1
|
Kaizuka T, Takumi T. Alteration of synaptic protein composition during developmental synapse maturation. Eur J Neurosci 2024; 59:2894-2914. [PMID: 38571321 DOI: 10.1111/ejn.16304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 01/02/2024] [Accepted: 02/07/2024] [Indexed: 04/05/2024]
Abstract
The postsynaptic density (PSD) is a collection of specialized proteins assembled beneath the postsynaptic membrane of dendritic spines. The PSD proteome comprises ~1000 proteins, including neurotransmitter receptors, scaffolding proteins and signalling enzymes. Many of these proteins have essential roles in synaptic function and plasticity. During brain development, changes are observed in synapse density and in the stability and shape of spines, reflecting the underlying molecular maturation of synapses. Synaptic protein composition changes in terms of protein abundance and the assembly of protein complexes, supercomplexes and the physical organization of the PSD. Here, we summarize the developmental alterations of postsynaptic protein composition during synapse maturation. We describe major PSD proteins involved in postsynaptic signalling that regulates synaptic plasticity and discuss the effect of altered expression of these proteins during development. We consider the abnormality of synaptic profiles and synaptic protein composition in the brain in neurodevelopmental disorders such as autism spectrum disorders. We also explain differences in synapse development between rodents and primates in terms of synaptic profiles and protein composition. Finally, we introduce recent findings related to synaptic diversity and nanoarchitecture and discuss their impact on future research. Synaptic protein composition can be considered a major determinant and marker of synapse maturation in normality and disease.
Collapse
Affiliation(s)
- Takeshi Kaizuka
- Department of Physiology and Cell Biology, Kobe University School of Medicine, Kobe, Japan
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Toru Takumi
- Department of Physiology and Cell Biology, Kobe University School of Medicine, Kobe, Japan
- RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| |
Collapse
|
2
|
McLaughlin T, Wang G, Medina A, Perkins J, Nihlawi R, Seyfried D, Hu Z, Wang JJ, Zhang SX. Essential Role of XBP1 in Maintaining Photoreceptor Synaptic Integrity in Early Diabetic Retinopathy. Invest Ophthalmol Vis Sci 2023; 64:40. [PMID: 38015176 PMCID: PMC10691399 DOI: 10.1167/iovs.64.14.40] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 11/04/2023] [Indexed: 11/29/2023] Open
Abstract
Purpose Diabetic retinopathy (DR) is a leading cause of blindness in working-age adults characterized by retinal dysfunction and neurovascular degeneration. We previously reported that deletion of X-box binding protein 1 (XBP1) leads to accelerated retinal neurodegeneration in diabetes; however, the mechanisms remain elusive. The goal of this study is to determine the role of XBP1 in the regulation of photoreceptor synaptic integrity in early DR. Methods Diabetes was induced by streptozotocin in retina-specific XBP1 conditional knockout (cKO) or wild-type (WT) mice to generate diabetic cKO (cKO/DM) or WT/DM mice for comparison with nondiabetic cKO (cKO/NDM) and WT/NDM mice. Retinal morphology, structure, and function were assessed by immunohistochemistry, optical coherence tomography, and electroretinogram (ERG) after 3 months of diabetes. The synapses between photoreceptors and bipolar cells were examined by confocal microscopy, and synaptic integrity was quantified using the QUANTOS algorithm. Results We found a thinning of the outer nuclear layer and a decline in the b-wave amplitude in dark- and light-adapted ERG in cKO/DM mice compared to all other groups. In line with these changes, cKO mice showed increased loss of synaptic integrity compared to WT mice, regardless of diabetes status. In searching for candidate molecules responsible for the loss of photoreceptor synaptic integrity in diabetic and XBP1-deficient retinas, we found decreased mRNA and protein levels of DLG4/PSD-95 in cKO/DM retina compared to WT/DM. Conclusions These findings suggest that XBP1 is a crucial regulator in maintaining synaptic integrity and retinal function, possibly through regulation of synaptic scaffold proteins.
Collapse
Affiliation(s)
- Todd McLaughlin
- Department of Ophthalmology and Ross Eye Institute, University at Buffalo, State University of New York, Buffalo, New York, United States
| | - Grant Wang
- Department of Ophthalmology and Ross Eye Institute, University at Buffalo, State University of New York, Buffalo, New York, United States
| | - Andy Medina
- Department of Ophthalmology and Ross Eye Institute, University at Buffalo, State University of New York, Buffalo, New York, United States
| | - Jacob Perkins
- Department of Ophthalmology and Ross Eye Institute, University at Buffalo, State University of New York, Buffalo, New York, United States
| | - Rhudwan Nihlawi
- Department of Ophthalmology and Ross Eye Institute, University at Buffalo, State University of New York, Buffalo, New York, United States
| | - Don Seyfried
- Department of Ophthalmology and Ross Eye Institute, University at Buffalo, State University of New York, Buffalo, New York, United States
| | - Zihua Hu
- Department of Ophthalmology and Ross Eye Institute, University at Buffalo, State University of New York, Buffalo, New York, United States
- Center for Computational Research, New York State Center of Excellence in Bioinformatics and Life Sciences, State University of New York, Buffalo, New York, United States
| | - Joshua J. Wang
- Department of Ophthalmology and Ross Eye Institute, University at Buffalo, State University of New York, Buffalo, New York, United States
- Department of Biochemistry, State University of New York, Buffalo, New York, United States
| | - Sarah X. Zhang
- Department of Ophthalmology and Ross Eye Institute, University at Buffalo, State University of New York, Buffalo, New York, United States
- Department of Biochemistry, State University of New York, Buffalo, New York, United States
| |
Collapse
|
3
|
Calabrese B, Jones SL, Shiraishi-Yamaguchi Y, Lingelbach M, Manor U, Svitkina TM, Higgs HN, Shih AY, Halpain S. INF2-mediated actin filament reorganization confers intrinsic resilience to neuronal ischemic injury. Nat Commun 2022; 13:6037. [PMID: 36229429 PMCID: PMC9558009 DOI: 10.1038/s41467-022-33268-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Accepted: 09/09/2022] [Indexed: 12/24/2022] Open
Abstract
During early ischemic brain injury, glutamate receptor hyperactivation mediates neuronal death via osmotic cell swelling. Here we show that ischemia and excess NMDA receptor activation cause actin to rapidly and extensively reorganize within the somatodendritic compartment. Normally, F-actin is concentrated within dendritic spines. However, <5 min after bath-applied NMDA, F-actin depolymerizes within spines and polymerizes into stable filaments within the dendrite shaft and soma. A similar actinification occurs after experimental ischemia in culture, and photothrombotic stroke in mouse. Following transient NMDA incubation, actinification spontaneously reverses. Na+, Cl-, water, and Ca2+ influx, and spine F-actin depolymerization are all necessary, but not individually sufficient, for actinification, but combined they induce activation of the F-actin polymerization factor inverted formin-2 (INF2). Silencing of INF2 renders neurons vulnerable to cell death and INF2 overexpression is protective. Ischemia-induced dendritic actin reorganization is therefore an intrinsic pro-survival response that protects neurons from death induced by cell edema.
Collapse
Affiliation(s)
- Barbara Calabrese
- Department of Neurobiology, School of Biological Sciences, University of California, San Diego, and Sanford Consortium for Regenerative Medicine, La Jolla, CA, 92093, USA
| | - Steven L Jones
- Department of Biology, University of Pennsylvania, Philadelphia, PA, 19104-4544, USA
| | | | - Michael Lingelbach
- Neurosciences Interdepartmental Program, Stanford University, Stanford, CA, 94305, USA
| | - Uri Manor
- The Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Tatyana M Svitkina
- Department of Biology, University of Pennsylvania, Philadelphia, PA, 19104-4544, USA
| | - Henry N Higgs
- Department of Biochemistry, Geisel School of Medicine, Hanover, NH, 03755, USA
| | - Andy Y Shih
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA, 98101, USA
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Shelley Halpain
- Department of Neurobiology, School of Biological Sciences, University of California, San Diego, and Sanford Consortium for Regenerative Medicine, La Jolla, CA, 92093, USA.
| |
Collapse
|
4
|
The Effects of Poria cocos on Rho Signaling-Induced Regulation of Mobility and F-Actin Aggregation in MK-801-Treated B35 and C6 Cells. Behav Neurol 2022; 2022:8225499. [PMID: 35864844 PMCID: PMC9296330 DOI: 10.1155/2022/8225499] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 06/28/2022] [Indexed: 01/05/2023] Open
Abstract
Methods B35 neuronal cells and C6 glial cells were incubated with MK-801 for 7 days followed by MK-801, MK801 in combination with water extracts of P. cocos (PRP for P. cocos cum Radix Pini or WP for White Poria) treatment for an additional 7 days. Analysis of cell mobility, F-actin aggregation, and Rho signaling modulation was performed to clarify the roles of PRP or WP in MK-801-treated B35 and C6 cells. Results MK-801 decreases B35 cell mobility, whereas the inhibited cell migration ability and F-actin aggregation in MK-801-treated B35 or C6 cells could be reversed by PRP or WP. The CDC42 expression in B35 or C6 cells would be reduced by MK-801 and restored by treating with PRP or WP. The RhoA expression was increased by MK-801 in both B35 and C6 cells but was differentially regulated by PRP or WP. In B35 cells, downregulation of PFN1, N-WASP, PAK1, and ARP2/3 induced by MK-801 can be reversely modulated by PRP or WP. PRP or WP reduced the increase in the p-MLC2 expression in B35 cells treated with MK-801. The reduction in ROCK1, PFN1, p-MLC2, and ARP2/3 expression in C6 cells induced by MK-801 was restored by PRP or WP. Reduced N-WASP and PAK1 expression was differentially regulated by PRP or WP in MK-801-treated C6 cells.
Collapse
|
5
|
Dutta P, Bharti P, Kumar J, Maiti S. Role of actin cytoskeleton in the organization and function of ionotropic glutamate receptors. Curr Res Struct Biol 2021; 3:277-289. [PMID: 34766008 PMCID: PMC8569634 DOI: 10.1016/j.crstbi.2021.10.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 09/04/2021] [Accepted: 10/09/2021] [Indexed: 12/22/2022] Open
Abstract
Neural networks with precise connection are compulsory for learning and memory. Various cellular events occur during the genesis of dendritic spines to their maturation, synapse formation, stabilization of the synapse, and proper signal transmission. The cortical actin cytoskeleton and its multiple regulatory proteins are crucial for the above cellular events. The different types of ionotropic glutamate receptors (iGluRs) present on the postsynaptic density (PSD) are also essential for learning and memory. Interaction of the iGluRs in association of their auxiliary proteins with actin cytoskeleton regulated by actin-binding proteins (ABPs) are required for precise long-term potentiation (LTP) and long-term depression (LTD). There has been a quest to understand the mechanistic detail of synapse function involving these receptors with dynamic actin cytoskeleton. A major, emerging area of investigation is the relationship between ABPs and iGluRs in synapse development. In this review we have summarized the current understanding of iGluRs functioning with respect to the actin cytoskeleton, scaffolding proteins, and their regulators. The AMPA, NMDA, Delta and Kainate receptors need the stable underlying actin cytoskeleton to anchor through synaptic proteins for precise synapse formation. The different types of ABPs present in neurons play a critical role in dynamizing/stabilizing the actin cytoskeleton needed for iGluRs function.
Collapse
Affiliation(s)
- Priyanka Dutta
- National Centre for Cell Science, Pune, Maharashtra, 411007, India
| | - Pratibha Bharti
- National Centre for Cell Science, Pune, Maharashtra, 411007, India
| | - Janesh Kumar
- National Centre for Cell Science, Pune, Maharashtra, 411007, India
| | - Sankar Maiti
- Indian Institute of Science Education and Research, Kolkata, 741246, India
| |
Collapse
|
6
|
Poria cocos Regulates Cell Migration and Actin Filament Aggregation in B35 and C6 Cells by Modulating the RhoA, CDC42, and Rho Signaling Pathways. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:6854860. [PMID: 34512781 PMCID: PMC8426088 DOI: 10.1155/2021/6854860] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 08/13/2021] [Accepted: 08/26/2021] [Indexed: 12/05/2022]
Abstract
Poria is used as a traditional Chinese herbal medicine with anti-inflammatory, anticancer, and mood-stabilizing properties. Poria contains triterpenoids and polysaccharides, which are reported to regulate the cytoplasmic free calcium associated with the N-methyl-D-aspartate receptor and affect the cell function of neonatal rat nerve cells and hippocampal neurons. Although the modulatory effects of Poria on neuronal function have been widely reported, the molecular mechanism of these effects is unclear. Cell migration ability and the reorganization of actin filaments are important biological functions during neuronal development, and they can be regulated mainly by the Rho signaling pathway. We found that the cell migration ability and actin condensation in B35 cells enhanced by P. cocos (a water solution of P. cocos cum Radix Pini (PRP) or White Poria (WP)) might be caused by increased RhoA and CDC42 activity and increased expression of downstream ROCK1, p-MLC2, N-WASP, and ARP2/3 in B35 cells. Similar modulations of cell migration ability, actin condensation, and Rho signaling pathway were also observed in the C6 glial cell line, except for the PRP-induced regulation of RhoA and CDC42 activities. Ketamine-induced inhibition of cell migration and actin condensation can be restored by P. cocos. In addition, we observed that the increased expression of RhoA and ROCK1 or the decreased expression of CDC42 and N-WASP caused by ketamine in B35 cells could also be restored by P. cocos. The results of this study suggest that the regulatory effects of P. cocos on cell migration and actin filament aggregation are closely related to the regulation of RhoA, CDC42, and Rho signaling pathways in both B35 and C6 cells. PRP and WP have the potential to restore neuronal cell Rho signaling abnormalities involved in some mental diseases.
