Physics and Astronomy, Aarhus University, Aarhus, Denmark
| | - Yury Shtyrov
- Center of Functionally Integrative Neuroscience, Aarhus University, Aarhus, Denmark
- Centre for Cognition and Decision making, HSE University, Moscow, Russia
| |
Collapse
|
27
|
Zhong H, Ceballos CC, Massengill CI, Muniak MA, Ma L, Qin M, Petrie SK, Mao T. High-fidelity, efficient, and reversible labeling of endogenous proteins using CRISPR-based designer exon insertion. eLife 2021; 10:64911. [PMID: 34100715 PMCID: PMC8211447 DOI: 10.7554/elife.64911] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Accepted: 06/07/2021] [Indexed: 12/26/2022] Open
Abstract
Precise and efficient insertion of large DNA fragments into somatic cells using gene editing technologies to label or modify endogenous proteins remains challenging. Non-specific insertions/deletions (INDELs) resulting from the non-homologous end joining pathway make the process error-prone. Further, the insert is not readily removable. Here, we describe a method called CRISPR-mediated insertion of exon (CRISPIE) that can precisely and reversibly label endogenous proteins using CRISPR/Cas9-based editing. CRISPIE inserts a designer donor module, which consists of an exon encoding the protein sequence flanked by intron sequences, into an intronic location in the target gene. INDELs at the insertion junction will be spliced out, leaving mRNAs nearly error-free. We used CRISPIE to fluorescently label endogenous proteins in mammalian neurons in vivo with previously unachieved efficiency. We demonstrate that this method is broadly applicable, and that the insert can be readily removed later. CRISPIE permits protein sequence insertion with high fidelity, efficiency, and flexibility.
Collapse
Affiliation(s)
- Haining Zhong
- Vollum Institute, Oregon Health & Science University, Portland, United States
| | - Cesar C Ceballos
- Vollum Institute, Oregon Health & Science University, Portland, United States
| | | | - Michael A Muniak
- Vollum Institute, Oregon Health & Science University, Portland, United States
| | - Lei Ma
- Vollum Institute, Oregon Health & Science University, Portland, United States
| | - Maozhen Qin
- Vollum Institute, Oregon Health & Science University, Portland, United States
| | - Stefanie Kaech Petrie
- Department of Neurology, Oregon Health & Science University, Portland, United States
| | - Tianyi Mao
- Vollum Institute, Oregon Health & Science University, Portland, United States
| |
Collapse
|
28
|
Kasai H, Ziv NE, Okazaki H, Yagishita S, Toyoizumi T. Spine dynamics in the brain, mental disorders and artificial neural networks. Nat Rev Neurosci 2021; 22:407-422. [PMID: 34050339 DOI: 10.1038/s41583-021-00467-3] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/14/2021] [Indexed: 12/15/2022]
Abstract
In the brain, most synapses are formed on minute protrusions known as dendritic spines. Unlike their artificial intelligence counterparts, spines are not merely tuneable memory elements: they also embody algorithms that implement the brain's ability to learn from experience and cope with new challenges. Importantly, they exhibit structural dynamics that depend on activity, excitatory input and inhibitory input (synaptic plasticity or 'extrinsic' dynamics) and dynamics independent of activity ('intrinsic' dynamics), both of which are subject to neuromodulatory influences and reinforcers such as dopamine. Here we succinctly review extrinsic and intrinsic dynamics, compare these with parallels in machine learning where they exist, describe the importance of intrinsic dynamics for memory management and adaptation, and speculate on how disruption of extrinsic and intrinsic dynamics may give rise to mental disorders. Throughout, we also highlight algorithmic features of spine dynamics that may be relevant to future artificial intelligence developments.
Collapse
Affiliation(s)
- Haruo Kasai
- Laboratory of Structural Physiology, Center for Disease Biology and Integrative Medicine, Faculty of Medicine, The University of Tokyo, Tokyo, Japan. .,International Research Center for Neurointelligence (WPI-IRCN), UTIAS, The University of Tokyo, Bunkyo-ku, Tokyo, Japan.
| | - Noam E Ziv
- Technion Faculty of Medicine and Network Biology Research Labs, Technion City, Haifa, Israel
| | - Hitoshi Okazaki
- Laboratory of Structural Physiology, Center for Disease Biology and Integrative Medicine, Faculty of Medicine, The University of Tokyo, Tokyo, Japan.,International Research Center for Neurointelligence (WPI-IRCN), UTIAS, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Sho Yagishita
- Laboratory of Structural Physiology, Center for Disease Biology and Integrative Medicine, Faculty of Medicine, The University of Tokyo, Tokyo, Japan.,International Research Center for Neurointelligence (WPI-IRCN), UTIAS, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Taro Toyoizumi
- Laboratory for Neural Computation and Adaptation, RIKEN Center for Brain Science, Saitama, Japan.,Department of Mathematical Informatics, Graduate School of Information Science and Technology, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
29
|
Mizuki Y, Sakamoto S, Okahisa Y, Yada Y, Hashimoto N, Takaki M, Yamada N. Mechanisms Underlying the Comorbidity of Schizophrenia and Type 2 Diabetes Mellitus. Int J Neuropsychopharmacol 2021; 24:367-382. [PMID: 33315097 PMCID: PMC8130204 DOI: 10.1093/ijnp/pyaa097] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 11/29/2020] [Accepted: 12/10/2020] [Indexed: 02/07/2023] Open
Abstract
The mortality rate of patients with schizophrenia is high, and life expectancy is shorter by 10 to 20 years. Metabolic abnormalities including type 2 diabetes mellitus (T2DM) are among the main reasons. The prevalence of T2DM in patients with schizophrenia may be epidemiologically frequent because antipsychotics induce weight gain as a side effect and the cognitive dysfunction of patients with schizophrenia relates to a disordered lifestyle, poor diet, and low socioeconomic status. Apart from these common risk factors and risk factors unique to schizophrenia, accumulating evidence suggests the existence of common susceptibility genes between schizophrenia and T2DM. Functional proteins translated from common genetic susceptibility genes are known to regulate neuronal development in the brain and insulin in the pancreas through several common cascades. In this review, we discuss common susceptibility genes, functional cascades, and the relationship between schizophrenia and T2DM. Many genetic and epidemiological studies have reliably associated the comorbidity of schizophrenia and T2DM, and it is probably safe to think that common cascades and mechanisms suspected from common genes' functions are related to the onset of both schizophrenia and T2DM. On the other hand, even when genetic analyses are performed on a relatively large number of comorbid patients, the results are sometimes inconsistent, and susceptibility genes may carry only a low or moderate risk. We anticipate future directions in this field.
