1
|
Teoh J, Bartolini F. Emerging roles for tubulin PTMs in neuronal function and neurodegenerative disease. Curr Opin Neurobiol 2025; 90:102971. [PMID: 39862522 PMCID: PMC11839326 DOI: 10.1016/j.conb.2025.102971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 12/10/2024] [Accepted: 01/02/2025] [Indexed: 01/27/2025]
Abstract
Neurons are equipped with microtubules of different stability with stable and dynamic domains often coexisting on the same microtubule. While dynamic microtubules undergo random transitions between disassembly and assembly, stable ones persist long enough to serve as platforms for tubulin-modifying enzymes (known as writers) that attach molecular components to the α- or β-tubulin subunits. The combination of these posttranslational modifications (PTMs) results in a "tubulin code," dictating the behavior of selected proteins (known as readers), some of which were shown to be crucial for neuronal function. Recent research has further highlighted that disturbances in tubulin PTMs can lead to neurodegeneration, sparking an emerging field of investigation with numerous questions such as whether and how tubulin PTMs can affect neurotransmission and synaptic plasticity and whether restoring balanced tubulin PTM levels could effectively prevent or mitigate neurodegenerative disease.
Collapse
Affiliation(s)
- JiaJie Teoh
- Department of Pathology & Cell Biology, Columbia University Irving Medical Center, 10032, New York, NY, USA
| | - Francesca Bartolini
- Department of Pathology & Cell Biology, Columbia University Irving Medical Center, 10032, New York, NY, USA.
| |
Collapse
|
2
|
Sun SY, Nie L, Zhang J, Fang X, Luo H, Fu C, Wei Z, Tang AH. The interaction between KIF21A and KANK1 regulates dendritic morphology and synapse plasticity in neurons. Neural Regen Res 2025; 20:209-223. [PMID: 38767486 PMCID: PMC11246154 DOI: 10.4103/1673-5374.391301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 07/12/2023] [Accepted: 11/07/2023] [Indexed: 05/22/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202501000-00029/figure1/v/2024-05-14T021156Z/r/image-tiff Morphological alterations in dendritic spines have been linked to changes in functional communication between neurons that affect learning and memory. Kinesin-4 KIF21A helps organize the microtubule-actin network at the cell cortex by interacting with KANK1; however, whether KIF21A modulates dendritic structure and function in neurons remains unknown. In this study, we found that KIF21A was distributed in a subset of dendritic spines, and that these KIF21A-positive spines were larger and more structurally plastic than KIF21A-negative spines. Furthermore, the interaction between KIF21A and KANK1 was found to be critical for dendritic spine morphogenesis and synaptic plasticity. Knockdown of either KIF21A or KANK1 inhibited dendritic spine morphogenesis and dendritic branching, and these deficits were fully rescued by coexpressing full-length KIF21A or KANK1, but not by proteins with mutations disrupting direct binding between KIF21A and KANK1 or binding between KANK1 and talin1. Knocking down KIF21A in the hippocampus of rats inhibited the amplitudes of long-term potentiation induced by high-frequency stimulation and negatively impacted the animals' cognitive abilities. Taken together, our findings demonstrate the function of KIF21A in modulating spine morphology and provide insight into its role in synaptic function.