Collapse
|
7
|
Murk K, Ornaghi M, Schiweck J. Profilin Isoforms in Health and Disease - All the Same but Different. Front Cell Dev Biol 2021; 9:681122. [PMID: 34458253 PMCID: PMC8387879 DOI: 10.3389/fcell.2021.681122] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 07/12/2021] [Indexed: 12/11/2022] Open
Abstract
Profilins are small actin binding proteins, which are structurally conserved throughout evolution. They are probably best known to promote and direct actin polymerization. However, they also participate in numerous cell biological processes beyond the roles typically ascribed to the actin cytoskeleton. Moreover, most complex organisms express several profilin isoforms. Their cellular functions are far from being understood, whereas a growing number of publications indicate that profilin isoforms are involved in the pathogenesis of various diseases. In this review, we will provide an overview of the profilin family and "typical" profilin properties including the control of actin dynamics. We will then discuss the profilin isoforms of higher animals in detail. In terms of cellular functions, we will focus on the role of Profilin 1 (PFN1) and Profilin 2a (PFN2a), which are co-expressed in the central nervous system. Finally, we will discuss recent findings that link PFN1 and PFN2a to neurological diseases, such as amyotrophic lateral sclerosis (ALS), Fragile X syndrome (FXS), Huntington's disease and spinal muscular atrophy (SMA).
Collapse
Affiliation(s)
- Kai Murk
- Institute of Biochemistry, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Marta Ornaghi
- Institute of Biochemistry, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Juliane Schiweck
- Institute of Biochemistry, Charité Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
8
|
Li ZZ, Han WJ, Sun ZC, Chen Y, Sun JY, Cai GH, Liu WN, Wang TZ, Xie YD, Mao HH, Wang F, Ma SB, Wang FD, Xie RG, Wu SX, Luo C. Extracellular matrix protein laminin β1 regulates pain sensitivity and anxiodepression-like behaviors in mice. J Clin Invest 2021; 131:e146323. [PMID: 34156983 DOI: 10.1172/jci146323] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 06/16/2021] [Indexed: 01/11/2023] Open
Abstract
Patients with neuropathic pain often experience comorbid psychiatric disorders. Cellular plasticity in the anterior cingulate cortex (ACC) is assumed to be a critical interface for pain perception and emotion. However, substantial efforts have thus far been focused on the intracellular mechanisms of plasticity rather than the extracellular alterations that might trigger and facilitate intracellular changes. Laminin, a key element of the extracellular matrix (ECM), consists of one α-, one β-, and one γ-chain and is implicated in several pathophysiological processes. Here, we showed in mice that laminin β1 (LAMB1) in the ACC was significantly downregulated upon peripheral neuropathy. Knockdown of LAMB1 in the ACC exacerbated pain sensitivity and induced anxiety and depression. Mechanistic analysis revealed that loss of LAMB1 caused actin dysregulation via interaction with integrin β1 and the subsequent Src-dependent RhoA/LIMK/cofilin pathway, leading to increased presynaptic transmitter release probability and abnormal postsynaptic spine remodeling, which in turn orchestrated the structural and functional plasticity of pyramidal neurons and eventually resulted in pain hypersensitivity and anxiodepression. This study sheds new light on the functional capability of ECM LAMB1 in modulating pain plasticity and identifies a mechanism that conveys extracellular alterations to intracellular plasticity. Moreover, we identified cingulate LAMB1/integrin β1 signaling as a promising therapeutic target for the treatment of neuropathic pain and associated anxiodepression.
Collapse
Affiliation(s)
- Zhen-Zhen Li
- Department of Neurobiology, School of Basic Medicine.,Department of Neurosurgery, Xijing Hospital, and
| | - Wen-Juan Han
- Department of Neurobiology, School of Basic Medicine
| | - Zhi-Chuan Sun
- Department of Neurobiology, School of Basic Medicine
| | - Yun Chen
- The Second Regiment, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Jun-Yi Sun
- The Second Regiment, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Guo-Hong Cai
- Department of Neurobiology, School of Basic Medicine
| | - Wan-Neng Liu
- Department of Neurobiology, School of Basic Medicine.,College of Life Sciences, Northwest University, Xi'an, China
| | - Tao-Zhi Wang
- Department of Neurobiology, School of Basic Medicine.,Department of Anesthesiology, The Second Hospital of Jilin University, Changchun, China
| | - Yang-Dan Xie
- The Second Regiment, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Hong-Hui Mao
- Department of Neurobiology, School of Basic Medicine
| | - Fei Wang
- Department of Neurobiology, School of Basic Medicine.,Medical Experiment Center, Shaanxi University of Chinese Medicine, China
| | - Sui-Bin Ma
- Department of Neurobiology, School of Basic Medicine
| | - Fu-Dong Wang
- Department of Neurobiology, School of Basic Medicine
| | - Rou-Gang Xie
- Department of Neurobiology, School of Basic Medicine
| | - Sheng-Xi Wu
- Department of Neurobiology, School of Basic Medicine
| | - Ceng Luo
- Department of Neurobiology, School of Basic Medicine
| |
Collapse
|
9
|
Vallejo D, Lindsay CB, González-Billault C, Inestrosa NC. Wnt5a modulates dendritic spine dynamics through the regulation of Cofilin via small Rho GTPase activity in hippocampal neurons. J Neurochem 2021; 158:673-693. [PMID: 34107066 DOI: 10.1111/jnc.15448] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 06/03/2021] [Accepted: 06/07/2021] [Indexed: 01/21/2023]
Abstract
Dendritic spines are small, actin-rich protrusions that act as the receiving sites of most excitatory inputs in the central nervous system. The remodeling of the synapse architecture is mediated by actin cytoskeleton dynamics, a process precisely regulated by the small Rho GTPase family. Wnt ligands exert their presynaptic and postsynaptic effects during formation and consolidation of the synaptic structure. Specifically, Wnt5a has been identified as an indispensable synaptogenic factor for the regulation and organization of the postsynaptic side; however, the molecular mechanisms through which Wnt5a induces morphological changes resulting from actin cytoskeleton dynamics within dendritic spines remain unclear. In this work, we employ primary rat hippocampal cultures and HT22 murine hippocampal neuronal cell models, molecular and pharmacological tools, and fluorescence microscopy (laser confocal and epifluorescence) to define the Wnt5a-induced molecular signaling involved in postsynaptic remodeling mediated via the regulation of the small Rho GTPase family. We report that Wnt5a differentially regulates the phosphorylation of Cofilin in neurons through both Ras-related C3 botulinum toxin substrate 1 and cell division cycle 42 depending on the subcellular compartment and the extracellular calcium levels. Additionally, we demonstrate that Wnt5a increases the density of dendritic spines and promotes their maturation via Ras-related C3 botulinum toxin substrate 1. Accordingly, we find that Wnt5a requires the combined activation of small Rho GTPases to increase the levels of filamentous actin, thus promoting the stability of actin filaments. Altogether, these results provide evidence for a new mechanism by which Wnt5a may target actin dynamics, thereby regulating the subsequent morphological changes in dendritic spine architecture.
Collapse
Affiliation(s)
- Daniela Vallejo
- Centro de Envejecimiento y Regeneración (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Carolina B Lindsay
- Centro de Envejecimiento y Regeneración (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Christian González-Billault
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile.,Geroscience Center for Brain Health and Metabolism (GERO), Santiago, Chile.,The Buck Institute for Research on Aging, Novato, CA, USA
| | - Nibaldo C Inestrosa
- Centro de Envejecimiento y Regeneración (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile
| |
Collapse
|
10
|
Whyte AJ, Trinoskey-Rice G, Davies RA, Woon EP, Foster SL, Shapiro LP, Li DC, Srikanth KD, Gil-Henn H, Gourley SL. Cell Adhesion Factors in the Orbitofrontal Cortex Control Cue-Induced Reinstatement of Cocaine Seeking and Amygdala-Dependent Goal Seeking. J Neurosci 2021; 41:5923-5936. [PMID: 34074735 PMCID: PMC8265806 DOI: 10.1523/jneurosci.0781-20.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 03/15/2021] [Accepted: 03/16/2021] [Indexed: 12/14/2022] Open
Abstract
Repeated cocaine exposure causes dendritic spine loss in the orbitofrontal cortex, which might contribute to poor orbitofrontal cortical function following drug exposure. One challenge, however, has been verifying links between neuronal structural plasticity and behavior, if any. Here we report that cocaine self-administration triggers the loss of dendritic spines on excitatory neurons in the orbitofrontal cortex of male and female mice (as has been reported in rats). To understand functional consequences, we locally ablated neuronal β1-integrins, cell adhesion receptors that adhere cells to the extracellular matrix and thus support dendritic spine stability. Degradation of β1-integrin tone: (1) caused dendritic spine loss, (2) exaggerated cocaine-seeking responses in a cue-induced reinstatement test, and (3) impaired the ability of mice to integrate new learning into familiar routines, a key function of the orbitofrontal cortex. Stimulating Abl-related gene kinase, overexpressing Proline-rich tyrosine kinase, and inhibiting Rho-associated coiled-coil containing kinase corrected response strategies, uncovering a β1-integrin-mediated signaling axis that controls orbitofrontal cortical function. Finally, use of a combinatorial gene silencing/chemogenetic strategy revealed that β1-integrins support the ability of mice to integrate new information into established behaviors by sustaining orbitofrontal cortical connections with the basolateral amygdala.SIGNIFICANCE STATEMENT Cocaine degenerates dendritic spines in the orbitofrontal cortex, a region of the brain involved in interlacing new information into established behaviors. One challenge has been verifying links between cellular structural stability and behavior, if any. In this second of two related investigations, we study integrin family receptors, which adhere cells to the extracellular matrix and thereby stabilize dendritic spines (see also DePoy et al., 2019). We reveal that β1-integrins in the orbitofrontal cortex control food- and cocaine-seeking behaviors. For instance, β1-integrin loss amplifies cocaine-seeking behavior and impairs the ability of mice to integrate new learning into familiar routines. We identify likely intracellular signaling partners by which β1-integrins support orbitofrontal cortical function and connectivity with the basolateral amygdala.
Collapse
Affiliation(s)
- Alonzo J Whyte
- Departments of Pediatrics and Psychiatry, Emory University School of Medicine, Atlanta, Georgia 30329
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia 30329
| | - Gracy Trinoskey-Rice
- Departments of Pediatrics and Psychiatry, Emory University School of Medicine, Atlanta, Georgia 30329
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia 30329
| | - Rachel A Davies
- Departments of Pediatrics and Psychiatry, Emory University School of Medicine, Atlanta, Georgia 30329
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia 30329
| | - Ellen P Woon
- Departments of Pediatrics and Psychiatry, Emory University School of Medicine, Atlanta, Georgia 30329
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia 30329
- Graduate Program in Neuroscience, Emory University, Atlanta, Georgia 30329
| | - Stephanie L Foster
- Departments of Pediatrics and Psychiatry, Emory University School of Medicine, Atlanta, Georgia 30329
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia 30329
- Graduate Program in Neuroscience, Emory University, Atlanta, Georgia 30329
| | - Lauren P Shapiro
- Departments of Pediatrics and Psychiatry, Emory University School of Medicine, Atlanta, Georgia 30329
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia 30329
- Graduate Program in Molecular and Systems Pharmacology, Emory University, Atlanta, Georgia 30329
| | - Dan C Li
- Departments of Pediatrics and Psychiatry, Emory University School of Medicine, Atlanta, Georgia 30329
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia 30329
- Graduate Program in Neuroscience, Emory University, Atlanta, Georgia 30329
| | | | - Hava Gil-Henn
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Shannon L Gourley
- Departments of Pediatrics and Psychiatry, Emory University School of Medicine, Atlanta, Georgia 30329
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia 30329
- Graduate Program in Neuroscience, Emory University, Atlanta, Georgia 30329
- Graduate Program in Molecular and Systems Pharmacology, Emory University, Atlanta, Georgia 30329
- Children's Healthcare of Atlanta, Atlanta, Georgia 30329
| |
Collapse
|
11
|
Glasgow SD, Ruthazer ES, Kennedy TE. Guiding synaptic plasticity: Novel roles for netrin-1 in synaptic plasticity and memory formation in the adult brain. J Physiol 2020; 599:493-505. [PMID: 32017127 DOI: 10.1113/jp278704] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 01/14/2020] [Indexed: 12/12/2022] Open
Abstract
Adult neural plasticity engages mechanisms that change synapse structure and function, yet many of the underlying events bear a striking similarity to processes that occur during the initial establishment of neural circuits during development. It is a long-standing hypothesis that the molecular mechanisms critical for neural development may also regulate synaptic plasticity related to learning and memory in adults. Netrins were initially described as chemoattractant guidance cues that direct cell and axon migration during embryonic development, yet they continue to be expressed by neurons in the adult brain. Recent findings have identified roles for netrin-1 in synaptogenesis during postnatal maturation, and in synaptic plasticity in the adult mammalian brain, regulating AMPA glutamate receptor trafficking at excitatory synapses. These findings provide an example of a conserved developmental guidance cue that is expressed by neurons in the adult brain and functions as a key regulator of activity-dependent synaptic plasticity. Notably, in humans, genetic polymorphisms in netrin-1 and its receptors have been linked to neurodevelopmental and neurodegenerative disorders. The molecular mechanisms associated with the synaptic function of netrin-1 therefore present new therapeutic targets for neuropathologies associated with memory dysfunction. Here, we summarize recent findings that link netrin-1 signalling to synaptic plasticity, and discuss the implications of these discoveries for the neurobiological basis of memory consolidation.