Collapse
Affiliation(s)
- Yutaka Mizuki
- Department of Neuropsychiatry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences
- Shimonoseki Hospital
| | - Shinji Sakamoto
- Department of Neuropsychiatry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences
| | - Yuko Okahisa
- Department of Neuropsychiatry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences
| | - Yuji Yada
- Department of Neuropsychiatry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences
- Okayama Psychiatric Medical Center
| | - Nozomu Hashimoto
- Department of Neuropsychiatry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences
- Okayama Psychiatric Medical Center
| | - Manabu Takaki
- Department of Neuropsychiatry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences
| | - Norihito Yamada
- Department of Neuropsychiatry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences
| |
Collapse
|
30
|
Chemical Stimulation of Rodent and Human Cortical Synaptosomes: Implications in Neurodegeneration. Cells 2021; 10:cells10051174. [PMID: 34065927 PMCID: PMC8151714 DOI: 10.3390/cells10051174] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 04/29/2021] [Accepted: 05/09/2021] [Indexed: 12/14/2022] Open
Abstract
Synaptic plasticity events, including long-term potentiation (LTP), are often regarded as correlates of brain functions of memory and cognition. One of the central players in these plasticity-related phenomena is the α-amino-3-hydroxy-5-methylisoxazole-4-propionate receptor (AMPAR). Increased levels of AMPARs on postsynaptic membranes thus constitute a biochemical measure of LTP. Isolated synaptic terminals (synaptosomes) are an excellent ex vivo tool to monitor synaptic physiology in healthy and diseased brains, particularly in human research. We herein describe three protocols for chemically-induced LTP (cLTP) in synaptosomes from both rodent and human brain tissues. Two of these chemical stimulation protocols are described for the first time in synaptosomes. A pharmacological block of synaptosomal actin dynamics confirmed the efficiency of the cLTP protocols. Furthermore, the study prototypically evaluated the deficiency of cLTP in cortical synaptosomes obtained from human cases of early-onset Alzheimer’s disease (EOAD) and frontotemporal lobar degeneration (FLTD), as well as an animal model that mimics FLTD.
Collapse
|
31
|
Schöpf CL, Ablinger C, Geisler SM, Stanika RI, Campiglio M, Kaufmann WA, Nimmervoll B, Schlick B, Brockhaus J, Missler M, Shigemoto R, Obermair GJ. Presynaptic α 2δ subunits are key organizers of glutamatergic synapses. Proc Natl Acad Sci U S A 2021; 118:e1920827118. [PMID: 33782113 PMCID: PMC8040823 DOI: 10.1073/pnas.1920827118] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
In nerve cells the genes encoding for α2δ subunits of voltage-gated calcium channels have been linked to synaptic functions and neurological disease. Here we show that α2δ subunits are essential for the formation and organization of glutamatergic synapses. Using a cellular α2δ subunit triple-knockout/knockdown model, we demonstrate a failure in presynaptic differentiation evidenced by defective presynaptic calcium channel clustering and calcium influx, smaller presynaptic active zones, and a strongly reduced accumulation of presynaptic vesicle-associated proteins (synapsin and vGLUT). The presynaptic defect is associated with the downscaling of postsynaptic AMPA receptors and the postsynaptic density. The role of α2δ isoforms as synaptic organizers is highly redundant, as each individual α2δ isoform can rescue presynaptic calcium channel trafficking and expression of synaptic proteins. Moreover, α2δ-2 and α2δ-3 with mutated metal ion-dependent adhesion sites can fully rescue presynaptic synapsin expression but only partially calcium channel trafficking, suggesting that the regulatory role of α2δ subunits is independent from its role as a calcium channel subunit. Our findings influence the current view on excitatory synapse formation. First, our study suggests that postsynaptic differentiation is secondary to presynaptic differentiation. Second, the dependence of presynaptic differentiation on α2δ implicates α2δ subunits as potential nucleation points for the organization of synapses. Finally, our results suggest that α2δ subunits act as transsynaptic organizers of glutamatergic synapses, thereby aligning the synaptic active zone with the postsynaptic density.
Collapse
Affiliation(s)
- Clemens L Schöpf
- Institute of Physiology, Medical University of Innsbruck, A-6020 Innsbruck, Austria
| | - Cornelia Ablinger
- Institute of Physiology, Medical University of Innsbruck, A-6020 Innsbruck, Austria
| | - Stefanie M Geisler
- Institute of Physiology, Medical University of Innsbruck, A-6020 Innsbruck, Austria
- Department of Pharmacology and Toxicology, University of Innsbruck, A-6020 Innsbruck, Austria
| | - Ruslan I Stanika
- Division of Physiology, Karl Landsteiner University of Health Sciences, A-3500 Krems, Austria
| | - Marta Campiglio
- Institute of Physiology, Medical University of Innsbruck, A-6020 Innsbruck, Austria
| | - Walter A Kaufmann
- Institute of Science and Technology Austria, A-3400 Klosterneuburg, Austria
| | - Benedikt Nimmervoll
- Institute of Physiology, Medical University of Innsbruck, A-6020 Innsbruck, Austria
| | - Bettina Schlick
- Institute of Physiology, Medical University of Innsbruck, A-6020 Innsbruck, Austria
| | - Johannes Brockhaus
- Institute of Anatomy and Molecular Neurobiology, Westfälische Wilhelms University, 48149 Münster, Germany
| | - Markus Missler
- Institute of Anatomy and Molecular Neurobiology, Westfälische Wilhelms University, 48149 Münster, Germany
| | - Ryuichi Shigemoto
- Institute of Science and Technology Austria, A-3400 Klosterneuburg, Austria
| | - Gerald J Obermair
- Institute of Physiology, Medical University of Innsbruck, A-6020 Innsbruck, Austria;
- Division of Physiology, Karl Landsteiner University of Health Sciences, A-3500 Krems, Austria
| |
Collapse
|
32
|
Ube2b-dependent degradation of DNMT3a relieves a transcriptional brake on opiate-induced synaptic and behavioral plasticity. Mol Psychiatry 2021; 26:1162-1177. [PMID: 31576007 DOI: 10.1038/s41380-019-0533-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 09/10/2019] [Accepted: 09/20/2019] [Indexed: 01/01/2023]
Abstract
Compelling evidence suggests that synaptic structural plasticity, driven by remodeling of the actin cytoskeleton, underlies addictive drugs-induced long-lasting behavioral plasticity. However, the signaling mechanisms leading to actin cytoskeleton remodeling remain poorly defined. DNA methylation is a critical mechanism used to control activity-dependent gene expression essential for long-lasting synaptic plasticity. Here, we provide evidence that DNA methyltransferase DNMT3a is degraded by the E2 ubiquitin-conjugating enzyme Ube2b-mediated ubiquitination in dorsal hippocampus (DH) of rats that repeatedly self-administrated heroin. DNMT3a degradation leads to demethylation in CaMKK1 gene promotor, thereby facilitating CaMKK1 expression and consequent activation of its downstream target CaMKIα, an essential regulator of spinogenesis. CaMKK1/CaMKIα signaling regulates actin cytoskeleton remodeling in the DH and behavioral plasticity by activation of Rac1 via acting Rac guanine-nucleotide-exchange factor βPIX. These data suggest that Ube2b-dependent degradation of DNMT3a relieves a transcriptional brake on CaMKK1 gene and thus activates CaMKK1/CaMKIα/βPIX/Rac1 cascade, leading to drug use-induced actin polymerization and behavior plasticity.