Collapse
Affiliation(s)
- Shi-Yan Sun
- Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, Ministry of Education Key Laboratory for Membrane-less Organelles and Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, Anhui Province, China
| | - Lingyun Nie
- Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, Ministry of Education Key Laboratory for Membrane-less Organelles and Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
- CAS Center for Excellence in Molecular Cell Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
| | - Jing Zhang
- Department of Neurobiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong Province, China
- Brain Research Center, Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Xue Fang
- Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, Ministry of Education Key Laboratory for Membrane-less Organelles and Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
| | - Hongmei Luo
- Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, Ministry of Education Key Laboratory for Membrane-less Organelles and Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, Anhui Province, China
| | - Chuanhai Fu
- Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, Ministry of Education Key Laboratory for Membrane-less Organelles and Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
- CAS Center for Excellence in Molecular Cell Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
| | - Zhiyi Wei
- Department of Neurobiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong Province, China
- Brain Research Center, Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Ai-Hui Tang
- Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, Ministry of Education Key Laboratory for Membrane-less Organelles and Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, Anhui Province, China
| |
Collapse
|
3
|
Jang EH, Choi H, Hur EM. Microtubule function and dysfunction in the nervous system. Mol Cells 2024; 47:100111. [PMID: 39265797 PMCID: PMC11474369 DOI: 10.1016/j.mocell.2024.100111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 09/05/2024] [Accepted: 09/06/2024] [Indexed: 09/14/2024] Open
Abstract
Microtubules are core components of the neuronal cytoskeleton, providing structural support for the complex cytoarchitecture of neurons and serving as tracks for long-distance transport. The properties and functions of neuronal microtubules are controlled by tubulin isoforms and a variety of post-translational modifications, collectively known as the "tubulin code." The tubulin code exerts direct control over the intrinsic properties of neuronal microtubules and regulates the repertoire of proteins that read the code, which in turn, has a significant impact on microtubule stability and dynamics. Here, we review progress in the understanding of the tubulin code in the nervous system, with a particular focus on tubulin post-translational modifications that have been proposed as potential contributors to the development and maintenance of the mammalian nervous system. Furthermore, we also discuss the potential links between disruptions in the tubulin code and neurological disorders, including neurodevelopmental abnormalities and neurodegenerative diseases.
Collapse
Affiliation(s)
- Eun-Hae Jang
- Laboratory of Neuroscience, College of Veterinary Medicine, Seoul National University, Seoul 08826, South Korea; Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, South Korea; Comparative Medicine Disease Research Center, Seoul National University, Seoul, South Korea
| | - Harryn Choi
- Laboratory of Neuroscience, College of Veterinary Medicine, Seoul National University, Seoul 08826, South Korea; Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, South Korea; BK21 Four Future Veterinary Medicine Leading Education & Research Center, College of Veterinary Medicine, Seoul National University, Seoul 08826, South Korea
| | - Eun-Mi Hur
- Laboratory of Neuroscience, College of Veterinary Medicine, Seoul National University, Seoul 08826, South Korea; Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, South Korea; Comparative Medicine Disease Research Center, Seoul National University, Seoul, South Korea; BK21 Four Future Veterinary Medicine Leading Education & Research Center, College of Veterinary Medicine, Seoul National University, Seoul 08826, South Korea; Interdisciplinary Program in Neuroscience, College of Natural Sciences, Seoul National University, Seoul 08826, South Korea.
| |
Collapse
|
4
|
Fok AHK, Huang Y, So BWL, Zheng Q, Tse CSC, Li X, Wong KKY, Huang J, Lai KO, Lai CSW. KIF5B plays important roles in dendritic spine plasticity and dendritic localization of PSD95 and FMRP in the mouse cortex in vivo. Cell Rep 2024; 43:113906. [PMID: 38451812 DOI: 10.1016/j.celrep.2024.113906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 12/21/2023] [Accepted: 02/16/2024] [Indexed: 03/09/2024] Open
Abstract
Kinesin 1 (KIF5) is one major type of motor protein in neurons, but its members' function in the intact brain remains less studied. Using in vivo two-photon imaging, we find that conditional knockout of Kif5b (KIF5B cKO) in CaMKIIα-Cre-expressing neurons shows heightened turnover and lower stability of dendritic spines in layer 2/3 pyramidal neurons with reduced spine postsynaptic density protein 95 acquisition in the mouse cortex. Furthermore, the RNA-binding protein fragile X mental retardation protein (FMRP) is translocated to the proximity of newly formed spines several hours before the spine formation events in vivo in control mice, but this preceding transport of FMRP is abolished in KIF5B cKO mice. We further find that FMRP is localized closer to newly formed spines after fear extinction, but this learning-dependent localization is disrupted in KIF5B cKO mice. Our findings provide the crucial in vivo evidence that KIF5B is involved in the dendritic targeting of synaptic proteins that underlies dendritic spine plasticity.