Collapse
Affiliation(s)
- Stephen D Glasgow
- Department of Neurology & Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, QC, H3A 2B4, Canada
| | - Edward S Ruthazer
- Department of Neurology & Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, QC, H3A 2B4, Canada
| | - Timothy E Kennedy
- Department of Neurology & Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, QC, H3A 2B4, Canada.,Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec, H3A 0C7, Canada
| |
Collapse
|
12
|
Hinton EA, Li DC, Allen AG, Gourley SL. Social Isolation in Adolescence Disrupts Cortical Development and Goal-Dependent Decision-Making in Adulthood, Despite Social Reintegration. eNeuro 2019; 6:ENEURO.0318-19.2019. [PMID: 31527057 PMCID: PMC6757188 DOI: 10.1523/eneuro.0318-19.2019] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 08/19/2019] [Indexed: 12/18/2022] Open
Abstract
The social environment influences neurodevelopment. Investigations using rodents to study this phenomenon commonly isolate subjects, then assess neurobehavioral consequences while animals are still isolated. This approach precludes one from dissociating the effects of on-going versus prior isolation, hindering our complete understanding of the consequences of social experience during particular developmental periods. Here, we socially isolated adolescent mice from postnatal day (P)31 to P60, then re-housed them into social groups. We tested their ability to select actions based on expected outcomes using multiple reinforcer devaluation and instrumental contingency degradation techniques. Social isolation in adolescence (but not adulthood) weakened instrumental response updating, causing mice to defer to habit-like behaviors. Habit biases were associated with glucocorticoid insufficiency in adolescence, oligodendrocyte marker loss throughout cortico-striatal regions, and dendritic spine and synaptic marker excess in the adult orbitofrontal cortex (OFC). Artificial, chemogenetic stimulation of the ventrolateral OFC in typical, healthy mice recapitulated response biases following isolation, causing habit-like behaviors. Meanwhile, correcting dendritic architecture by inhibiting the cytoskeletal regulatory protein ROCK remedied instrumental response updating defects in socially isolated mice. Our findings suggest that adolescence is a critical period during which social experience optimizes one's ability to seek and attain goals later in life. Age-typical dendritic spine elimination appears to be an essential factor, and in its absence, organisms may defer to habit-based behaviors.
Collapse
Affiliation(s)
- Elizabeth A Hinton
- Graduate Program in Neuroscience, Emory University, Atlanta, GA, 30329
- Center for Translational and Social Neuroscience, Emory University, Atlanta, GA, 30329
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, 30329
- Department of Pediatrics, Emory University, Atlanta, GA, 30329
- Department of Psychiatry, Emory University, Atlanta, GA, 30329
| | - Dan C Li
- Graduate Program in Neuroscience, Emory University, Atlanta, GA, 30329
- Center for Translational and Social Neuroscience, Emory University, Atlanta, GA, 30329
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, 30329
- Department of Pediatrics, Emory University, Atlanta, GA, 30329
- Department of Psychiatry, Emory University, Atlanta, GA, 30329
| | - Aylet G Allen
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, 30329
- Department of Pediatrics, Emory University, Atlanta, GA, 30329
| | - Shannon L Gourley
- Graduate Program in Neuroscience, Emory University, Atlanta, GA, 30329
- Center for Translational and Social Neuroscience, Emory University, Atlanta, GA, 30329
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, 30329
- Department of Pediatrics, Emory University, Atlanta, GA, 30329
- Department of Psychiatry, Emory University, Atlanta, GA, 30329
| |
Collapse
|
13
|
Rampérez A, Bartolomé-Martín D, García-Pascual A, Sánchez-Prieto J, Torres M. Photoconversion of FM1-43 Reveals Differences in Synaptic Vesicle Recycling and Sensitivity to Pharmacological Disruption of Actin Dynamics in Individual Synapses. ACS Chem Neurosci 2019; 10:2045-2059. [PMID: 30763065 DOI: 10.1021/acschemneuro.8b00712] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
The cycling of synaptic vesicles ensures that neurons can communicate adequately through their synapses on repeated occasions when activity is sustained, and several steps in this cycle are modulated by actin. The effects of pharmacological stabilization of actin with jasplakinolide or its depolymerization with latrunculin A was assessed on the synaptic vesicle cycle at individual boutons of cerebellar granule cells, using FM1-43 imaging to track vesicle recycling and its photoconversion to specifically label recycled organelles. Remarkable differences in the recycling capacity of individual boutons are evident, and their dependence on the actin cytoskeleton for recycling is clear. Disrupting actin dynamics causes a loss of functional boutons, and while this indicates that exo/endocytotic cycling in boutons is fully dependent on such events, this dependence is only partial in other boutons. Indeed, exocytosis and vesicle trafficking are impaired significantly by stabilizing or depolymerizing actin, whereas repositioning recycled vesicles at the active zone seems to be dependent on actin polymerization alone. These findings support the hypothesis that different steps of synaptic vesicle cycling depend on actin dynamics and that such dependence varies among individual boutons.
Collapse
Affiliation(s)
- Alberto Rampérez
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid 28040, Spain
| | - David Bartolomé-Martín
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid 28040, Spain
| | - Angeles García-Pascual
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid 28040, Spain
| | - Jose Sánchez-Prieto
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid 28040, Spain
| | - Magdalena Torres
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid 28040, Spain
| |
Collapse
|
14
|
Shapiro LP, Kietzman HW, Guo J, Rainnie DG, Gourley SL. Rho-kinase inhibition has antidepressant-like efficacy and expedites dendritic spine pruning in adolescent mice. Neurobiol Dis 2019; 124:520-530. [PMID: 30593834 PMCID: PMC6365018 DOI: 10.1016/j.nbd.2018.12.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 11/15/2018] [Accepted: 12/21/2018] [Indexed: 12/29/2022] Open
Abstract
Adolescence represents a critical period of neurodevelopment, defined by structural and synaptic pruning within the prefrontal cortex. While characteristic of typical development, this structural instability may open a window of vulnerability to developing neuropsychiatric disorders, including depression. Thus, therapeutic interventions that support or expedite neural remodeling in adolescence may be advantageous. Here, we inhibited the neuronally-expressed cytoskeletal regulatory factor Rho-kinase (ROCK), focusing primarily on the clinically-viable ROCK inhibitor fasudil. ROCK inhibition had rapid antidepressant-like effects in adolescent mice, and its efficacy was comparable to ketamine and fluoxetine. It also modified levels of the antidepressant-related signaling factors, tropomyosin/tyrosine receptor kinase B and Akt, as well as the postsynaptic marker PSD-95, in the ventromedial prefrontal cortex (vmPFC). Meanwhile, adolescent-typical dendritic spine pruning on excitatory pyramidal neurons in the vmPFC was expedited. Further, vmPFC-specific shRNA-mediated reduction of ROCK2, the dominant ROCK isoform in the brain, had antidepressant-like consequences. We cautiously suggest that ROCK inhibitors may have therapeutic potential for adolescent-onset depression.
Collapse
Affiliation(s)
- Lauren P Shapiro
- Molecular and Systems Pharmacology, Emory University, Atlanta, GA, United States; Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States; Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States
| | - Henry W Kietzman
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States; Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States; Graduate Program in Neuroscience, Emory University, Atlanta, GA, United States
| | - Jidong Guo
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States; Department of Psychiatry, Emory University School of Medicine, Atlanta, GA, United States
| | - Donald G Rainnie
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States; Department of Psychiatry, Emory University School of Medicine, Atlanta, GA, United States
| | - Shannon L Gourley
- Molecular and Systems Pharmacology, Emory University, Atlanta, GA, United States; Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States; Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States; Graduate Program in Neuroscience, Emory University, Atlanta, GA, United States; Department of Psychiatry, Emory University School of Medicine, Atlanta, GA, United States.
| |
Collapse
|
15
|
Wang Q, Wang D, Shibata S, Ji T, Zhang L, Zhang R, Yang H, Ma L, Jiao J. Group I metabotropic glutamate receptor activation induces TRPC6-dependent calcium influx and RhoA activation in cultured human kidney podocytes. Biochem Biophys Res Commun 2019; 511:374-380. [PMID: 30782481 DOI: 10.1016/j.bbrc.2019.02.062] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Accepted: 02/11/2019] [Indexed: 12/20/2022]
Abstract
Researches have shown that mice lacking the metabotropic glutamate receptor 1 (mGluR) showed albuminuria, remodeling of F-actin, with loss of stress fibers. Selective group I mGluRs agonist (S)-3,5-dihydroxyphenylglycine (DHPG) attenuated albuminuria in several rodent models of nephrotic syndrome. However, the molecular mechanism is obscure. Using a human podocyte cell line, we here investigated the molecular mechanisms of group I mGluRs-induced calcium influx and the formation of stress fibers. Our data showed that group I mGluRs activation by DHPG induced a significant calcium influx, and promoted cytoskeletal stress fiber formation and focal adhesions in podocytes. Pre-incubating podocytes with non-selective inhibitor of transient receptor potential channels (TRPC), or the knockdown of TRPC6 attenuated the calcium influx and the stress fiber formation induced by DHPG. Further, DHPG resulted in an increase of active RhoA expression. However, the knockdown of RhoA by siRNA abolished the DHPG-induced increase in stress fibers. Additionally, nonselective inhibitors of TRPC or TRPC6 knockdown clearly inhibited RhoA activation induced by DHPG, as assessed by Glutathione-S-transferase pull-down assay followed by Western blotting. Taken together, our findings suggest TRPC6 regulates actin stress fiber formation and focal adhesions via the RhoA pathway in response to group I mGluRs activation. Our data can potentially explain the mechanism of protective action of group I mGluRs in glomerular podocyte injury.
Collapse
Affiliation(s)
- Qin Wang
- Department of Nephrology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Derun Wang
- Department of Geriatrics, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shigeru Shibata
- Division of Nephrology, Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| | - Tianrong Ji
- Department of Nephrology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Lei Zhang
- Department of Pediatrics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Rui Zhang
- Department of Nephrology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - He Yang
- Department of Nephrology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Linlin Ma
- Department of Nephrology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jundong Jiao
- Department of Nephrology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China; Institute of Nephrology, Harbin Medical University, Harbin, China.
| |
Collapse
|
16
|
Pyk2 Signaling through Graf1 and RhoA GTPase Is Required for Amyloid-β Oligomer-Triggered Synapse Loss. J Neurosci 2019; 39:1910-1929. [PMID: 30626696 DOI: 10.1523/jneurosci.2983-18.2018] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Revised: 12/20/2018] [Accepted: 12/22/2018] [Indexed: 12/15/2022] Open
Abstract
The intracellular tyrosine kinase Pyk2 (PTK2B) is related to focal adhesion kinase and localizes to postsynaptic sites in brain. Pyk2 genetic variation contributes to late onset Alzheimer's disease (AD) risk. We recently observed that Pyk2 is required for synapse loss and for learning deficits in a transgenic mouse model of AD. Here, we explore the cellular and biochemical basis for the action of Pyk2 tyrosine kinase in amyloid-β oligomer (Aβo)-induced dendritic spine loss. Overexpression of Pyk2 reduces dendritic spine density of hippocampal neurons by a kinase-dependent mechanism. Biochemical isolation of Pyk2-interacting proteins from brain identifies Graf1c, a RhoA GTPase-activating protein inhibited by Pyk2. Aβo-induced reductions in dendritic spine motility and chronic spine loss require both Pyk2 kinase and RhoA activation. Thus, Pyk2 functions at postsynaptic sites to modulate F-actin control by RhoA and regulate synapse maintenance of relevance to AD risk.SIGNIFICANCE STATEMENT Genetic variation at the Pyk2 locus is a risk for Alzheimer's disease. We have observed that Pyk2 is required for AD transgenic synapse loss and memory dysfunction. However, the cellular and biochemical basis for Pyk2 function related to AD is not defined. Here, we show that brain Pyk2 interacts with the RhoGAP protein Graf1 to alter dendritic spine stability via RhoA GTPase. Amyloid-β oligomer-induced dendritic spine loss requires the Pyk2/Graf1 pathway.