Collapse
|
33
|
Bonilla-Quintana M, Wörgötter F. Exploring new roles for actin upon LTP induction in dendritic spines. Sci Rep 2021; 11:7072. [PMID: 33782451 PMCID: PMC8007616 DOI: 10.1038/s41598-021-86367-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 03/15/2021] [Indexed: 01/21/2023] Open
Abstract
Dendritic spines, small protrusions of the dendrites, enlarge upon LTP induction, linking morphological and functional properties. Although the role of actin in spine enlargement has been well studied, little is known about its relationship with mechanical membrane properties, such as membrane tension, which is involved in many cell processes, like exocytosis. Here, we use a 3D model of the dendritic spine to investigate how polymerization of actin filaments can effectively elevate the membrane tension to trigger exocytosis in a domain close to the tip of the spine. Moreover, we show that the same pool of actin promotes full membrane fusion after exocytosis and spine stabilization.
Collapse
|
34
|
Abl2:Cortactin Interactions Regulate Dendritic Spine Stability via Control of a Stable Filamentous Actin Pool. J Neurosci 2021; 41:3068-3081. [PMID: 33622779 DOI: 10.1523/jneurosci.2472-20.2021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 01/15/2021] [Accepted: 02/16/2021] [Indexed: 11/21/2022] Open
Abstract
Dendritic spines act as the receptive contacts at most excitatory synapses. Spines are enriched in a network of actin filaments comprised of two kinetically distinct pools. The majority of spine actin is highly dynamic and regulates spine size, structural plasticity, and postsynaptic density organization. The remainder of the spine actin network is more stable, but the function of this minor actin population is not well understood, as tools to study it have not been available. Previous work has shown that disruption of the Abl2/Arg nonreceptor tyrosine kinase in mice compromises spine stability and size. Here, using cultured hippocampal neurons pooled from both sexes of mice, we provide evidence that binding to cortactin tethers Abl2 in spines, where Abl2 and cortactin maintain the small pool of stable actin required for dendritic spine stability. Using fluorescence recovery after photobleaching of GFP-actin, we find that disruption of Abl2:cortactin interactions eliminates stable actin filaments in dendritic spines, significantly reducing spine density. A subset of spines remaining after Abl2 depletion retain their stable actin pool and undergo activity-dependent spine enlargement, associated with increased cortactin and GluN2B levels. Finally, tonic increases in synaptic activity rescue spine loss following Abl2 depletion by promoting cortactin enrichment in vulnerable spines. Together, our findings strongly suggest that Abl2:cortactin interactions promote spine stability by maintaining pools of stable actin filaments in spines.SIGNIFICANCE STATEMENT Dendritic spines contain two kinetically distinct pools of actin. The more abundant, highly dynamic pool regulates spine shape, size, and plasticity. The function of the smaller, stable actin network is not well understood, as tools to study it have not been available. We demonstrate here that Abl2 and its substrate and interaction partner, cortactin, are essential to maintain the stable pool in spines. Depletion of the stable actin pool via disruption of Abl2 or cortactin, or interactions between the proteins, significantly reduces spine stability. We also provide evidence that tonic increases in synaptic activity promote spine stability via enrichment of cortactin in spines, suggesting that synaptic activity acts on the stable actin pool to stabilize dendritic spines.
Collapse
|
35
|
Wang JL, Wang Y, Sun W, Yu Y, Wei N, Du R, Yang Y, Liang T, Wang XL, Ou CH, Chen J. Spinophilin modulates pain through suppressing dendritic spine morphogenesis via negative control of Rac1-ERK signaling in rat spinal dorsal horn. Neurobiol Dis 2021; 152:105302. [PMID: 33609640 DOI: 10.1016/j.nbd.2021.105302] [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: 07/09/2020] [Revised: 01/07/2021] [Accepted: 02/14/2021] [Indexed: 01/07/2023] Open
Abstract
Both spinophilin (SPN, also known as neurabin 2) and Rac1 (a member of Rho GTPase family) are believed to play key roles in dendritic spine (DS) remodeling and spinal nociception. However, how SPN interacts with Rac1 in the above process is unknown. Here, we first demonstrated natural existence of SPN-protein phosphatase 1-Rac1 complex in the spinal dorsal horn (DH) neurons by both double immunofluorescent labeling and co-immunoprecipitation, then the effects of SPN over-expression and down-regulation on mechanical and thermal pain sensitivity, GTP-bound Rac1-ERK signaling activity, and spinal DS density were studied. Over-expression of SPN in spinal neurons by intra-DH pAAV-CMV-SPN-3FLAG could block both mechanical and thermal pain hypersensitivity induced by intraplantar bee venom injection, however it had no effect on the basal pain sensitivity. Over-expression of SPN also resulted in a significant decrease in GTP-Rac1-ERK activities, relative to naive and irrelevant control (pAAV-MCS). In sharp contrast, knockdown of SPN in spinal neurons by intra-DH pAAV-CAG-eGFP-U6-shRNA[SPN] produced both pain hypersensitivity and dramatic elevation of GTP-Rac1-ERK activities, relative to naive and irrelevant control (pAAV-shRNA [NC]). Moreover, knockdown of SPN resulted in increase in DS density while over-expression of it had no such effect. Collectively, SPN is likely to serve as a regulator of Rac1 signaling to suppress DS morphogenesis via negative control of GTP-bound Rac1-ERK activities at postsynaptic component in rat DH neurons wherein both mechanical and thermal pain sensitivity are controlled.