Collapse
Affiliation(s)
- Albert Hiu Ka Fok
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Yuhua Huang
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China; Advanced Biomedical Instrumentation Centre, Hong Kong Science Park, Shatin, New Territories, Hong Kong SAR, China
| | - Beth Wing Lam So
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Qiyu Zheng
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Chun Sing Carlos Tse
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Xiaoyang Li
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Kenneth Kin-Yip Wong
- Advanced Biomedical Instrumentation Centre, Hong Kong Science Park, Shatin, New Territories, Hong Kong SAR, China; Department of Electrical and Electronic Engineering, The University of Hong Kong, Hong Kong SAR, China
| | - Jiandong Huang
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China; Chinese Academy of Sciences (CAS) Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; Clinical Oncology Center, Shenzhen Key Laboratory for Cancer Metastasis and Personalized Therapy, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China; Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen University, Guangzhou, China; State Key Laboratory of Cognitive and Brain Research, The University of Hong Kong, Hong Kong SAR, China
| | - Kwok-On Lai
- Department of Neuroscience, City University of Hong Kong, Hong Kong SAR, China; Hong Kong Institute for Advanced Study, City University of Hong Kong, Hong Kong SAR, China.
| | - Cora Sau Wan Lai
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China; State Key Laboratory of Cognitive and Brain Research, The University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
5
|
McKenna ED, Sarbanes SL, Cummings SW, Roll-Mecak A. The Tubulin Code, from Molecules to Health and Disease. Annu Rev Cell Dev Biol 2023; 39:331-361. [PMID: 37843925 DOI: 10.1146/annurev-cellbio-030123-032748] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2023]
Abstract
Microtubules are essential dynamic polymers composed of α/β-tubulin heterodimers. They support intracellular trafficking, cell division, cellular motility, and other essential cellular processes. In many species, both α-tubulin and β-tubulin are encoded by multiple genes with distinct expression profiles and functionality. Microtubules are further diversified through abundant posttranslational modifications, which are added and removed by a suite of enzymes to form complex, stereotyped cellular arrays. The genetic and chemical diversity of tubulin constitute a tubulin code that regulates intrinsic microtubule properties and is read by cellular effectors, such as molecular motors and microtubule-associated proteins, to provide spatial and temporal specificity to microtubules in cells. In this review, we synthesize the rapidly expanding tubulin code literature and highlight limitations and opportunities for the field. As complex microtubule arrays underlie essential physiological processes, a better understanding of how cells employ the tubulin code has important implications for human disease ranging from cancer to neurological disorders.
Collapse
Affiliation(s)
- Elizabeth D McKenna
- Cell Biology and Biophysics Unit, Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, USA;
| | - Stephanie L Sarbanes
- Cell Biology and Biophysics Unit, Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, USA;
| | - Steven W Cummings
- Cell Biology and Biophysics Unit, Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, USA;
| | - Antonina Roll-Mecak
- Cell Biology and Biophysics Unit, Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, USA;
- Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA
| |
Collapse
|
6
|
Pero ME, Chowdhury F, Bartolini F. Role of tubulin post-translational modifications in peripheral neuropathy. Exp Neurol 2023; 360:114274. [PMID: 36379274 PMCID: PMC11320756 DOI: 10.1016/j.expneurol.2022.114274] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 11/06/2022] [Accepted: 11/08/2022] [Indexed: 11/14/2022]
Abstract
Peripheral neuropathy is a common disorder that results from nerve damage in the periphery. The degeneration of sensory axon terminals leads to changes or loss of sensory functions, often manifesting as debilitating pain, weakness, numbness, tingling, and disability. The pathogenesis of most peripheral neuropathies remains to be fully elucidated. Cumulative evidence from both early and recent studies indicates that tubulin damage may provide a common underlying mechanism of axonal injury in various peripheral neuropathies. In particular, tubulin post-translational modifications have been recently implicated in both toxic and inherited forms of peripheral neuropathy through regulation of axonal transport and mitochondria dynamics. This knowledge forms a new area of investigation with the potential for developing therapeutic strategies to prevent or delay peripheral neuropathy by restoring tubulin homeostasis.