Collapse
|
17
|
Werner CT, Viswanathan R, Martin JA, Gobira PH, Mitra S, Thomas SA, Wang ZJ, Liu JF, Stewart AF, Neve RL, Li JX, Gancarz AM, Dietz DM. E3 Ubiquitin-Protein Ligase SMURF1 in the Nucleus Accumbens Mediates Cocaine Seeking. Biol Psychiatry 2018; 84:881-892. [PMID: 30158054 PMCID: PMC6260585 DOI: 10.1016/j.biopsych.2018.07.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 06/25/2018] [Accepted: 07/10/2018] [Indexed: 12/15/2022]
Abstract
BACKGROUND Substance use disorder is a neurobiological disease characterized by episodes of relapse despite periods of withdrawal. It is thought that neuroadaptations in discrete brain areas of the reward pathway, including the nucleus accumbens, underlie these aberrant behaviors. The ubiquitin-proteasome system degrades proteins and has been shown to be involved in cocaine-induced plasticity, but the role of E3 ubiquitin ligases, which conjugate ubiquitin to substrates, is unknown. Here, we examined E3 ubiquitin-protein ligase SMURF1 (SMURF1) in neuroadaptations and relapse behavior during withdrawal following cocaine self-administration. METHODS SMURF1 and downstream targets ras homolog gene family, member A (RhoA), SMAD1/5, and Runt-related transcript factor 2 were examined using Western blotting (n = 9-11/group), quantitative polymerase chain reaction (n = 6-9/group), co-immunoprecipitation (n = 9-11/group), tandem ubiquitin binding entities affinity purification (n = 5-6/group), and quantitative chromatin immunoprecipitation (n = 3-6/group) (2 rats/sample). Viral-mediated gene transfer (n = 7-12/group) and intra-accumbal microinjections (n = 9-10/group) were used to examine causal roles of SMURF1 and substrate RhoA, respectively, in cue-induced cocaine seeking. RESULTS SMURF1 protein expression was decreased, while SMURF1 substrates RhoA and SMAD1/5 were increased, in the nucleus accumbens on withdrawal day 7, but not on withdrawal day 1, following cocaine self-administration. Viral-mediated gene transfer of Smurf1 or constitutive activation of RhoA attenuated cue-induced cocaine seeking, while catalytically inactive Smurf1 enhanced cocaine seeking. Furthermore, SMURF1-regulated, SMAD1/5-associated transcription factor Runt-related transcript factor 2 displayed increased binding at promoter regions of genes previously associated with cocaine-induced plasticity. CONCLUSIONS SMURF1 is a key mediator of neuroadaptations in the nucleus accumbens following cocaine exposure and mediates cue-induced cocaine seeking during withdrawal.
Collapse
Affiliation(s)
- Craig T Werner
- Department of Pharmacology and Toxicology, Program in Neuroscience, Research Institute on Addictions, The State University of New York at Buffalo, Buffalo, New York
| | - Rathipriya Viswanathan
- Department of Pharmacology and Toxicology, Program in Neuroscience, Research Institute on Addictions, The State University of New York at Buffalo, Buffalo, New York
| | - Jennifer A Martin
- Department of Pharmacology and Toxicology, Program in Neuroscience, Research Institute on Addictions, The State University of New York at Buffalo, Buffalo, New York
| | - Pedro H Gobira
- Department of Pharmacology and Toxicology, Program in Neuroscience, Research Institute on Addictions, The State University of New York at Buffalo, Buffalo, New York; Department of Physics and Chemistry, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Swarup Mitra
- Department of Pharmacology and Toxicology, Program in Neuroscience, Research Institute on Addictions, The State University of New York at Buffalo, Buffalo, New York
| | - Shruthi A Thomas
- Department of Pharmacology and Toxicology, Program in Neuroscience, Research Institute on Addictions, The State University of New York at Buffalo, Buffalo, New York
| | - Zi-Jun Wang
- Department of Pharmacology and Toxicology, Program in Neuroscience, Research Institute on Addictions, The State University of New York at Buffalo, Buffalo, New York
| | - Jian-Feng Liu
- Department of Pharmacology and Toxicology, Program in Neuroscience, Research Institute on Addictions, The State University of New York at Buffalo, Buffalo, New York
| | - Andrew F Stewart
- Department of Pharmacology and Toxicology, Program in Neuroscience, Research Institute on Addictions, The State University of New York at Buffalo, Buffalo, New York
| | - Rachael L Neve
- Gene Delivery Technology Core, Massachusetts General Hospital, Cambridge, Massachusetts
| | - Jun-Xu Li
- Department of Pharmacology and Toxicology, Program in Neuroscience, Research Institute on Addictions, The State University of New York at Buffalo, Buffalo, New York
| | - Amy M Gancarz
- Department of Pharmacology and Toxicology, Program in Neuroscience, Research Institute on Addictions, The State University of New York at Buffalo, Buffalo, New York; Department of Psychology, California State University, Bakersfield, Bakersfield, California
| | - David M Dietz
- Department of Pharmacology and Toxicology, Program in Neuroscience, Research Institute on Addictions, The State University of New York at Buffalo, Buffalo, New York; Department of Psychology, The State University of New York at Buffalo, Buffalo, New York.
| |
Collapse
|
18
|
Swanson AM, DePoy LM, Gourley SL. Inhibiting Rho kinase promotes goal-directed decision making and blocks habitual responding for cocaine. Nat Commun 2017; 8:1861. [PMID: 29187752 PMCID: PMC5707361 DOI: 10.1038/s41467-017-01915-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2017] [Accepted: 10/25/2017] [Indexed: 01/04/2023] Open
Abstract
The prelimbic prefrontal cortex is necessary for associating actions with their consequences, enabling goal-directed decision making. We find that the strength of action–outcome conditioning correlates with dendritic spine density in prelimbic cortex, suggesting that new action–outcome learning involves dendritic spine plasticity. To test this, we inhibited the cytoskeletal regulatory factor Rho kinase. We find that the inhibitor fasudil enhances action–outcome memory, resulting in goal-directed behavior in mice that would otherwise express stimulus-response habits. Fasudil transiently reduces prelimbic cortical dendritic spine densities during a period of presumed memory consolidation, but only when paired with new learning. Fasudil also blocks habitual responding for cocaine, an effect that persists over time, across multiple contexts, and depends on actin polymerization. We suggest that Rho kinase inhibition promotes goal-oriented action selection by augmenting the plasticity of prelimbic cortical dendritic spines during the formation of new action–outcome memories. Action-outcome learning requires the prelimbic prefrontal cortex. Here the authors report that fasudil, a Rho kinase inhibitor, reduces dendritic spine densities on prelimbic neurons in an activity-dependent manner, stimulating goal-directed actions, and reducing habitual responding for cocaine.
Collapse
Affiliation(s)
- Andrew M Swanson
- Departments of Pediatrics and Psychiatry, Emory University School of Medicine, 954 Gatewood Road NE, Atlanta, GA, 30329, USA.,Yerkes National Primate Research Center, Graduate Program in Neuroscience, Emory University, 954 Gatewood Road NE, Atlanta, GA, 30329, USA
| | - Lauren M DePoy
- Departments of Pediatrics and Psychiatry, Emory University School of Medicine, 954 Gatewood Road NE, Atlanta, GA, 30329, USA.,Yerkes National Primate Research Center, Graduate Program in Neuroscience, Emory University, 954 Gatewood Road NE, Atlanta, GA, 30329, USA
| | - Shannon L Gourley
- Departments of Pediatrics and Psychiatry, Emory University School of Medicine, 954 Gatewood Road NE, Atlanta, GA, 30329, USA. .,Yerkes National Primate Research Center, Graduate Program in Neuroscience, Emory University, 954 Gatewood Road NE, Atlanta, GA, 30329, USA.
| |
Collapse
|
19
|
Lacampagne A, Liu X, Reiken S, Bussiere R, Meli AC, Lauritzen I, Teich AF, Zalk R, Saint N, Arancio O, Bauer C, Duprat F, Briggs CA, Chakroborty S, Stutzmann GE, Shelanski ML, Checler F, Chami M, Marks AR. Post-translational remodeling of ryanodine receptor induces calcium leak leading to Alzheimer's disease-like pathologies and cognitive deficits. Acta Neuropathol 2017. [PMID: 28631094 DOI: 10.1007/s00401-017-1733-7] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The mechanisms underlying ryanodine receptor (RyR) dysfunction associated with Alzheimer disease (AD) are still not well understood. Here, we show that neuronal RyR2 channels undergo post-translational remodeling (PKA phosphorylation, oxidation, and nitrosylation) in brains of AD patients, and in two murine models of AD (3 × Tg-AD, APP +/- /PS1 +/-). RyR2 is depleted of calstabin2 (KFBP12.6) in the channel complex, resulting in endoplasmic reticular (ER) calcium (Ca2+) leak. RyR-mediated ER Ca2+ leak activates Ca2+-dependent signaling pathways, contributing to AD pathogenesis. Pharmacological (using a novel RyR stabilizing drug Rycal) or genetic rescue of the RyR2-mediated intracellular Ca2+ leak improved synaptic plasticity, normalized behavioral and cognitive functions and reduced Aβ load. Genetically altered mice with congenitally leaky RyR2 exhibited premature and severe defects in synaptic plasticity, behavior and cognitive function. These data provide a mechanism underlying leaky RyR2 channels, which could be considered as potential AD therapeutic targets.
Collapse
|
20
|
TRPC Channels and Neuron Development, Plasticity, and Activities. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 976:95-110. [PMID: 28508316 DOI: 10.1007/978-94-024-1088-4_9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
In this chapter, we mainly focus on the functions of TRPC channels in brain development, including neural progenitor proliferation, neurogenesis, neuron survival, axon guidance, dendritic morphology, synaptogenesis, and neural plasticity. We also notice emerging advances in understanding the functions of TRPC channels in periphery, especially their functions in sensation and nociception in dorsal root ganglion (DRG). Because TRPC channels are expressed in all major types of glial cells, which account for at least half of total cells in the brain, TRPC channels may act as modulators for glial functions as well. The future challenges for studying these channels could be (1) the detailed protein structures of these channels, (2) their cell type-specific functions, (3) requirement for their specific blockers or activators, and (4) change in the channel conformation in the brain.
Collapse
|
21
|
Korkotian E, Oni-Biton E, Segal M. The role of the store-operated calcium entry channel Orai1 in cultured rat hippocampal synapse formation and plasticity. J Physiol 2016; 595:125-140. [PMID: 27393042 DOI: 10.1113/jp272645] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 06/30/2016] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS The role of non-synaptic calcium entry in the formation and functions of dendritic spines was studied in dissociated cultured rat hippocampal neurons. Orai1, a store-operated calcium channel, is found in dendritic spines. Orai1 co-localizes in dendritic spines with STIM2 under conditions of lower [Ca2+ ]o. Orai1 channels are associated with the formation of new dendritic spines in response to elevated [Ca2+ ]o. Lack of Orai1, either by transfection with a dominant negative construct or with small interfering RNA to Orai1, results in retarded dendritic spines, an increase in density of filopodia, lower synaptic connectivity and the ability to undergo plastic changes. These results highlight a novel role for Orai1 in synapse formation, maturation and plasticity. ABSTRACT The possible role of store operated calcium entry (SOCE) through the Orai1 channel in the formation and functions of dendritic spines was studied in cultured hippocampal neurons. In calcium store-depleted neurons, a transient elevation of extracellular calcium concentration ([Ca2+ ]o ) caused a rise in [Ca2+ ]i that was mediated by activation of the SOCE. The store depletion resulted in an increase in stromal interacting molecule 2 (an endoplasmic calcium sensor) association with Orai1 in dendritic spines. The response to the rise in [Ca2+ ]o was larger in spines endowed with a cluster of Orai1 molecules than in spines devoid of Orai1. Transfection of neurons with a dominant negative Orai1 resulted in retarded maturation of dendritic spines, a reduction in synaptic connectivity with afferent neurons and a reduction in the ability to undergo morphological changes following induction of chemical long-term potentiation. Similarly, small interfering RNA (siRNA)-treated neurons had fewer mature dendritic spines, and lower rates of mEPSCs compared to scrambled control siRNA-treated neurons. Thus, influx of calcium through Orai1 channels facilitates the maturation of dendritic spines and the formation of functional synapses in central neurons.