Collapse
Affiliation(s)
- Jiang-Lin Wang
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an 710038, Shaanxi Province, PR China; Department of Pain Management, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan Province, PR China
| | - Yan Wang
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an 710038, Shaanxi Province, PR China; Key Laboratory of Brain Stress and Behavior, People's Liberation Army, Xi'an 710038, PR China
| | - Wei Sun
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an 710038, Shaanxi Province, PR China; Key Laboratory of Brain Stress and Behavior, People's Liberation Army, Xi'an 710038, PR China
| | - Yang Yu
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an 710038, Shaanxi Province, PR China; Key Laboratory of Brain Stress and Behavior, People's Liberation Army, Xi'an 710038, PR China
| | - Na Wei
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an 710038, Shaanxi Province, PR China; Key Laboratory of Brain Stress and Behavior, People's Liberation Army, Xi'an 710038, PR China
| | - Rui Du
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an 710038, Shaanxi Province, PR China; Key Laboratory of Brain Stress and Behavior, People's Liberation Army, Xi'an 710038, PR China
| | - Yan Yang
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an 710038, Shaanxi Province, PR China; Key Laboratory of Brain Stress and Behavior, People's Liberation Army, Xi'an 710038, PR China
| | - Ting Liang
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an 710038, Shaanxi Province, PR China; Key Laboratory of Brain Stress and Behavior, People's Liberation Army, Xi'an 710038, PR China
| | - Xiao-Liang Wang
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an 710038, Shaanxi Province, PR China; Key Laboratory of Brain Stress and Behavior, People's Liberation Army, Xi'an 710038, PR China
| | - Ce-Hua Ou
- Department of Pain Management, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan Province, PR China
| | - Jun Chen
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an 710038, Shaanxi Province, PR China; Key Laboratory of Brain Stress and Behavior, People's Liberation Army, Xi'an 710038, PR China.
| |
Collapse
|
36
|
Reproducing asymmetrical spine shape fluctuations in a model of actin dynamics predicts self-organized criticality. Sci Rep 2021; 11:4012. [PMID: 33597561 PMCID: PMC7889935 DOI: 10.1038/s41598-021-83331-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 01/29/2021] [Indexed: 12/19/2022] Open
Abstract
Dendritic spines change their size and shape spontaneously, but the function of this remains unclear. Here, we address this in a biophysical model of spine fluctuations, which reproduces experimentally measured spine fluctuations. For this, we characterize size- and shape fluctuations from confocal microscopy image sequences using autoregressive models and a new set of shape descriptors derived from circular statistics. Using the biophysical model, we extrapolate into longer temporal intervals and find the presence of 1/f noise. When investigating its origins, the model predicts that the actin dynamics underlying shape fluctuations self-organizes into a critical state, which creates a fine balance between static actin filaments and free monomers. In a comparison against a non-critical model, we show that this state facilitates spine enlargement, which happens after LTP induction. Thus, ongoing spine shape fluctuations might be necessary to react quickly to plasticity events.
Collapse
|
37
|
Shen W, Jin L, Zhu A, Lin Y, Pan G, Zhou S, Cheng J, Zhang J, Tu F, Liu C, Xie Q, Chen X. Treadmill exercise enhances synaptic plasticity in the ischemic penumbra of MCAO mice by inducing the expression of Camk2a via CYFIP1 upregulation. Life Sci 2021; 270:119033. [PMID: 33497737 DOI: 10.1016/j.lfs.2021.119033] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 12/29/2020] [Accepted: 01/01/2021] [Indexed: 10/22/2022]
Abstract
AIMS Physical exercise is beneficial to the recovery of patients with ischemic stroke. However, the underlying mechanism by which exercise promotes dendritic remodeling and synaptic plasticity is still obscure. This study explored the mechanism by which treadmill exercise enhances synaptic plasticity and dendritic remodeling in the ischemic penumbra. MAIN METHODS A middle cerebral artery occlusion (MCAO) model was generated in C57BL/6 mice, and lentivirus-mediated cytoplasmic FMRP-associated protein 1 (CYFIP1) shRNA expression was utilized to confirm the role of CYFIP1 in the exercise-induced increase in synaptic plasticity and dendritic remodeling. Neurological deficits were measured using the Zea Longa scale. Hematoxylin-eosin (H&E) staining and Nissl staining were performed to assess cerebral ischemic injury. Golgi-Cox staining was used to observe changes in dendritic remodeling and synaptic plasticity. Transmission electron microscopy (TEM) was performed to observe the synaptic ultrastructure. Molecular mechanisms were explored using immunofluorescence staining and western blotting. KEY FINDINGS Treadmill training enhanced synaptic plasticity in the penumbra. Additionally, we observed significant increases in the expression of CYFIP1 and calcium/calmodulin-dependent kinase 2a (Camk2a); enhanced neurological recovery and a decreased infarct volume. However, the injection of a lentivirus containing CYFIP1 shRNA into the lateral ventricle exerted negative effects on synaptic plasticity. Moreover, the exercise-induced neuroprotective effects were abolished by lentivirus-mediated CYFIP1 shRNA expression, consistent with the downregulation of Camk2a expression and the deterioration of neurological function. SIGNIFICANCE Treadmill training enhances synaptic plasticity and dendritic remodeling in the ischemic penumbra by inducing the expression of Camk2a via upregulation of CYFIP1.
Collapse
Affiliation(s)
- Weimin Shen
- Physical Medicine and Rehabilitation Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, No. 109, Xueyuanxi Road, Wenzhou, Zhejiang, China
| | - Lingqin Jin
- Physical Medicine and Rehabilitation Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, No. 109, Xueyuanxi Road, Wenzhou, Zhejiang, China
| | - Anqi Zhu
- Physical Medicine and Rehabilitation Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, No. 109, Xueyuanxi Road, Wenzhou, Zhejiang, China
| | - Yao Lin
- Physical Medicine and Rehabilitation Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, No. 109, Xueyuanxi Road, Wenzhou, Zhejiang, China
| | - Guoyuan Pan
- Physical Medicine and Rehabilitation Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, No. 109, Xueyuanxi Road, Wenzhou, Zhejiang, China
| | - Shanshan Zhou
- Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jingyan Cheng
- The Second Hospital Affiliated to Anhui University of Chinese Medicine, No.300, Shouchun Road, Hefei, Anhui, China
| | - Jieqiong Zhang
- Physical Medicine and Rehabilitation Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, No. 109, Xueyuanxi Road, Wenzhou, Zhejiang, China
| | - Fengxia Tu
- Physical Medicine and Rehabilitation Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, No. 109, Xueyuanxi Road, Wenzhou, Zhejiang, China
| | - Chan Liu
- Physical Medicine and Rehabilitation Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, No. 109, Xueyuanxi Road, Wenzhou, Zhejiang, China
| | - Qingfeng Xie
- Physical Medicine and Rehabilitation Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, No. 109, Xueyuanxi Road, Wenzhou, Zhejiang, China.
| | - Xiang Chen
- Physical Medicine and Rehabilitation Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, No. 109, Xueyuanxi Road, Wenzhou, Zhejiang, China.