Collapse
Affiliation(s)
- Maria Elena Pero
- Department of Pathology and Cell Biology, Columbia University, New York, USA; Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Italy
| | - Farihah Chowdhury
- Department of Pathology and Cell Biology, Columbia University, New York, USA
| | - Francesca Bartolini
- Department of Pathology and Cell Biology, Columbia University, New York, USA.
| |
Collapse
|
7
|
Jia S, Rybalchenko N, Kunwar K, Farmer GE, Little JT, Toney GM, Cunningham JT. Chronic intermittent hypoxia enhances glycinergic inhibition in nucleus tractus solitarius. J Neurophysiol 2022; 128:1383-1394. [PMID: 36321700 PMCID: PMC9678432 DOI: 10.1152/jn.00241.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Chronic intermittent hypoxia (CIH), an animal model of sleep apnea, has been shown to alter the activity of second-order chemoreceptor neurons in the caudal nucleus of the solitary tract (cNTS). Although numerous studies have focused on excitatory plasticity, few studies have explored CIH-induced plasticity impacting inhibitory inputs to NTS neurons, and the roles of GABAergic and glycinergic inputs on heightened cNTS excitability following CIH are unknown. In addition, changes in astrocyte function may play a role in cNTS plasticity responses to CIH. This study tested the effects of a 7-day CIH protocol on miniature inhibitory postsynaptic currents (mIPSCs) in cNTS neurons receiving chemoreceptor afferents. Normoxia-treated rats primarily displayed GABA mIPSCs, whereas CIH-treated rats exhibited a shift toward combined GABA/glycine-mediated mIPSCs. CIH increased glycinergic mIPSC amplitude and area. This shift was not observed in dorsal motor nucleus of the vagus neurons or cNTS cells from females. Immunohistochemistry showed that strengthened glycinergic mIPSCs were associated with increased glycine receptor protein and were dependent on receptor trafficking in CIH-treated rats. In addition, CIH altered astrocyte morphology in the cNTS, and inactivation of astrocytes following CIH reduced glycine receptor-mediated mIPSC frequency and overall mIPSC amplitude. In cNTS, CIH produced changes in glycine signaling that appear to reflect increased trafficking of glycine receptors to the cell membrane. Increased glycine signaling in cNTS associated with CIH also appears to be dependent on astrocytes. Additional studies will be needed to determine how CIH influences glycine receptor expression and astrocyte function in cNTS.NEW & NOTEWORTHY Chronic intermittent hypoxia (CIH) has been used to mimic the hypoxemia associated with sleep apnea and determine how these hypoxemias influence neural function. The nucleus of the solitary tract is the main site for chemoreceptor input to the CNS, but how CIH influences NTS inhibition has not been determined. These studies show that CIH increases glycine-mediated miniature IPSCs through mechanisms that depend on protein trafficking and astrocyte activation.