Collapse
Affiliation(s)
- Eduard Korkotian
- Department of Neurobiology, The Weizmann Institute, Rehovot, Israel
| | - Efrat Oni-Biton
- Department of Neurobiology, The Weizmann Institute, Rehovot, Israel
| | - Menahem Segal
- Department of Neurobiology, The Weizmann Institute, Rehovot, Israel
| |
Collapse
|
22
|
Inan SY, Soner BC, Sahin AS. Behavioural effects of basal ganglia rho-kinase inhibition in the unilateral 6-hydroxydopamine rat model of Parkinson's disease. Metab Brain Dis 2016; 31:849-57. [PMID: 26996632 DOI: 10.1007/s11011-016-9820-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 03/17/2016] [Indexed: 11/25/2022]
Abstract
Parkinson's disease (PD) is one of the most common neurodegenerative disorders, which affects more than six million people in the world. While current available pharmacological therapies for PD in the early stages of the disease usually improve motor symptoms, they cause side effects, such as fluctuations and dyskinesias in the later stages. In this later stage, high frequency deep brain stimulation of the subthalamic nucleus (STN-DBS) is a treatment option which is most successful to treat drug resistant advanced PD. It has previously been demonstrated that activation of Rho/Rho-kinase pathway is involved in the dopaminergic cell degeneration which is one of the main characteristics of PD pathology. In addition, the involvement of this pathway has been suggested in diverse cellular events in the central nervous system; such as epilepsy, anxiety-related behaviors, regulation of dendritic and axonal morphology, antinociception, subarachnoid haemorrhage, spinal cord injury and amyotrophic lateral sclerosis. However, up to date, to our knowledge there are no previous reports showing the beneficial effects of the potent Rho-kinase inhibitor Y-27632 in the 6-hydroxydopamine (6-OHDA) rat model of PD. Therefore, in the present study, we investigated the behavioural effects of basal ganglia Y-27632 microinjections in this PD model. Our results indicated that basal ganglia Y-27632 microinjections significantly decreased the number of contralateral rotations-induced by apomorphine, significantly increased line crossings in the open-field test, contralateral forelimb use in the limb-use asymmetry test and contralateral tape playing time in the somatosensory asymmetry test, which may suggest that Y-27632 could be a potentially active antiparkinsonian agent.
Collapse
Affiliation(s)
- Salim Yalcin Inan
- Department of Medical Pharmacology, Meram Faculty of Medicine, University of Konya-NE, Akyokus, 42080, Konya, Meram, Turkey.
| | - Burak Cem Soner
- Department of Medical Pharmacology, Meram Faculty of Medicine, University of Konya-NE, Akyokus, 42080, Konya, Meram, Turkey
| | - Ayse Saide Sahin
- Department of Medical Pharmacology, Meram Faculty of Medicine, University of Konya-NE, Akyokus, 42080, Konya, Meram, Turkey
| |
Collapse
|
23
|
Haws HJ, McNeil MA, Hansen MDH. Control of cell mechanics by RhoA and calcium fluxes during epithelial scattering. Tissue Barriers 2016; 4:e1187326. [PMID: 27583192 DOI: 10.1080/21688370.2016.1187326] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 04/25/2016] [Accepted: 05/01/2016] [Indexed: 12/17/2022] Open
Abstract
Epithelial tissues use adherens junctions to maintain tight interactions and coordinate cellular activities. Adherens junctions are remodeled during epithelial morphogenesis, including instances of epithelial-mesenchymal transition, or EMT, wherein individual cells detach from the tissue and migrate as individual cells. EMT has been recapitulated by growth factor induction of epithelial scattering in cell culture. In culture systems, cells undergo a highly reproducible series of cell morphology changes, most notably cell spreading followed by cellular compaction and cell migration. These morphology changes are accompanied by striking actin rearrangements. The current evidence suggests that global changes in actomyosin-based cellular contractility, first a loss of contractility during spreading and its activation during cell compaction, are the main drivers of epithelial scattering. In this review, we focus on how spreading and contractility might be controlled during epithelial scattering. While we propose a central role for RhoA, which is well known to control cellular contractility in multiple systems and whose role in epithelial scattering is well accepted, we suggest potential roles for additional cellular systems whose role in epithelial cell biology has been less well documented. In particular, we propose critical roles for vesicle recycling, calcium channels, and calcium-dependent kinases.
Collapse
Affiliation(s)
- Hillary J Haws
- Physiology and Developmental Biology, Brigham Young University , Provo, UT, USA
| | - Melissa A McNeil
- Physiology and Developmental Biology, Brigham Young University , Provo, UT, USA
| | - Marc D H Hansen
- Physiology and Developmental Biology, Brigham Young University , Provo, UT, USA
| |
Collapse
|
24
|
Huang KH, Lan YT, Chen MH, Chao Y, Lo SS, Li AFY, Wu CW, Chiou SH, Yang MH, Shyr YM, Fang WL. The Correlation Between RhoA Expression and Clinicopathological Characteristics in Gastric Cancer Patients After Curative Surgery. World J Surg 2016; 39:2289-99. [PMID: 26013205 DOI: 10.1007/s00268-015-3095-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
BACKGROUND The expression of RhoA, a member of the ras homologue family, is reported to be involved in tumorigenesis in some cancers; however, its prognostic value in gastric cancer is controversial. METHODS Between April 1988 and January 2005, a total of 206 gastric cancer patients receiving curative surgery were enrolled in this study. Immunohistochemical staining of the RhoA protein was performed, and the clinicopathological characteristics and initial recurrence patterns were compared between low RhoA expression (n = 55) and high RhoA expression (n = 151) gastric cancer patients. RESULTS For intestinal-type (n = 134) gastric cancer, there is no significant difference between the clinicopathological characteristics and RhoA expression. However, for diffuse-type (n = 82) gastric cancer, high RhoA expression was associated with more advanced pathological N category compared to low RhoA expression. A multivariate analysis revealed that age, pathological T and N categories, and RhoA expression were independent prognostic factors for overall survival after curative surgery. For all patients, the five-year overall survival rates and disease-free survival rates were higher in patients with low RhoA expression compared to those with high RhoA expression, which was observed in diffuse-type gastric cancer, not in intestinal-type gastric cancer. With regard to the initial recurrence pattern, patients with high RhoA expression had more distant metastasis compared to those with low RhoA expression, especially more liver metastasis. CONCLUSIONS RhoA expression is an independent prognostic factor for gastric cancer, especially for diffuse-type. We should be aware of liver metastasis during the follow-up of gastric cancer with high RhoA expression.
Collapse
Affiliation(s)
- Kuo-Hung Huang
- Division of General Surgery, Department of Surgery, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Rd., Beitou District, Taipei City, 11217, Taiwan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Schreiber J, Végh MJ, Dawitz J, Kroon T, Loos M, Labonté D, Li KW, Van Nierop P, Van Diepen MT, De Zeeuw CI, Kneussel M, Meredith RM, Smit AB, Van Kesteren RE. Ubiquitin ligase TRIM3 controls hippocampal plasticity and learning by regulating synaptic γ-actin levels. J Cell Biol 2015; 211:569-86. [PMID: 26527743 PMCID: PMC4639863 DOI: 10.1083/jcb.201506048] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 10/06/2015] [Indexed: 12/24/2022] Open
Abstract
TRIM3 regulates synaptic γ-actin levels. TRIM3-deficient mice consequently have higher hippocampal spine densities, increased long-term potentiation, and enhanced contextual fear memory consolidation, indicating that temporal control of ACTG1 levels by TRIM3 is required to constrain hippocampal plasticity within physiological boundaries. Synaptic plasticity requires remodeling of the actin cytoskeleton. Although two actin isoforms, β- and γ-actin, are expressed in dendritic spines, the specific contribution of γ-actin in the expression of synaptic plasticity is unknown. We show that synaptic γ-actin levels are regulated by the E3 ubiquitin ligase TRIM3. TRIM3 protein and Actg1 transcript are colocalized in messenger ribonucleoprotein granules responsible for the dendritic targeting of messenger RNAs. TRIM3 polyubiquitylates γ-actin, most likely cotranslationally at synaptic sites. Trim3−/− mice consequently have increased levels of γ-actin at hippocampal synapses, resulting in higher spine densities, increased long-term potentiation, and enhanced short-term contextual fear memory consolidation. Interestingly, hippocampal deletion of Actg1 caused an increase in long-term fear memory. Collectively, our findings suggest that temporal control of γ-actin levels by TRIM3 is required to regulate the timing of hippocampal plasticity. We propose a model in which TRIM3 regulates synaptic γ-actin turnover and actin filament stability and thus forms a transient inhibitory constraint on the expression of hippocampal synaptic plasticity.
Collapse
Affiliation(s)
- Joerg Schreiber
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, VU University, 1081 HV Amsterdam, Netherlands
| | - Marlene J Végh
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, VU University, 1081 HV Amsterdam, Netherlands
| | - Julia Dawitz
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, VU University, 1081 HV Amsterdam, Netherlands
| | - Tim Kroon
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, VU University, 1081 HV Amsterdam, Netherlands
| | - Maarten Loos
- Sylics (Synaptologics BV), 1008 BA Amsterdam, Netherlands
| | - Dorthe Labonté
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, D-20251 Hamburg, Germany
| | - Ka Wan Li
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, VU University, 1081 HV Amsterdam, Netherlands
| | - Pim Van Nierop
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, VU University, 1081 HV Amsterdam, Netherlands
| | - Michiel T Van Diepen
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, VU University, 1081 HV Amsterdam, Netherlands
| | - Chris I De Zeeuw
- Department of Neuroscience, Erasmus Medical Center, 3000 DR Rotterdam, Netherlands Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Science, 1105 BA Amsterdam, Netherlands
| | - Matthias Kneussel
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, D-20251 Hamburg, Germany
| | - Rhiannon M Meredith
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, VU University, 1081 HV Amsterdam, Netherlands
| | - August B Smit
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, VU University, 1081 HV Amsterdam, Netherlands
| | - Ronald E Van Kesteren
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, VU University, 1081 HV Amsterdam, Netherlands
| |
Collapse
|
26
|
Abstract
Dendritic protein synthesis and actin cytoskeleton reorganization are important events required for the consolidation of hippocampal LTP and memory. However, the temporal and spatial relationships between these two processes remain unclear. Here, we report that treatment of adult rat hippocampal slices with BDNF or with tetraethylammonium (TEA), which induces a chemical form of LTP, produces a rapid and transient increase in RhoA protein levels. Changes in RhoA were restricted to dendritic spines of CA3 and CA1 and require de novo protein synthesis regulated by mammalian target of rapamycin (mTOR). BDNF-mediated stimulation of RhoA activity, cofilin phosphorylation, and actin polymerization were completely suppressed by protein synthesis inhibitors. Furthermore, intrahippocampal injections of RhoA antisense oligodeoxynucleotides inhibited theta burst stimulation (TBS)-induced RhoA upregulation in dendritic spines and prevented LTP consolidation. Addition of calpain inhibitors after BDNF or TEA treatment maintained RhoA levels elevated and prolonged the effects of BDNF and TEA on actin polymerization. Finally, the use of isoform-selective calpain inhibitors revealed that calpain-2 was involved in RhoA synthesis, whereas calpain-1 mediated RhoA degradation. Overall, this mechanism provides a novel link between dendritic protein synthesis and reorganization of the actin cytoskeleton in hippocampal dendritic spines during LTP consolidation.
Collapse
|
27
|
Chazeau A, Garcia M, Czöndör K, Perrais D, Tessier B, Giannone G, Thoumine O. Mechanical coupling between transsynaptic N-cadherin adhesions and actin flow stabilizes dendritic spines. Mol Biol Cell 2015; 26:859-73. [PMID: 25568337 PMCID: PMC4342023 DOI: 10.1091/mbc.e14-06-1086] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
A combination of quantitative live imaging of fluorescently tagged actin, N-cadherin, and myosin in primary neurons and computer modeling of actin dynamics shows that a clutch-like mechanism connecting N-cadherin–based transsynaptic adhesions and the actin/myosin network drives the stabilization of dendritic filopodia into spines. The morphology of neuronal dendritic spines is a critical indicator of synaptic function. It is regulated by several factors, including the intracellular actin/myosin cytoskeleton and transcellular N-cadherin adhesions. To examine the mechanical relationship between these molecular components, we performed quantitative live-imaging experiments in primary hippocampal neurons. We found that actin turnover and structural motility were lower in dendritic spines than in immature filopodia and increased upon expression of a nonadhesive N-cadherin mutant, resulting in an inverse relationship between spine motility and actin enrichment. Furthermore, the pharmacological stimulation of myosin II induced the rearward motion of actin structures in spines, showing that myosin II exerts tension on the actin network. Strikingly, the formation of stable, spine-like structures enriched in actin was induced at contacts between dendritic filopodia and N-cadherin–coated beads or micropatterns. Finally, computer simulations of actin dynamics mimicked various experimental conditions, pointing to the actin flow rate as an important parameter controlling actin enrichment in dendritic spines. Together these data demonstrate that a clutch-like mechanism between N-cadherin adhesions and the actin flow underlies the stabilization of dendritic filopodia into mature spines, a mechanism that may have important implications in synapse initiation, maturation, and plasticity in the developing brain.