| |
Collapse
|
38
|
Horn D, Fernández-Núñez E, Gomez-Carmona R, Rivera-Barahona A, Nevado J, Schwartzmann S, Ehmke N, Lapunzina P, Otaify GA, Temtamy S, Aglan M, Boschann F, Ruiz-Perez VL. Biallelic truncating variants in MAPKAPK5 cause a new developmental disorder involving neurological, cardiac, and facial anomalies combined with synpolydactyly. Genet Med 2021; 23:679-688. [PMID: 33442026 DOI: 10.1038/s41436-020-01052-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 11/19/2020] [Accepted: 11/20/2020] [Indexed: 11/09/2022] Open
Abstract
PURPOSE This study aimed to identify the genetic cause of a new multiple congenital anomalies syndrome observed in three individuals from two unrelated families. METHODS Clinical assessment was conducted prenatally and at different postnatal stages. Genetic studies included exome sequencing (ES) combined with single-nucleotide polymorphism (SNP) array based homozygosity mapping and trio ES. Dermal fibroblasts were used for functional assays. RESULTS A clinically recognizable syndrome characterized by severe developmental delay, variable brain anomalies, congenital heart defects, dysmorphic facial features, and a distinctive type of synpolydactyly with an additional hypoplastic digit between the fourth and fifth digits of hands and/or feet was identified. Additional features included eye abnormalities, hearing impairment, and electroencephalogram anomalies. ES detected different homozygous truncating variants in MAPKAPK5 in both families. Patient-derived cells showed no expression of MAPKAPK5 protein isoforms and reduced levels of the MAPKAPK5-interacting protein ERK3. F-actin recovery after latrunculin B treatment was found to be less efficient in patient-derived fibroblasts than in control cells, supporting a role of MAPKAPK5 in F-actin polymerization. CONCLUSION Our data indicate that loss-of-function variants in MAPKAPK5 result in a severe developmental disorder and reveal a major role of this gene in human brain, heart, and limb development.
Collapse
Affiliation(s)
- Denise Horn
- Institute of Medical Genetics and Human Genetics, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.
| | - Elisa Fernández-Núñez
- Instituto de Investigaciones Biomédicas "Alberto Sols", Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Ricardo Gomez-Carmona
- Instituto de Investigaciones Biomédicas "Alberto Sols", Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Ana Rivera-Barahona
- Instituto de Investigaciones Biomédicas "Alberto Sols", Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid (UAM), Madrid, Spain.,CIBER de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Julian Nevado
- CIBER de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.,Instituto de Genética Médica y Molecular (INGEMM)-IdiPAZ, Hospital Universitario La Paz, Universidad Autónoma, Madrid, Spain.,ITHACA, European Reference Network on Rare Congenital Malformations and Rare Intellectual Disability, Paris, France
| | - Sarina Schwartzmann
- Institute of Medical Genetics and Human Genetics, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Nadja Ehmke
- Institute of Medical Genetics and Human Genetics, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Pablo Lapunzina
- CIBER de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.,Instituto de Genética Médica y Molecular (INGEMM)-IdiPAZ, Hospital Universitario La Paz, Universidad Autónoma, Madrid, Spain.,ITHACA, European Reference Network on Rare Congenital Malformations and Rare Intellectual Disability, Paris, France
| | - Ghada A Otaify
- Department of Clinical Genetics, Division of Human Genetics and Genome Research, Center of Excellence for Human Genetics, National Research Centre, Cairo, Egypt
| | - Samia Temtamy
- Department of Clinical Genetics, Division of Human Genetics and Genome Research, Center of Excellence for Human Genetics, National Research Centre, Cairo, Egypt
| | - Mona Aglan
- Department of Clinical Genetics, Division of Human Genetics and Genome Research, Center of Excellence for Human Genetics, National Research Centre, Cairo, Egypt
| | - Felix Boschann
- Institute of Medical Genetics and Human Genetics, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Victor L Ruiz-Perez
- Instituto de Investigaciones Biomédicas "Alberto Sols", Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid (UAM), Madrid, Spain. .,CIBER de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III (ISCIII), Madrid, Spain. .,Instituto de Genética Médica y Molecular (INGEMM)-IdiPAZ, Hospital Universitario La Paz, Universidad Autónoma, Madrid, Spain. .,ITHACA, European Reference Network on Rare Congenital Malformations and Rare Intellectual Disability, Paris, France.
| |
Collapse
|
39
|
Ji B, Skup M. Roles of palmitoylation in structural long-term synaptic plasticity. Mol Brain 2021; 14:8. [PMID: 33430908 PMCID: PMC7802216 DOI: 10.1186/s13041-020-00717-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 12/15/2020] [Indexed: 11/30/2022] Open
Abstract
Long-term potentiation (LTP) and long-term depression (LTD) are important cellular mechanisms underlying learning and memory processes. N-Methyl-d-aspartate receptor (NMDAR)-dependent LTP and LTD play especially crucial roles in these functions, and their expression depends on changes in the number and single channel conductance of the major ionotropic glutamate receptor α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) located on the postsynaptic membrane. Structural changes in dendritic spines comprise the morphological platform and support for molecular changes in the execution of synaptic plasticity and memory storage. At the molecular level, spine morphology is directly determined by actin cytoskeleton organization within the spine and indirectly stabilized and consolidated by scaffold proteins at the spine head. Palmitoylation, as a uniquely reversible lipid modification with the ability to regulate protein membrane localization and trafficking, plays significant roles in the structural and functional regulation of LTP and LTD. Altered structural plasticity of dendritic spines is also considered a hallmark of neurodevelopmental disorders, while genetic evidence strongly links abnormal brain function to impaired palmitoylation. Numerous studies have indicated that palmitoylation contributes to morphological spine modifications. In this review, we have gathered data showing that the regulatory proteins that modulate the actin network and scaffold proteins related to AMPAR-mediated neurotransmission also undergo palmitoylation and play roles in modifying spine architecture during structural plasticity.
Collapse
Affiliation(s)
- Benjun Ji
- Nencki Institute of Experimental Biology, 02-093, Warsaw, Poland.
| | - Małgorzata Skup
- Nencki Institute of Experimental Biology, 02-093, Warsaw, Poland.