Collapse
Affiliation(s)
- Shuping Jia
- 1Department of Physiology and Anatomy, University of Texas Health Science Center, Fort Worth, Texas
| | - Nataliya Rybalchenko
- 1Department of Physiology and Anatomy, University of Texas Health Science Center, Fort Worth, Texas
| | - Kishor Kunwar
- 2Microscopy Core, Division of Research and Innovation, University of Texas Health Science Center, Fort Worth, Texas
| | - George E. Farmer
- 1Department of Physiology and Anatomy, University of Texas Health Science Center, Fort Worth, Texas
| | - Joel T. Little
- 1Department of Physiology and Anatomy, University of Texas Health Science Center, Fort Worth, Texas
| | - Glenn M. Toney
- 3Department of Cellular & Integrative Physiology, University of Texas Health San Antonio, San Antonio, Texas
| | - J. Thomas Cunningham
- 1Department of Physiology and Anatomy, University of Texas Health Science Center, Fort Worth, Texas
| |
Collapse
|
8
|
Radwitz J, Hausrat TJ, Heisler FF, Janiesch PC, Pechmann Y, Rübhausen M, Kneussel M. Tubb3 expression levels are sensitive to neuronal activity changes and determine microtubule growth and kinesin-mediated transport. Cell Mol Life Sci 2022; 79:575. [DOI: 10.1007/s00018-022-04607-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 10/11/2022] [Accepted: 10/11/2022] [Indexed: 11/03/2022]
Abstract
AbstractMicrotubules are dynamic polymers of α/β-tubulin. They regulate cell structure, cell division, cell migration, and intracellular transport. However, functional contributions of individual tubulin isotypes are incompletely understood. The neuron-specific β-tubulin Tubb3 displays highest expression around early postnatal periods characterized by exuberant synaptogenesis. Although Tubb3 mutations are associated with neuronal disease, including abnormal inhibitory transmission and seizure activity in patients, molecular consequences of altered Tubb3 levels are largely unknown. Likewise, it is unclear whether neuronal activity triggers Tubb3 expression changes in neurons. In this study, we initially asked whether chemical protocols to induce long-term potentiation (cLTP) affect microtubule growth and the expression of individual tubulin isotypes. We found that growing microtubules and Tubb3 expression are sensitive to changes in neuronal activity and asked for consequences of Tubb3 downregulation in neurons. Our data revealed that reduced Tubb3 levels accelerated microtubule growth in axons and dendrites. Remarkably, Tubb3 knockdown induced a specific upregulation of Tubb4 gene expression, without changing other tubulin isotypes. We further found that Tubb3 downregulation reduces tubulin polyglutamylation, increases KIF5C motility and boosts the transport of its synaptic cargo N-Cadherin, which is known to regulate synaptogenesis and long-term potentiation. Due to the large number of tubulin isotypes, we developed and applied a computational model based on a Monte Carlo simulation to understand consequences of tubulin expression changes in silico. Together, our data suggest a feedback mechanism with neuronal activity regulating tubulin expression and consequently microtubule dynamics underlying the delivery of synaptic cargoes.
Collapse
|
9
|
Disruption of tubulin-alpha4a polyglutamylation prevents aggregation of hyper-phosphorylated tau and microglia activation in mice. Nat Commun 2022; 13:4192. [PMID: 35858909 PMCID: PMC9300677 DOI: 10.1038/s41467-022-31776-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 06/30/2022] [Indexed: 11/14/2022] Open
Abstract
Dissociation of hyper-phosphorylated Tau from neuronal microtubules and its pathological aggregates, are hallmarks in the etiology of tauopathies. The Tau-microtubule interface is subject to polyglutamylation, a reversible posttranslational modification, increasing negative charge at tubulin C-terminal tails. Here, we asked whether tubulin polyglutamylation may contribute to Tau pathology in vivo. Since polyglutamylases modify various proteins other than tubulin, we generated a knock-in mouse carrying gene mutations to abolish Tuba4a polyglutamylation in a substrate-specific manner. We found that Tuba4a lacking C-terminal polyglutamylation prevents the binding of Tau and GSK3 kinase to neuronal microtubules, thereby strongly reducing phospho-Tau levels. Notably, crossbreeding of the Tuba4a knock-in mouse with the hTau tauopathy model, expressing a human Tau transgene, reversed hyper-phosphorylation and oligomerization of Tau and normalized microglia activation in brain. Our data highlight tubulin polyglutamylation as a potential therapeutic strategy in fighting tauopathies. Pathologic oligomerization of hyper-phosphorylated Tau is a hallmark of tauopathies. Here the authors show that the loss of tubulin a4 polyglutamylation reverses tau hyperphosphorylation, oligomerization and microglia activation in a tauopathy mouse.