Collapse
Affiliation(s)
- Anaël Chazeau
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, Unité Mixte de Recherche 5297, F-33000 Bordeaux, France Interdisciplinary Institute for Neuroscience, Centre Nationale de la Recherche Scientifique, Unité Mixte de Recherche 5297, F-33000 Bordeaux, France
| | - Mikael Garcia
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, Unité Mixte de Recherche 5297, F-33000 Bordeaux, France Interdisciplinary Institute for Neuroscience, Centre Nationale de la Recherche Scientifique, Unité Mixte de Recherche 5297, F-33000 Bordeaux, France CYTOO, Minatec, Grenoble, 38054 Grenoble, France
| | - Katalin Czöndör
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, Unité Mixte de Recherche 5297, F-33000 Bordeaux, France Interdisciplinary Institute for Neuroscience, Centre Nationale de la Recherche Scientifique, Unité Mixte de Recherche 5297, F-33000 Bordeaux, France
| | - David Perrais
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, Unité Mixte de Recherche 5297, F-33000 Bordeaux, France Interdisciplinary Institute for Neuroscience, Centre Nationale de la Recherche Scientifique, Unité Mixte de Recherche 5297, F-33000 Bordeaux, France
| | - Béatrice Tessier
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, Unité Mixte de Recherche 5297, F-33000 Bordeaux, France Interdisciplinary Institute for Neuroscience, Centre Nationale de la Recherche Scientifique, Unité Mixte de Recherche 5297, F-33000 Bordeaux, France
| | - Grégory Giannone
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, Unité Mixte de Recherche 5297, F-33000 Bordeaux, France Interdisciplinary Institute for Neuroscience, Centre Nationale de la Recherche Scientifique, Unité Mixte de Recherche 5297, F-33000 Bordeaux, France
| | - Olivier Thoumine
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, Unité Mixte de Recherche 5297, F-33000 Bordeaux, France Interdisciplinary Institute for Neuroscience, Centre Nationale de la Recherche Scientifique, Unité Mixte de Recherche 5297, F-33000 Bordeaux, France
| |
Collapse
|
28
|
Lamprecht R. The actin cytoskeleton in memory formation. Prog Neurobiol 2014; 117:1-19. [DOI: 10.1016/j.pneurobio.2014.02.001] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Revised: 02/02/2014] [Accepted: 02/03/2014] [Indexed: 01/21/2023]
|
29
|
Franco-Villanueva A, Fernández-López E, Gabandé-Rodríguez E, Bañón-Rodríguez I, Esteban JA, Antón IM, Ledesma MD. WIP modulates dendritic spine actin cytoskeleton by transcriptional control of lipid metabolic enzymes. Hum Mol Genet 2014; 23:4383-95. [PMID: 24698977 DOI: 10.1093/hmg/ddu155] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
We identify Wiskott-Aldrich syndrome protein (WASP)-interacting protein (WIP) as a novel component of neuronal synapses whose absence increases dendritic spine size and filamentous actin levels in an N-WASP/Arp2/3-independent, RhoA/ROCK/profilinIIa-dependent manner. These effects depend on the reduction of membrane sphingomyelin (SM) due to transcriptional upregulation of neutral sphingomyelinase (NSM) through active RhoA; this enhances RhoA binding to the membrane, raft partitioning and activation in steady state but prevents RhoA changes in response to stimulus. Inhibition of NSM or SM addition reverses RhoA, filamentous actin and functional anomalies in synapses lacking WIP. Our findings characterize WIP as a link between membrane lipid composition and actin cytoskeleton at dendritic spines. They also contribute to explain cognitive deficits shared by individuals bearing mutations in the region assigned to the gene encoding for WIP.
Collapse
Affiliation(s)
- Ana Franco-Villanueva
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM) and Centro Nacional de Biotecnología (CNB-CSIC), Madrid 28049, Spain
| | - Estefanía Fernández-López
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM) and Centro Nacional de Biotecnología (CNB-CSIC), Madrid 28049, Spain
| | | | | | | | - Inés M Antón
- Centro Nacional de Biotecnología (CNB-CSIC), Madrid 28049, Spain
| | | |
Collapse
|
30
|
p140Cap regulates memory and synaptic plasticity through Src-mediated and citron-N-mediated actin reorganization. J Neurosci 2014; 34:1542-53. [PMID: 24453341 DOI: 10.1523/jneurosci.2341-13.2014] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
A major challenge in the neuroscience field is the identification of molecules and pathways that control synaptic plasticity and memory. Dendritic spines play a pivotal role in these processes, as the major sites of excitatory synapses in neuronal communication. Previous studies have shown that the scaffold protein p140Cap localizes into dendritic spines and that its knockdown negatively modulates spine shape in culture. However, so far, there is no information on its in vivo relevance. By using a knock-out mouse model, we here demonstrate that p140Cap is a key element for both learning and synaptic plasticity. Indeed, p140Cap(-/-) mice are impaired in object recognition test, as well as in LTP and in LTD measurements. The in vivo effects of p140Cap loss are presumably attenuated by noncell-autonomous events, since primary neurons obtained from p140Cap(-/-) mice show a strong reduction in number of mushroom spines and abnormal organization of synapse-associated F-actin. These phenotypes are most likely caused by a local reduction of the inhibitory control of RhoA and of cortactin toward the actin-depolymerizing factor cofilin. These events can be controlled by p140Cap through its capability to directly inhibit the activation of Src kinase and by its binding to the scaffold protein Citron-N. Altogether, our results provide new insight into how protein associated with dynamic microtubules may regulate spine actin organization through interaction with postsynaptic density components.
Collapse
|
31
|
Herrera-Molina R, Sarto-Jackson I, Montenegro-Venegas C, Heine M, Smalla KH, Seidenbecher CI, Beesley PW, Gundelfinger ED, Montag D. Structure of excitatory synapses and GABAA receptor localization at inhibitory synapses are regulated by neuroplastin-65. J Biol Chem 2014; 289:8973-88. [PMID: 24554721 DOI: 10.1074/jbc.m113.514992] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Formation, maintenance, and activity of excitatory and inhibitory synapses are essential for neuronal network function. Cell adhesion molecules (CAMs) are crucially involved in these processes. The CAM neuroplastin-65 (Np65) highly expressed during periods of synapse formation and stabilization is present at the pre- and postsynaptic membranes. Np65 can translocate into synapses in response to electrical stimulation and it interacts with subtypes of GABAA receptors in inhibitory synapses. Here, we report that in the murine hippocampus and in hippocampal primary culture, neurons of the CA1 region and the dentate gyrus (DG) express high Np65 levels, whereas expression in CA3 neurons is lower. In neuroplastin-deficient (Np(-/-)) mice the number of excitatory synapses in CA1 and DG, but not CA3 regions is reduced. Notably this picture is mirrored in mature Np(-/-) hippocampal cultures or in mature CA1 and DG wild-type (Np(+/+)) neurons treated with a function-blocking recombinant Np65-Fc extracellular fragment. Although the number of GABAergic synapses was unchanged in Np(-/-) neurons or in mature Np65-Fc-treated Np(+/+) neurons, the ratio of excitatory to inhibitory synapses was significantly lower in Np(-/-) cultures. Furthermore, GABAA receptor composition was altered at inhibitory synapses in Np(-/-) neurons as the α1 to α2 GABAA receptor subunit ratio was increased. Changes of excitatory and inhibitory synaptic function in Np(-/-) neurons were confirmed evaluating the presynaptic release function and using patch clamp recording. These data demonstrate that Np65 is an important regulator of the number and function of synapses in the hippocampus.
Collapse
|
32
|
Arroyo AI, Camoletto PG, Morando L, Sassoe-Pognetto M, Giustetto M, Van Veldhoven PP, Schuchman EH, Ledesma MD. Pharmacological reversion of sphingomyelin-induced dendritic spine anomalies in a Niemann Pick disease type A mouse model. EMBO Mol Med 2014; 6:398-413. [PMID: 24448491 PMCID: PMC3958313 DOI: 10.1002/emmm.201302649] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Understanding the role of lipids in synapses and the aberrant molecular mechanisms causing the cognitive deficits that characterize most lipidosis is necessary to develop therapies for these diseases. Here we describe sphingomyelin (SM) as a key modulator of the dendritic spine actin cytoskeleton. We show that increased SM levels in neurons of acid sphingomyelinase knock out mice (ASMko), which mimic Niemann Pick disease type A (NPA), result in reduced spine number and size and low levels of filamentous actin. Mechanistically, SM accumulation decreases the levels of metabotropic glutamate receptors type I (mGluR1/5) at the synaptic membrane impairing membrane attachment and activity of RhoA and its effectors ROCK and ProfilinIIa. Pharmacological enhancement of the neutral sphingomyelinase rescues the aberrant molecular and morphological phenotypes in vitro and in vivo and improves motor and memory deficits in ASMko mice. Altogether, these data demonstrate the influence of SM and its catabolic enzymes in dendritic spine physiology and contribute to our understanding of the cognitive deficits of NPA patients, opening new perspectives for therapeutic interventions. Subject Categories Genetics, Gene Therapy & Genetic Disease; Neuroscience
Collapse
Affiliation(s)
- Ana I Arroyo
- Department of Neurobiology, Centro Biologia Molecular Severo Ochoa CSIC-UAM, Madrid, Spain
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Caspase-2 is required for dendritic spine and behavioural alterations in J20 APP transgenic mice. Nat Commun 2013; 4:1939. [PMID: 23748737 PMCID: PMC4398315 DOI: 10.1038/ncomms2927] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Accepted: 04/24/2013] [Indexed: 11/09/2022] Open
Abstract
Caspases have critical roles in Alzheimer's disease pathogenesis. Here we show that caspase-2 is required for the cognitive decline seen in human amyloid precursor protein transgenic mice (J20). The age-related changes in behaviour and dendritic spine density observed in these mice are absent when they lack caspase-2, in spite of similar levels of amyloid beta (Aβ) deposition and inflammation. A similar degree of protection is observed in cultured hippocampal neurons lacking caspase-2, which are immune to the synaptotoxic effects of Aβ. Our studies suggest that caspase-2 is a critical mediator in the activation of the RhoA/ROCK-II signalling pathway, leading to the collapse of dendritic spines. We propose that this is controlled by an inactive caspase-2/RhoA/ROCK-II complex localized in dendrites, which dissociates in the presence of Aβ, allowing for their activation and entry in the spine. These findings directly implicate caspase-2 as key driver of synaptic dysfunction in Alzheimer's disease and offer novel therapeutic targets.
Collapse
|
34
|
MAP1B-dependent Rac activation is required for AMPA receptor endocytosis during long-term depression. EMBO J 2013; 32:2287-99. [PMID: 23881099 DOI: 10.1038/emboj.2013.166] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Accepted: 07/02/2013] [Indexed: 02/07/2023] Open
Abstract
The microtubule-associated protein 1B (MAP1B) plays critical roles in neurite growth and synapse maturation during brain development. This protein is well expressed in the adult brain. However, its function in mature neurons remains unknown. We have used a genetically modified mouse model and shRNA techniques to assess the role of MAP1B at established synapses, bypassing MAP1B functions during neuronal development. Under these conditions, we found that MAP1B deficiency alters synaptic plasticity by specifically impairing long-term depression (LTD) expression. Interestingly, this is due to a failure to trigger AMPA receptor endocytosis and spine shrinkage during LTD. These defects are accompanied by an impaired targeting of the Rac1 activator Tiam1 at synaptic compartments. Accordingly, LTD and AMPA receptor endocytosis are restored in MAP1B-deficient neurons by providing additional Rac1. Therefore, these results indicate that the MAP1B-Tiam1-Rac1 relay is essential for spine structural plasticity and removal of AMPA receptors from synapses during LTD. This work highlights the importance of MAPs as signalling hubs controlling the actin cytoskeleton and receptor trafficking during plasticity in mature neurons.