| |
Collapse
|
40
|
Leterrier C. A Pictorial History of the Neuronal Cytoskeleton. J Neurosci 2021; 41:11-27. [PMID: 33408133 PMCID: PMC7786211 DOI: 10.1523/jneurosci.2872-20.2020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 11/24/2020] [Accepted: 11/24/2020] [Indexed: 11/21/2022] Open
Affiliation(s)
- Christophe Leterrier
- Aix Marseille Université, Centre National de la Recherche Scientifique, INP Unité Mixte de Recherche 7051, NeuroCyto, Marseille 13005, France
| |
Collapse
|
41
|
Briševac D, Scholz R, Du D, Elagabani MN, Köhr G, Kornau HC. The small GTPase Arf6 is dysregulated in a mouse model for fragile X syndrome. J Neurochem 2020; 157:666-683. [PMID: 33125726 DOI: 10.1111/jnc.15230] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 10/09/2020] [Accepted: 10/27/2020] [Indexed: 11/29/2022]
Abstract
Fragile X syndrome (FXS), the most common inherited cause of intellectual disability, results from silencing of the fragile X mental retardation gene 1 (FMR1). The analyses of FXS patients' brain autopsies revealed an increased density of immature dendritic spines in cortical areas. We hypothesize that the small GTPase Arf6, an actin regulator critical for the development of glutamatergic synapses and dendritic spines, is implicated in FXS. Here, we determined the fraction of active, GTP-bound Arf6 in cortical neuron cultures and synaptoneurosomes from Fmr1 knockout mice, measured actin polymerization in neurons expressing Arf6 mutants with variant GTP- or GDP-binding properties, and recorded hippocampal long-term depression induced by metabotropic glutamate receptors (mGluR-LTD) in acute brain slices. We detected a persistently elevated Arf6 activity, a loss of Arf6 sensitivity to synaptic stimulation and an increased Arf6-dependent dendritic actin polymerization in mature Fmr1 knockout neurons. Similar imbalances in Arf6-GTP levels and actin filament assembly were caused in wild-type neurons by RNAi-mediated depletion of the postsynaptic Arf6 guanylate exchange factors IQSEC1 (BRAG2) or IQSEC2 (BRAG1). Targeted deletion of Iqsec1 in hippocampal neurons of 3-week-old mice interfered with mGluR-LTD in wild-type, but not in Fmr1 knockout mice. Collectively, these data suggest an aberrant Arf6 regulation in Fmr1 knockout neurons with consequences for the actin cytoskeleton, spine morphology, and synaptic plasticity. Moreover, FXS and syndromes caused by genetic variants in IQSEC1 and IQSEC2 share intellectual disabilities and developmental delay as main symptoms. Therefore, dysregulation of Arf6 may contribute to the cognitive impairment in FXS.
Collapse
Affiliation(s)
- Dušica Briševac
- Neuroscience Research Center (NWFZ), Charité - Universitätsmedizin Berlin, Berlin, Germany.,Department of Biology, Chemistry, and Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Ralf Scholz
- Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Dan Du
- Central Institute of Mental Health, Mannheim Center for Translational Neuroscience, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | | | - Georg Köhr
- Central Institute of Mental Health, Mannheim Center for Translational Neuroscience, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.,Department of Neurophysiology, Mannheim Center for Translational Neuroscience, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Hans-Christian Kornau
- Neuroscience Research Center (NWFZ), Charité - Universitätsmedizin Berlin, Berlin, Germany.,Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.,German Center for Neurodegenerative Diseases (DZNE) Berlin, Berlin, Germany
| |
Collapse
|
42
|
Vázquez-Hernández N, Martínez-Torres NI, González-Burgos I. Plastic changes to dendritic spines in the cerebellar and prefrontal cortices underlie the decline in motor coordination and working memory during successful aging. Behav Brain Res 2020; 400:113014. [PMID: 33309738 DOI: 10.1016/j.bbr.2020.113014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 11/03/2020] [Accepted: 11/09/2020] [Indexed: 10/22/2022]
Abstract
Old age is the last stage of life and by taking a multidimensional view of aging, Neuroscientists have been able to characterize pathological or successful aging. Psychomotor and cognitive performance are recognized as two major domains of successful aging, with a loss of motor coordination and working memory deficits two of the most characteristic features of elderly people. Dendritic spines in both the cerebellar and prefrontal cortices diminish in aging, yet the plastic changes in dendritic spines have not been related to behavioral performance neither the changes in the cerebellar or prefrontal cortices. As such, motor coordination and visuospatial working memory (vsWM) was evaluated here in aged, 22-month-old rats, calculating the density of spines and the proportion of the different types of spines. These animals performed erratically and slowly in a motor coordination-related paradigm, and the vsWM was resolved deficiently. Spine density was reduced in aged animals, and the proportional density of each of the spine types studied diminished in both the brain regions studied. The loss of dendritic spines and particularly, the changes in the proportional density of the different spine types could underlie, at least in part, the behavioral deficits observed during aging. To our knowledge, this is the first study of the plastic changes in different dendritic spine types that might underlie the behavioral alterations in motor and cognitive abilities associated with aging. Further neurochemical and molecular studies will help better understand the functional significance of the plastic changes to dendritic spines in both successful and pathological aging.
Collapse
Affiliation(s)
- N Vázquez-Hernández
- División de Neurociencias, Centro de Investigación Biomédica de Occidente, IMSS, Guadalajara, Jal, Mexico
| | - N I Martínez-Torres
- División de Neurociencias, Centro de Investigación Biomédica de Occidente, IMSS, Guadalajara, Jal, Mexico; Centro Universitario del Norte, Universidad de Guadalajara, Colotlán, Jal, Mexico
| | - I González-Burgos
- División de Neurociencias, Centro de Investigación Biomédica de Occidente, IMSS, Guadalajara, Jal, Mexico.
| |
Collapse
|
43
|
Leonard CE, Baydyuk M, Stepler MA, Burton DA, Donoghue MJ. EphA7 isoforms differentially regulate cortical dendrite development. PLoS One 2020; 15:e0231561. [PMID: 33275600 PMCID: PMC7717530 DOI: 10.1371/journal.pone.0231561] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 11/22/2020] [Indexed: 02/06/2023] Open
Abstract
The shape of a neuron facilitates its functionality within neural circuits. Dendrites integrate incoming signals from axons, receiving excitatory input onto small protrusions called dendritic spines. Therefore, understanding dendritic growth and development is fundamental for discerning neural function. We previously demonstrated that EphA7 receptor signaling during cortical development impacts dendrites in two ways: EphA7 restricts dendritic growth early and promotes dendritic spine formation later. Here, the molecular basis for this shift in EphA7 function is defined. Expression analyses reveal that EphA7 full-length (EphA7-FL) and truncated (EphA7-T1; lacking kinase domain) isoforms are dynamically expressed in the developing cortex. Peak expression of EphA7-FL overlaps with dendritic elaboration around birth, while highest expression of EphA7-T1 coincides with dendritic spine formation in early postnatal life. Overexpression studies in cultured neurons demonstrate that EphA7-FL inhibits both dendritic growth and spine formation, while EphA7-T1 increases spine density. Furthermore, signaling downstream of EphA7 shifts during development, such that in vivo inhibition of mTOR by rapamycin in EphA7-mutant neurons ameliorates dendritic branching, but not dendritic spine phenotypes. Finally, direct interaction between EphA7-FL and EphA7-T1 is demonstrated in cultured cells, which results in reduction of EphA7-FL phosphorylation. In cortex, both isoforms are colocalized to synaptic fractions and both transcripts are expressed together within individual neurons, supporting a model where EphA7-T1 modulates EphA7-FL repulsive signaling during development. Thus, the divergent functions of EphA7 during cortical dendrite development are explained by the presence of two variants of the receptor.