Collapse
|
10
|
Bai G, Zhang M. Inhibitory postsynaptic density from the lens of phase separation. OXFORD OPEN NEUROSCIENCE 2022; 1:kvac003. [PMID: 38596704 PMCID: PMC10913824 DOI: 10.1093/oons/kvac003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/22/2022] [Accepted: 02/22/2022] [Indexed: 04/11/2024]
Abstract
To faithfully transmit and decode signals released from presynaptic termini, postsynaptic compartments of neuronal synapses deploy hundreds of various proteins. In addition to distinct sets of proteins, excitatory and inhibitory postsynaptic apparatuses display very different organization features and regulatory properties. Decades of extensive studies have generated a wealth of knowledge on the molecular composition, assembly architecture and activity-dependent regulatory mechanisms of excitatory postsynaptic compartments. In comparison, our understanding of the inhibitory postsynaptic apparatus trails behind. Recent studies have demonstrated that phase separation is a new paradigm underlying the formation and plasticity of both excitatory and inhibitory postsynaptic molecular assemblies. In this review, we discuss molecular composition, organizational and regulatory features of inhibitory postsynaptic densities through the lens of the phase separation concept and in comparison with the excitatory postsynaptic densities.
Collapse
Affiliation(s)
- Guanhua Bai
- School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Mingjie Zhang
- School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
- Greater Bay Biomedical Innocenter, Shenzhen Bay Laboratory, Shenzhen 518036, China
| |
Collapse
|
11
|
Peña-Ortega F, Robles-Gómez ÁA, Xolalpa-Cueva L. Microtubules as Regulators of Neural Network Shape and Function: Focus on Excitability, Plasticity and Memory. Cells 2022; 11:cells11060923. [PMID: 35326374 PMCID: PMC8946818 DOI: 10.3390/cells11060923] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 02/09/2022] [Accepted: 02/17/2022] [Indexed: 12/19/2022] Open
Abstract
Neuronal microtubules (MTs) are complex cytoskeletal protein arrays that undergo activity-dependent changes in their structure and function as a response to physiological demands throughout the lifespan of neurons. Many factors shape the allostatic dynamics of MTs and tubulin dimers in the cytosolic microenvironment, such as protein–protein interactions and activity-dependent shifts in these interactions that are responsible for their plastic capabilities. Recently, several findings have reinforced the role of MTs in behavioral and cognitive processes in normal and pathological conditions. In this review, we summarize the bidirectional relationships between MTs dynamics, neuronal processes, and brain and behavioral states. The outcomes of manipulating the dynamicity of MTs by genetic or pharmacological approaches on neuronal morphology, intrinsic and synaptic excitability, the state of the network, and behaviors are heterogeneous. We discuss the critical position of MTs as responders and adaptative elements of basic neuronal function whose impact on brain function is not fully understood, and we highlight the dilemma of artificially modulating MT dynamics for therapeutic purposes.
Collapse
|
12
|
Fan R, Lai KO. Understanding how kinesin motor proteins regulate postsynaptic function in neuron. FEBS J 2021; 289:2128-2144. [PMID: 34796656 DOI: 10.1111/febs.16285] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 11/08/2021] [Accepted: 11/17/2021] [Indexed: 01/07/2023]
Abstract
The Kinesin superfamily proteins (KIFs) are major molecular motors that transport diverse set of cargoes along microtubules to both the axon and dendrite of a neuron. Much of our knowledge about kinesin function is obtained from studies on axonal transport. Emerging evidence reveals how specific kinesin motor proteins carry cargoes to dendrites, including proteins, mRNAs and organelles that are crucial for synapse development and plasticity. In this review, we will summarize the major kinesin motors and their associated cargoes that have been characterized to regulate postsynaptic function in neuron. We will also discuss how specific kinesins are selectively involved in the development of excitatory and inhibitory postsynaptic compartments, their regulation by post-translational modifications (PTM), as well as their roles beyond conventional transport carrier.