Collapse
|
35
|
Cho IH, Lee MJ, Kim DH, Kim B, Bae J, Choi KY, Kim SM, Huh YH, Lee KH, Kim CH, Song WK. SPIN90 dephosphorylation is required for cofilin-mediated actin depolymerization in NMDA-stimulated hippocampal neurons. Cell Mol Life Sci 2013; 70:4369-83. [PMID: 23765104 PMCID: PMC3825632 DOI: 10.1007/s00018-013-1391-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 05/11/2013] [Accepted: 05/29/2013] [Indexed: 11/30/2022]
Abstract
Actin plays a fundamental role in the regulation of spine morphology (both shrinkage and enlargement) upon synaptic activation. In particular, actin depolymerization is crucial for the spine shrinkage in NMDAR-mediated synaptic depression. Here, we define the role of SPIN90 phosphorylation/dephosphorylation in regulating actin depolymerization via modulation of cofilin activity. When neurons were treated with NMDA, SPIN90 was dephosphorylated by STEP61 (striatal-enriched protein tyrosine phosphatase) and translocated from the spines to the dendritic shafts. In addition, phosphorylated SPIN90 bound cofilin and then inhibited cofilin activity, suggesting that SPIN90 dephosphorylation is a prerequisite step for releasing cofilin so that cofilin can adequately sever actin filaments into monomeric form. We found that SPIN90 YE, a phosphomimetic mutant, remained in the spines after NMDAR activation where it bound cofilin, thereby effectively preventing actin depolymerization. This led to inhibition of the activity-dependent redistribution of cortactin and drebrin A, as well as of the morphological changes in the spines that underlie synaptic plasticity. These findings indicate that NMDA-induced SPIN90 dephosphorylation and translocation initiates cofilin-mediated actin dynamics and spine shrinkage within dendritic spines, thereby modulating synaptic activity.
Collapse
Affiliation(s)
- In Ha Cho
- Bio Imaging and Cell Dynamics Research Center, Gwangju Institute of Science and Technology, 261 Cheomdan-gwagiro, Buk-Gu, Gwangju, 500-712, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
A role for the tyrosine kinase ACK1 in neurotrophin signaling and neuronal extension and branching. Cell Death Dis 2013; 4:e602. [PMID: 23598414 PMCID: PMC3668633 DOI: 10.1038/cddis.2013.99] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Neurotrophins are involved in many crucial cellular functions, including neurite outgrowth, synapse formation, and plasticity. Although these events have long been known, the molecular determinants underlying neuritogenesis have not been fully characterized. Ack1 (activated Cdc42-associated tyrosine kinase) is a non-receptor tyrosine kinase that is highly expressed in the brain. Here, we demonstrate that Ack1 is a molecular constituent of neurotrophin signaling cascades in neurons and PC12 cells. We report that Ack1 interacts with Trk receptors and becomes tyrosine phosphorylated and its kinase activity is increased in response to neurotrophins. Moreover, our data indicate that Ack1 acts upstream of the Akt and MAPK pathways. We show that Ack1 overexpression induces neuritic outgrowth and promotes branching in neurotrophin-treated neuronal cells, whereas the expression of Ack1 dominant negatives or short-hairpin RNAs counteract neurotrophin-stimulated differentiation. Our results identify Ack1 as a novel regulator of neurotrophin-mediated events in primary neurons and in PC12 cells.
Collapse
|
37
|
Abl2/Arg controls dendritic spine and dendrite arbor stability via distinct cytoskeletal control pathways. J Neurosci 2013; 33:1846-57. [PMID: 23365224 DOI: 10.1523/jneurosci.4284-12.2013] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Rho family GTPases coordinate cytoskeletal rearrangements in neurons, and mutations in their regulators are associated with mental retardation and other neurodevelopmental disorders (Billuart et al., 1998; Kutsche et al., 2000; Newey et al., 2005; Benarroch, 2007). Chromosomal microdeletions encompassing p190RhoGAP or its upstream regulator, the Abl2/Arg tyrosine kinase, have been observed in cases of mental retardation associated with developmental defects (Scarbrough et al., 1988; James et al., 1996; Takano et al., 1997; Chaabouni et al., 2006; Leal et al., 2009). Genetic knock-out of Arg in mice leads to synapse, dendritic spine, and dendrite arbor loss accompanied by behavioral deficits (Moresco et al., 2005; Sfakianos et al., 2007). To elucidate the cell-autonomous mechanisms by which Arg regulates neuronal stability, we knocked down Arg in mouse hippocampal neuronal cultures. We find that Arg knockdown significantly destabilizes dendrite arbors and reduces dendritic spine density by compromising dendritic spine stability. Inhibiting RhoA prevents dendrite arbor loss following Arg knockdown in neurons, but does not block spine loss. Interestingly, Arg-deficient neurons exhibit increased miniature EPSC amplitudes, and their remaining spines exhibit larger heads deficient in the actin stabilizing protein cortactin. Spine destabilization in Arg knockdown neurons is prevented by blocking NMDA receptor-dependent relocalization of cortactin from spines, or by forcing cortactin into spines via fusion to an actin-binding region of Arg. Thus, Arg employs distinct mechanisms to selectively regulate spine and dendrite stability: Arg dampens activity-dependent disruption of cortactin localization to stabilize spines and attenuates Rho activity to stabilize dendrite arbors.
Collapse
|
38
|
Iizuka M, Kimura K, Wang S, Kato K, Amano M, Kaibuchi K, Mizoguchi A. Distinct distribution and localization of Rho-kinase in mouse epithelial, muscle and neural tissues. Cell Struct Funct 2012; 37:155-75. [PMID: 22986902 DOI: 10.1247/csf.12018] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
The small GTP-binding protein Rho plays a crucial role in a wide variety of cellular functions through various effector proteins. Rho-kinase is a key effector protein of Rho, which is composed of two isoforms, ROCK1 and ROCK2. To clarify the site of action of ROCK1 and ROCK2, we performed immunofluorescence and immunoelectron microscopic analyses using isoform-specific antibodies in mouse tissues. In the large and small intestines, ROCK1 immunoreactivity was predominantly identified in epithelial cells, and ROCK2 immunoreactivity was negligible. In these epithelial cells, ROCK1 immunoreactivity was distributed on plasma membranes, while ROCK1 immunogold signals were localized at cell-cell contacts and cell adhesion sites, especially at the adherens junctions at the ultrastructural level. In the bladder epithelium, however, ROCK1 and ROCK2 signals were identified at intermediate filaments, and ROCK2 signals were also observed in nuclei. In the three types of muscular cells-smooth, cardiac, and skeletal muscle cells-ROCK1 and ROCK2 also showed differential distribution. ROCK1 signals were localized at actin filaments, plasma membranes, and vesicles near plasma membranes in smooth muscle cells; at the lysosomes in skeletal muscle cells; and were undetectable in cardiac muscle cells. ROCK2 signals were localized at actin filaments and centrosomes in smooth muscle cells, at intercalated discs in cardiac muscle cells, and at Z-discs and sarcoplasmic reticulum in skeletal muscle cells. In the brain, ROCK1 immunoreactivity was distributed in glia, whereas ROCK2 immunoreactivity was observed in neurons. These results indicate that the two isoforms of Rho-kinase distribute differentially to accomplish their specific functions.
Collapse
Affiliation(s)
- Michiro Iizuka
- Department of Neural Regeneration and Cell Communication, Mie University Graduate School of Medicine, Tsu, Japan
| | | | | | | | | | | | | |
Collapse
|
39
|
Soria Fregozo C, Pérez Vega M. Actin-binding proteins and signalling pathways associated with the formation and maintenance of dendritic spines. NEUROLOGÍA (ENGLISH EDITION) 2012. [DOI: 10.1016/j.nrleng.2011.10.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
40
|
Lontay B, Pál B, Serfőző Z, Kőszeghy Á, Szücs G, Rusznák Z, Erdődi F. Protein phosphatase-1M and Rho-kinase affect exocytosis from cortical synaptosomes and influence neurotransmission at a glutamatergic giant synapse of the rat auditory system. J Neurochem 2012; 123:84-99. [PMID: 22817114 DOI: 10.1111/j.1471-4159.2012.07882.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Protein phosphatase-1M (PP1M, myosin phosphatase) consists of a PP1 catalytic subunit (PP1c) and the myosin phosphatase target subunit-1 (MYPT1). RhoA-activated kinase (ROK) regulates PP1M via inhibitory phosphorylation of MYPT1. Using multidisciplinary approaches, we have studied the roles of PP1M and ROK in neurotransmission. Electron microscopy demonstrated the presence of MYPT1 and ROK in both pre- and post-synaptic terminals. Tautomycetin (TMC), a PP1-specific inhibitor, decreased the depolarization-induced exocytosis from cortical synaptosomes. trans-4-[(1R)-1-aminoethyl]-N-4-pyridinylcyclohexanecarboxamide dihydrochloride, a ROK-specific inhibitor, had the opposite effect. Mass spectrometry analysis identified several MYPT1-bound synaptosomal proteins, of which interactions of synapsin-I, syntaxin-1, calcineurin-A subunit, and Ca(2+) /calmodulin-dependent kinase II with MYPT1 were confirmed. In intact synaptosomes, TMC increased, whereas Y27632 decreased the phosphorylation levels of MYPT1(Thr696) , myosin-II light chain(Ser19) , synapsin-I(Ser9) , and syntaxin-1(Ser14) , indicating that PP1M and ROK influence their phosphorylation status. Confocal microscopy indicated that MYPT1 and ROK are present in the rat ventral cochlear nucleus both pre- and post-synaptically. Analysis of the neurotransmission in an auditory glutamatergic giant synapse demonstrated that PP1M and ROK affect neurotransmission via both pre- and post-synaptic mechanisms. Our data suggest that both PP1M and ROK influence synaptic transmission, but further studies are needed to give a full account of their mechanism of action.
Collapse
Affiliation(s)
- Beáta Lontay
- Department of Medical Chemistry and Cell Biology and Signaling Research Group of the Hungarian Academy of Sciences, Medical and Health Science Center, University of Debrecen, Debrecen, Hungary
| | | | | | | | | | | | | |
Collapse
|
41
|
Murk K, Wittenmayer N, Michaelsen-Preusse K, Dresbach T, Schoenenberger CA, Korte M, Jockusch BM, Rothkegel M. Neuronal profilin isoforms are addressed by different signalling pathways. PLoS One 2012; 7:e34167. [PMID: 22470532 PMCID: PMC3314592 DOI: 10.1371/journal.pone.0034167] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2011] [Accepted: 02/23/2012] [Indexed: 01/29/2023] Open
Abstract
Profilins are prominent regulators of actin dynamics. While most mammalian cells express only one profilin, two isoforms, PFN1 and PFN2a are present in the CNS. To challenge the hypothesis that the expression of two profilin isoforms is linked to the complex shape of neurons and to the activity-dependent structural plasticity, we analysed how PFN1 and PFN2a respond to changes of neuronal activity. Simultaneous labelling of rodent embryonic neurons with isoform-specific monoclonal antibodies revealed both isoforms in the same synapse. Immunoelectron microscopy on brain sections demonstrated both profilins in synapses of the mature rodent cortex, hippocampus and cerebellum. Both isoforms were significantly more abundant in postsynaptic than in presynaptic structures. Immunofluorescence showed PFN2a associated with gephyrin clusters of the postsynaptic active zone in inhibitory synapses of embryonic neurons. When cultures were stimulated in order to change their activity level, active synapses that were identified by the uptake of synaptotagmin antibodies, displayed significantly higher amounts of both isoforms than non-stimulated controls. Specific inhibition of NMDA receptors by the antagonist APV in cultured rat hippocampal neurons resulted in a decrease of PFN2a but left PFN1 unaffected. Stimulation by the brain derived neurotrophic factor (BDNF), on the other hand, led to a significant increase in both synaptic PFN1 and PFN2a. Analogous results were obtained for neuronal nuclei: both isoforms were localized in the same nucleus, and their levels rose significantly in response to KCl stimulation, whereas BDNF caused here a higher increase in PFN1 than in PFN2a. Our results strongly support the notion of an isoform specific role for profilins as regulators of actin dynamics in different signalling pathways, in excitatory as well as in inhibitory synapses. Furthermore, they suggest a functional role for both profilins in neuronal nuclei.