Collapse
Affiliation(s)
- Carrie E. Leonard
- Department of Biology, Georgetown University, Washington, DC, United States of America
- Interdisciplinary Program in Neuroscience, Georgetown University, Washington, DC, United States of America
| | - Maryna Baydyuk
- Department of Biology, Georgetown University, Washington, DC, United States of America
| | - Marissa A. Stepler
- Department of Biology, Georgetown University, Washington, DC, United States of America
| | - Denver A. Burton
- Department of Biology, Georgetown University, Washington, DC, United States of America
| | - Maria J. Donoghue
- Department of Biology, Georgetown University, Washington, DC, United States of America
- Interdisciplinary Program in Neuroscience, Georgetown University, Washington, DC, United States of America
| |
Collapse
|
44
|
Feuge J, Scharkowski F, Michaelsen-Preusse K, Korte M. FMRP Modulates Activity-Dependent Spine Plasticity by Binding Cofilin1 mRNA and Regulating Localization and Local Translation. Cereb Cortex 2020; 29:5204-5216. [PMID: 30953439 DOI: 10.1093/cercor/bhz059] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 02/14/2019] [Accepted: 02/27/2019] [Indexed: 11/13/2022] Open
Abstract
Multiple variants of intellectual disability, e.g., the Fragile X Syndrome are associated with alterations in dendritic spine morphology, thereby pointing to dysregulated actin dynamics during development and processes of synaptic plasticity. Surprisingly, although the necessity of spine actin remodeling was demonstrated repeatedly, the importance and precise role of actin regulators is often undervalued. Here, we provide evidence that structural and functional plasticity are severely impaired after NMDAR-dependent LTP in the hippocampus of Fmr1 KO mice. We can link these defects to an aberrant activity-dependent regulation of Cofilin 1 (cof1) as activity-dependent modulations of local cof1 mRNA availability, local cof1 translation as well as total cof1 expression are impaired in the absence of FMRP. Finally, we can rescue activity-dependent structural plasticity in KO neurons by mimicking the regulation of cof1 observed in WT cells, thereby illustrating the potential of actin modulators to provide novel treatment strategies for the Fragile X Syndrome.
Collapse
Affiliation(s)
- Jonas Feuge
- Division of Cellular Neurobiology, Zoological Institute, TU Braunschweig, Germany
| | | | | | - Martin Korte
- Division of Cellular Neurobiology, Zoological Institute, TU Braunschweig, Germany.,Helmholtz Center for Infection Research, Research group Neuroinflammation and Neurodegeneration, Braunschweig, Germany
| |
Collapse
|
45
|
Runge K, Cardoso C, de Chevigny A. Dendritic Spine Plasticity: Function and Mechanisms. Front Synaptic Neurosci 2020. [DOI: 10.3389/fnsyn.2020.00036
expr 823669561 + 872784217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
|
46
|
Runge K, Cardoso C, de Chevigny A. Dendritic Spine Plasticity: Function and Mechanisms. Front Synaptic Neurosci 2020; 12:36. [PMID: 32982715 PMCID: PMC7484486 DOI: 10.3389/fnsyn.2020.00036] [Citation(s) in RCA: 131] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 07/28/2020] [Indexed: 12/12/2022] Open
Abstract
Dendritic spines are small protrusions studding neuronal dendrites, first described in 1888 by Ramón y Cajal using his famous Golgi stainings. Around 50 years later the advance of electron microscopy (EM) confirmed Cajal's intuition that spines constitute the postsynaptic site of most excitatory synapses in the mammalian brain. The finding that spine density decreases between young and adult ages in fixed tissues suggested that spines are dynamic. It is only a decade ago that two-photon microscopy (TPM) has unambiguously proven the dynamic nature of spines, through the repeated imaging of single spines in live animals. Spine dynamics comprise formation, disappearance, and stabilization of spines and are modulated by neuronal activity and developmental age. Here, we review several emerging concepts in the field that start to answer the following key questions: What are the external signals triggering spine dynamics and the molecular mechanisms involved? What is, in return, the role of spine dynamics in circuit-rewiring, learning, and neuropsychiatric disorders?
Collapse
Affiliation(s)
- Karen Runge
- Institut de Neurobiologie de la Méditerranée (INMED) INSERM U1249, Aix-Marseille University, Marseille, France
| | - Carlos Cardoso
- Institut de Neurobiologie de la Méditerranée (INMED) INSERM U1249, Aix-Marseille University, Marseille, France
| | - Antoine de Chevigny
- Institut de Neurobiologie de la Méditerranée (INMED) INSERM U1249, Aix-Marseille University, Marseille, France
| |
Collapse
|
47
|
Natarajaseenivasan K, Shanmughapriya S, Velusamy P, Sayre M, Garcia A, Gomez NM, Langford D. Inflammation-induced PINCH expression leads to actin depolymerization and mitochondrial mislocalization in neurons. Transl Neurodegener 2020; 9:32. [PMID: 32746944 PMCID: PMC7397656 DOI: 10.1186/s40035-020-00211-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 07/21/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Diseases and disorders with a chronic neuroinflammatory component are often linked with changes in brain metabolism. Among neurodegenerative disorders, people living with human immunodeficiency virus (HIV) and Alzheimer's disease (AD) are particularly vulnerable to metabolic disturbances, but the mechanistic connections of inflammation, neurodegeneration and bioenergetic deficits in the central nervous system (CNS) are poorly defined. The particularly interesting new cysteine histidine-rich-protein (PINCH) is nearly undetectable in healthy mature neurons, but is robustly expressed in tauopathy-associated neurodegenerative diseases including HIV infection and AD. Although robust PINCH expression has been reported in neurons in the brains of patients with HIV and AD, the molecular mechanisms and cellular consequences of increased PINCH expression in CNS disease remain largely unknown. METHODS We investigated the regulatory mechanisms responsible for PINCH protein-mediated changes in bioenergetics, mitochondrial subcellular localization and bioenergetic deficits in neurons exposed to physiological levels of TNFα or the HIV protein Tat. Changes in the PINCH-ILK-Parvin (PIP) complex association with cofilin and TESK1 were assessed to identify factors responsible for actin depolymerization and mitochondrial mislocalization. Lentiviral and pharmacological inhibition experiments were conducted to confirm PINCH specificity and to reinstate proper protein-protein complex communication. RESULTS We identified MEF2A as the PINCH transcription factor in neuroinflammation and determined the biological consequences of increased PINCH in neurons. TNFα-mediated activation of MEF2A via increased cellular calcium induced PINCH, leading to disruption of the PIP ternary complex, cofilin activation by TESK1 inactivation, and actin depolymerization. The disruption of actin led to perinuclear mislocalization of mitochondria by destabilizing the kinesin-dependent mitochondrial transport machinery, resulting in impaired neuronal metabolism. Blocking TNFα-induced PINCH expression preserved mitochondrial localization and maintained metabolic functioning. CONCLUSIONS This study reported for the first time the mechanistic and biological consequences of PINCH expression in CNS neurons in diseases with a chronic neuroinflammation component. Our findings point to the maintenance of PINCH at normal physiological levels as a potential new therapeutic target for neurodegenerative diseases with impaired metabolisms.