Collapse
Affiliation(s)
- Ruolin Fan
- Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
| | - Kwok-On Lai
- Department of Neuroscience, City University of Hong Kong, Hong Kong, China
| |
Collapse
|
13
|
Zhu S, Yang BS, Li SJ, Tong G, Tan JY, Wu GF, Li L, Chen GL, Chen Q, Lin LJ. Protein post-translational modifications after spinal cord injury. Neural Regen Res 2021; 16:1935-1943. [PMID: 33642363 PMCID: PMC8343325 DOI: 10.4103/1673-5374.308068] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 09/11/2020] [Accepted: 11/22/2020] [Indexed: 11/04/2022] Open
Abstract
Deficits in intrinsic neuronal capacities in the spinal cord, a lack of growth support, and suppression of axonal outgrowth by inhibitory molecules mean that spinal cord injury almost always has devastating consequences. As such, one of the primary targets for the treatment of spinal cord injury is to develop strategies to antagonize extrinsic or intrinsic axonal growth-inhibitory factors or enhance the factors that support axonal growth. Among these factors, a series of individual protein level disorders have been identified during the generation of axons following spinal cord injury. Moreover, an increasing number of studies have indicated that post-translational modifications of these proteins have important implications for axonal growth. Some researchers have discovered a variety of post-translational modifications after spinal cord injury, such as tyrosination, acetylation, and phosphorylation. In this review, we reviewed the post-translational modifications for axonal growth, functional recovery, and neuropathic pain after spinal cord injury, a better understanding of which may elucidate the dynamic change of spinal cord injury-related molecules and facilitate the development of a new therapeutic strategy for spinal cord injury.
Collapse
Affiliation(s)
- Shuang Zhu
- Department of Joint and Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Bing-Sheng Yang
- Department of Joint and Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Si-Jing Li
- Department of Joint and Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Ge Tong
- Department of Medical Ultrasonics, Guangdong Province Key Laboratory of Hepatology Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Jian-Ye Tan
- Department of Joint and Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Guo-Feng Wu
- Department of Joint and Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Lin Li
- Department of Joint and Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Guo-Li Chen
- Department of Orthopedics, Affiliated Hospital of Putian University, Putian, Fujian Province, China
| | - Qian Chen
- Cell and Molecular Biology Laboratory, Department of Orthopaedics, Alpert Medical School of Brown University/Rhode Island Hospital, Providence, RI, USA
| | - Li-Jun Lin
- Department of Joint and Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| |
Collapse
|
14
|
Fischhaber N, Faber J, Bakirci E, Dalton PD, Budday S, Villmann C, Schaefer N. Spinal Cord Neuronal Network Formation in a 3D Printed Reinforced Matrix-A Model System to Study Disease Mechanisms. Adv Healthc Mater 2021; 10:e2100830. [PMID: 34350717 PMCID: PMC11469053 DOI: 10.1002/adhm.202100830] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 06/24/2021] [Indexed: 12/29/2022]
Abstract
3D cell cultures allow a better mimicry of the biological and mechanical environment of cells in vivo compared to 2D cultures. However, 3D cell cultures have been challenging for ultrasoft tissues such as the brain. The present study uses a microfiber reinforcement approach combining mouse primary spinal cord neurons in Matrigel with melt electrowritten (MEW) frames. Within these 3D constructs, neuronal network development is followed for 21 days in vitro. To evaluate neuronal development in 3D constructs, the maturation of inhibitory glycinergic synapses is analyzed using protein expression, the complex mechanical properties by assessing nonlinearity, conditioning, and stress relaxation, and calcium imaging as readouts. Following adaptation to the 3D matrix-frame, mature inhibitory synapse formation is faster than in 2D demonstrated by a steep increase in glycine receptor expression between days 3 and 10. The 3D expression pattern of marker proteins at the inhibitory synapse and the mechanical properties resemble the situation in native spinal cord tissue. Moreover, 3D spinal cord neuronal networks exhibit intensive neuronal activity after 14 days in culture. The spinal cord cell culture model using ultrasoft matrix reinforced by MEW fibers provides a promising tool to study and understand biomechanical mechanisms in health and disease.