Collapse
Affiliation(s)
- Kai Murk
- Cellular Neurobiology, Zoological Institute, TU Braunschweig, Braunschweig, Germany
| | - Nina Wittenmayer
- Department of Anatomy and Cell Biology, Center of Anatomy, Georg August University Göttingen, Göttingen, Germany
| | | | - Thomas Dresbach
- Department of Anatomy and Cell Biology, Center of Anatomy, Georg August University Göttingen, Göttingen, Germany
| | | | - Martin Korte
- Cellular Neurobiology, Zoological Institute, TU Braunschweig, Braunschweig, Germany
| | | | - Martin Rothkegel
- Cellular Neurobiology, Zoological Institute, TU Braunschweig, Braunschweig, Germany
- * E-mail:
| |
Collapse
|
42
|
Sodero AO, Vriens J, Ghosh D, Stegner D, Brachet A, Pallotto M, Sassoè-Pognetto M, Brouwers JF, Helms JB, Nieswandt B, Voets T, Dotti CG. Cholesterol loss during glutamate-mediated excitotoxicity. EMBO J 2012; 31:1764-73. [PMID: 22343944 DOI: 10.1038/emboj.2012.31] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2011] [Accepted: 01/10/2012] [Indexed: 11/09/2022] Open
Abstract
The deregulation of brain cholesterol metabolism is typical in acute neuronal injury (such as stroke, brain trauma and epileptic seizures) and chronic neurodegenerative diseases (Alzheimer's disease). Since both conditions are characterized by excessive stimulation of glutamate receptors, we have here investigated to which extent excitatory neurotransmission plays a role in brain cholesterol homeostasis. We show that a short (30 min) stimulation of glutamatergic neurotransmission induces a small but significant loss of membrane cholesterol, which is paralleled by release to the extracellular milieu of the metabolite 24S-hydroxycholesterol. Consistent with a cause-effect relationship, knockdown of the enzyme cholesterol 24-hydroxylase (CYP46A1) prevented glutamate-mediated cholesterol loss. Functionally, the loss of cholesterol modulates the magnitude of the depolarization-evoked calcium response. Mechanistically, glutamate-induced cholesterol loss requires high levels of intracellular Ca(2+), a functional stromal interaction molecule 2 (STIM2) and mobilization of CYP46A1 towards the plasma membrane. This study underscores the key role of excitatory neurotransmission in the control of membrane lipid composition, and consequently in neuronal membrane organization and function.
Collapse
Affiliation(s)
- Alejandro O Sodero
- VIB Center for Biology of Disease, Katholieke Universiteit Leuven, Leuven, Belgium
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Membrane-initiated estradiol signaling induces spinogenesis required for female sexual receptivity. J Neurosci 2012; 31:17583-9. [PMID: 22131419 DOI: 10.1523/jneurosci.3030-11.2011] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Estrogens have profound actions on the structure of the nervous system during development and in adulthood. One of the signature actions of estradiol is to alter the morphology of neural processes. In the hippocampus, estradiol modulates spines and cellular excitability that affect cognitive behaviors. In the hypothalamus, estradiol increases spine density in mediobasal hypothalamic nuclei that regulate reproduction. The hypothalamic arcuate nucleus (ARH), an important site for modulation of female sexual receptivity, has a sexual dimorphism in dendritic spine density that favors females. In the present study, we used both β-actin immunostaining and Golgi staining to visualize estradiol-induced changes in spine density in Long-Evans rats. Golgi impregnation was used to visualize spine shape, and then β-actin immunoreactivity was used as a semiquantitative measure of spine plasticity since actin forms the core of dendritic spines. At 4 h after estradiol treatment, both β-actin immunofluorescence and filopodial spines were increased (from 70.57 ± 1.09% to 78.01 ± 1.05%, p < 0.05). Disruption of estradiol-induced β-actin polymerization with cytochalasin D attenuated lordosis behavior, indicating the importance of estradiol-mediated spinogenesis for female sexual receptivity (81.43 ± 7.05 to 35.00 ± 11.76, p < 0.05). Deactivation of cofilin, an actin depolymerizing factor is required for spinogenesis. Membrane-initiated estradiol signaling involving the metabotropic glutamate receptor 1a was responsible for the phosphorylation and thereby deactivation of cofilin. These data demonstrate that estradiol-induced spinogenesis in the ARH is an important cellular mechanism for the regulation of female sexual behavior.
Collapse
|
44
|
Soria Fregozo C, Pérez Vega MI. Actin-binding proteins and signalling pathways associated with the formation and maintenance of dendritic spines. Neurologia 2011; 27:421-31. [PMID: 22178050 DOI: 10.1016/j.nrl.2011.10.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2011] [Accepted: 10/09/2011] [Indexed: 12/20/2022] Open
Abstract
INTRODUCTION Dendritic spines are the main sites of excitatory synaptic contacts. Moreover, they present plastic responses to different stimuli present in synaptic activity or damage, ranging from an increase or decrease in their total number, to redistribution of progenitor dendritic spines, to variations in their size or shape. However, the spines can remain stable for a long time. BACKGROUND The use of experimental models has shown that different molecules of the F-actin binding and signalling pathways are closely related to the development, maintenance and plasticity of excitatory synapses, which could affect the number, size and shape of the dendritic spines; these mechanisms affect and depend on the reorganisation of the actin cytoskeleton. DEVELOPMENT It is proposed that the filopodia are precursors of dendritic spines. Drebrin is an F-actin binding protein, and it is responsible for concentrating F-actin and PSD-95 in filopodia that will guide the formation of the new spines. CONCLUSION The specific mechanisms of actin regulation are an integral part in the formation, maturing process and plasticity of dendritic spines in association with the various actin cytoskeleton-binding proteins The signalling pathways mediated by small GTPases and the equilibrium between G-actin and F-actin are also involved.
Collapse
Affiliation(s)
- C Soria Fregozo
- Laboratorio de Psicobiología y Biología Molecular, Departamento de Ciencias de la Tierra y de la Vida, Centro Universitario de los Lagos, Universidad de Guadalajara, Guadalajara, Mexico.
| | | |
Collapse
|
45
|
Bennett M. Schizophrenia: susceptibility genes, dendritic-spine pathology and gray matter loss. Prog Neurobiol 2011; 95:275-300. [DOI: 10.1016/j.pneurobio.2011.08.003] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2011] [Revised: 08/12/2011] [Accepted: 08/15/2011] [Indexed: 02/01/2023]
|
46
|
Role of GluN2A and GluN2B subunits in the formation of filopodia and secondary dendrites in cultured hippocampal neurons. Naunyn Schmiedebergs Arch Pharmacol 2011; 385:171-80. [DOI: 10.1007/s00210-011-0701-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2011] [Accepted: 10/10/2011] [Indexed: 10/16/2022]
|
47
|
Bakshi K, Kosciuk M, Nagele RG, Friedman E, Wang HY. Prenatal cocaine exposure increases synaptic localization of a neuronal RasGEF, GRASP-1 via hyperphosphorylation of AMPAR anchoring protein, GRIP. PLoS One 2011; 6:e25019. [PMID: 21980374 PMCID: PMC3181332 DOI: 10.1371/journal.pone.0025019] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2011] [Accepted: 08/23/2011] [Indexed: 11/18/2022] Open
Abstract
Prenatal cocaine exposure causes sustained phosphorylation of the synaptic anchoring protein, glutamate receptor interacting protein (GRIP1/2), preventing synaptic targeting of the GluR2/3-containing alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid-type glutamate receptors (AMPARs; J. Neurosci. 29: 6308–6319, 2009). Because overexpression of GRIP-associated neuronal rasGEF protein (GRASP-1) specifically reduces the synaptic targeting of AMPARs, we hypothesized that prenatal cocaine exposure enhances GRASP-1 synaptic membrane localization leading to hyper-activation of ras family proteins and heightened actin polymerization. Our results show a markedly increased GRIP1-associated GRASP-1 content with approximately 40% reduction in its rasGEF activity in frontal cortices (FCX) of 21-day-old (P21) prenatal cocaine-exposed rats. This cocaine effect is the result of a persistent protein kinase C (PKC)- and downstream Src tyrosine kinase-mediated GRIP phosphorylation. The hyperactivated PKC also increased membrane-associated GRASP-1 and activated small G-proteins RhoA, cdc42/Rac1 and Rap1 as well as filamentous actin (F-actin) levels without an effect on the phosphorylation state of actin. Since increased F-actin facilitates protein transport, our results suggest that increased GRASP-1 synaptic localization in prenatal cocaine-exposed brains is an adaptive response to restoring the synaptic expression of AMPA-GluR2/3. Our earlier data demonstrated that persistent PKC-mediated GRIP phosphorylation reduces GluR2/3 synaptic targeting in prenatal cocaine-exposed brains, we now show that the increased GRIP-associated GRASP-1 may contribute to the reduction in GluR2/3 synaptic expression and AMPAR signaling defects.
Collapse
Affiliation(s)
- Kalindi Bakshi
- Departments of Physiology, Pharmacology and Neuroscience, Sophie Davis School of Biomedical Education, The City University of New York Medical School, New York, New York, United States of America
- Department of Biology & Neuroscience, Graduate Center of the City University of New York, New York, New York, United States of America
- Center for Developmental Neuroscience/Institute for Basic Research/City University of New York Graduate School, Staten Island, New York, United States of America
| | - Mary Kosciuk
- New Jersey Institute for Successful Aging, University of Medicine and Dentistry New Jersey-School of Osteopathic Medicine, Stratford, New Jersey, United States of America
| | - Robert G. Nagele
- New Jersey Institute for Successful Aging, University of Medicine and Dentistry New Jersey-School of Osteopathic Medicine, Stratford, New Jersey, United States of America
| | - Eitan Friedman
- Departments of Physiology, Pharmacology and Neuroscience, Sophie Davis School of Biomedical Education, The City University of New York Medical School, New York, New York, United States of America
- Department of Biology & Neuroscience, Graduate Center of the City University of New York, New York, New York, United States of America
| | - Hoau-Yan Wang
- Departments of Physiology, Pharmacology and Neuroscience, Sophie Davis School of Biomedical Education, The City University of New York Medical School, New York, New York, United States of America
- Department of Biology & Neuroscience, Graduate Center of the City University of New York, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
48
|
Lamprecht R. The roles of the actin cytoskeleton in fear memory formation. Front Behav Neurosci 2011; 5:39. [PMID: 21808614 PMCID: PMC3139223 DOI: 10.3389/fnbeh.2011.00039] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2011] [Accepted: 07/02/2011] [Indexed: 01/08/2023] Open
Abstract
The formation and storage of fear memory is needed to adapt behavior and avoid danger during subsequent fearful events. However, fear memory may also play a significant role in stress and anxiety disorders. When fear becomes disproportionate to that necessary to cope with a given stimulus, or begins to occur in inappropriate situations, a fear or anxiety disorder exists. Thus, the study of cellular and molecular mechanisms underpinning fear memory may shed light on the formation of memory and on anxiety and stress related disorders. Evidence indicates that fear learning leads to changes in neuronal synaptic transmission and morphology in brain areas underlying fear memory formation including the amygdala and hippocampus. The actin cytoskeleton has been shown to participate in these key neuronal processes. Recent findings show that the actin cytoskeleton is needed for fear memory formation and extinction. Moreover, the actin cytoskeleton is involved in synaptic plasticity and in neuronal morphogenesis in brain areas that mediate fear memory. The actin cytoskeleton may therefore mediate between synaptic transmission during fear learning and long-term cellular alterations mandatory for fear memory formation.
Collapse
Affiliation(s)
- Raphael Lamprecht
- Faculty of Natural Sciences, Department of Neurobiology and Ethology, University of Haifa Haifa, Israel
| |
Collapse
|
49
|
A network of networks: cytoskeletal control of compartmentalized function within dendritic spines. Curr Opin Neurobiol 2011; 20:578-87. [PMID: 20667710 DOI: 10.1016/j.conb.2010.06.009] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2010] [Revised: 06/04/2010] [Accepted: 06/26/2010] [Indexed: 12/19/2022]
Abstract
Almost 30 years ago, actin was identified as the major cytoskeletal component of dendritic spines. Since then, its role in the remarkable dynamics of spine morphology have been detailed with live-cell views establishing that spine shape dynamics are an important requirement for synaptogenesis and synaptic plasticity. However, the actin cytoskeleton is critical to numerous and varied processes within the spine which contribute to the maintenance and plasticity of synaptic function. Here, we argue that the spatial and temporal distribution of actin-dependent processes within spines suggests that the spine cytoskeleton should not be considered a single entity, but an interacting network of nodes or hubs that are independently regulated and balanced to maintain synapse function. Disruptions of this balance within the spine are likely to lead to psychiatric and neurological dysfunction.
Collapse
|
50
|
Lin YC, Koleske AJ. Mechanisms of synapse and dendrite maintenance and their disruption in psychiatric and neurodegenerative disorders. Annu Rev Neurosci 2011; 33:349-78. [PMID: 20367247 DOI: 10.1146/annurev-neuro-060909-153204] [Citation(s) in RCA: 188] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Emerging evidence indicates that once established, synapses and dendrites can be maintained for long periods, if not for the organism's entire lifetime. In contrast to the wealth of knowledge regarding axon, dendrite, and synapse development, we understand comparatively little about the cellular and molecular mechanisms that enable long-term synapse and dendrite maintenance. Here, we review how the actin cytoskeleton and its regulators, adhesion receptors, and scaffolding proteins mediate synapse and dendrite maintenance. We examine how these mechanisms are reinforced by trophic signals passed between the pre- and postsynaptic compartments. We also discuss how synapse and dendrite maintenance mechanisms are compromised in psychiatric and neurodegenerative disorders.
Collapse
Affiliation(s)
- Yu-Chih Lin
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut 06520-8024, USA.
| | | |
Collapse
|