Collapse
Affiliation(s)
- Kalimuthusamy Natarajaseenivasan
- Department of Neurosciences and Center for Neurovirology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140 USA
- Department of Microbiology, Bharathidasan University, Tiruchirappalli, 620024 India
| | - Santhanam Shanmughapriya
- Heart and Vascular Institute, Department of Medicine, Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA 17033 USA
| | - Prema Velusamy
- Heart and Vascular Institute, Department of Medicine, Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA 17033 USA
| | - Matthew Sayre
- Department of Neurosciences and Center for Neurovirology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140 USA
| | - Alvaro Garcia
- Department of Neurosciences and Center for Neurovirology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140 USA
| | - Nestor Mas Gomez
- Department of Neurosciences and Center for Neurovirology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140 USA
| | - Dianne Langford
- Department of Neurosciences and Center for Neurovirology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140 USA
| |
Collapse
|
48
|
Klimas A, Zhao Y. Expansion Microscopy: Toward Nanoscale Imaging of a Diverse Range of Biomolecules. ACS NANO 2020; 14:7689-7695. [PMID: 32628828 PMCID: PMC7456618 DOI: 10.1021/acsnano.0c04374] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Expansion microscopy (ExM) has become a powerful imaging tool for visualizing the nanoscale organization of protein and nucleic acid targets in cells and tissues using only a conventional microscope. Until recently, current ExM approaches have had limited applicability to imaging other biomolecules, such as lipids and small molecules. With the new TRITON probes reported by Wen et al. in this issue of ACS Nano, ExM can now be used to perform nanoscale imaging of the cytoskeleton and lipid membranes. In this Perspective, we offer a brief overview of recent developments in ExM, with a focus on biomolecule anchoring and labeling strategies that target a wide range of biomolecules to the water-swellable polymer formed in situ, a key step that ensures biomolecules or labels of interest are separated in space and can be resolved on a conventional microscope. In addition to these new advancements, we discuss challenges and future directions in this exciting field.
Collapse
Affiliation(s)
- Aleksandra Klimas
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Yongxin Zhao
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA, USA
| |
Collapse
|
49
|
Wen G, Vanheusden M, Acke A, Valli D, Neely RK, Leen V, Hofkens J. Evaluation of Direct Grafting Strategies via Trivalent Anchoring for Enabling Lipid Membrane and Cytoskeleton Staining in Expansion Microscopy. ACS NANO 2020; 14:7860-7867. [PMID: 32176475 DOI: 10.1021/acsnano.9b09259] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
Super-resolution fluorescence microscopy is a key tool in the elucidation of biological fine structures, providing insights into the distribution and interactions of biomolecular complexes down to the nanometer scale. Expansion microscopy is a recently developed approach for achieving nanoscale resolution on a conventional microscope. Here, biological samples are embedded in an isotropically swollen hydrogel. This physical expansion of the sample allows imaging with resolutions down to the tens-of-nanometers. However, because of the requirement that fluorescent labels are covalently bound to the hydrogel, standard, small-molecule targeting of fluorophores has proven incompatible with expansion microscopy. Here, we show a chemical linking approach that enables direct, covalent grafting of a targeting molecule and fluorophore to the hydrogel in expansion microscopy. We show application of this series of molecules in the antibody-free targeting of the cell cytoskeleton and in an example of lipid membrane staining for expansion microscopy. Furthermore, using this trivalent linker strategy, we demonstrate the benefit of introducing fluorescent labels post-expansion by visualizing an immunostaining through fluorescent oligonucleotide hybridization after expanding the polymer. Our probes allow different labeling approaches that are compatible with expansion microscopy.
Collapse
Affiliation(s)
- Gang Wen
- Department of Chemistry, KU Leuven, Leuven, 3001, Belgium
| | | | - Aline Acke
- Department of Chemistry, KU Leuven, Leuven, 3001, Belgium
| | - Donato Valli
- Department of Chemistry, KU Leuven, Leuven, 3001, Belgium
| | - Robert K Neely
- School of Chemistry, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Volker Leen
- Department of Chemistry, KU Leuven, Leuven, 3001, Belgium
| | - Johan Hofkens
- Department of Chemistry, KU Leuven, Leuven, 3001, Belgium
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| |
Collapse
|
50
|
Fox ME, Figueiredo A, Menken MS, Lobo MK. Dendritic spine density is increased on nucleus accumbens D2 neurons after chronic social defeat. Sci Rep 2020; 10:12393. [PMID: 32709968 PMCID: PMC7381630 DOI: 10.1038/s41598-020-69339-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 07/09/2020] [Indexed: 02/07/2023] Open
Abstract
Stress alters the structure and function of brain reward circuitry and is an important risk factor for developing depression. In the nucleus accumbens (NAc), structural and physiological plasticity of medium spiny neurons (MSNs) have been linked to increased stress-related and depression-like behaviors. NAc MSNs have opposing roles in driving stress-related behaviors that is dependent on their dopamine receptor expression. After chronic social defeat stress, NAc MSNs exhibit increased dendritic spine density. However, it remains unclear if the dendritic spine plasticity is MSN subtype specific. Here we use viral labeling to characterize dendritic spine morphology specifically in dopamine D2 receptor expressing MSNs (D2-MSNs). After chronic social defeat, D2-MSNs exhibit increased spine density that is correlated with enhanced social avoidance behavior. Together, our data indicate dendritic spine plasticity is MSN subtype specific, improving our understanding of structural plasticity after chronic stress.
Collapse
Affiliation(s)
- Megan E Fox
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, 20 Penn St, HSFII Building, Rm 265, Baltimore, MD, 21201, USA
| | - Antonio Figueiredo
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, 20 Penn St, HSFII Building, Rm 265, Baltimore, MD, 21201, USA
| | - Miriam S Menken
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, 20 Penn St, HSFII Building, Rm 265, Baltimore, MD, 21201, USA
| | - Mary Kay Lobo
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, 20 Penn St, HSFII Building, Rm 265, Baltimore, MD, 21201, USA.
| |
Collapse
|