Collapse
Affiliation(s)
- Natalie Fischhaber
- Institute for Clinical NeurobiologyUniversity Hospital WürzburgVersbacherstr. 597078WürzburgGermany
| | - Jessica Faber
- Department of Mechanical EngineeringInstitute of Applied MechanicsFriedrich‐Alexander‐University Erlangen‐NürnbergEgerlandstrasse 591058ErlangenGermany
| | - Ezgi Bakirci
- Department of Functional Materials in Medicine and Dentistry and Bavarian Polymer InstituteUniversity Hospital WürzburgPleicherwall 297070WürzburgGermany
| | - Paul D. Dalton
- Department of Functional Materials in Medicine and Dentistry and Bavarian Polymer InstituteUniversity Hospital WürzburgPleicherwall 297070WürzburgGermany
- Phil and Penny Knight Campus for Accelerating Scientific ImpactUniversity of Oregon1505 Franklin Blvd.EugeneOR97403USA
| | - Silvia Budday
- Department of Mechanical EngineeringInstitute of Applied MechanicsFriedrich‐Alexander‐University Erlangen‐NürnbergEgerlandstrasse 591058ErlangenGermany
| | - Carmen Villmann
- Institute for Clinical NeurobiologyUniversity Hospital WürzburgVersbacherstr. 597078WürzburgGermany
| | - Natascha Schaefer
- Institute for Clinical NeurobiologyUniversity Hospital WürzburgVersbacherstr. 597078WürzburgGermany
| |
Collapse
|
15
|
Teo S, Salinas PC. Wnt-Frizzled Signaling Regulates Activity-Mediated Synapse Formation. Front Mol Neurosci 2021; 14:683035. [PMID: 34194299 PMCID: PMC8236581 DOI: 10.3389/fnmol.2021.683035] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 05/17/2021] [Indexed: 11/26/2022] Open
Abstract
The formation of synapses is a tightly regulated process that requires the coordinated assembly of the presynaptic and postsynaptic sides. Defects in synaptogenesis during development or in the adult can lead to neurodevelopmental disorders, neurological disorders, and neurodegenerative diseases. In order to develop therapeutic approaches for these neurological conditions, we must first understand the molecular mechanisms that regulate synapse formation. The Wnt family of secreted glycoproteins are key regulators of synapse formation in different model systems from invertebrates to mammals. In this review, we will discuss the role of Wnt signaling in the formation of excitatory synapses in the mammalian brain by focusing on Wnt7a and Wnt5a, two Wnt ligands that play an in vivo role in this process. We will also discuss how changes in neuronal activity modulate the expression and/or release of Wnts, resulting in changes in the localization of surface levels of Frizzled, key Wnt receptors, at the synapse. Thus, changes in neuronal activity influence the magnitude of Wnt signaling, which in turn contributes to activity-mediated synapse formation.
Collapse
Affiliation(s)
| | - Patricia C. Salinas
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| |
Collapse
|
16
|
Gutiérrez Y, López-García S, Lario A, Gutiérrez-Eisman S, Delevoye C, Esteban JA. KIF13A drives AMPA receptor synaptic delivery for long-term potentiation via endosomal remodeling. J Cell Biol 2021; 220:212112. [PMID: 33999113 PMCID: PMC8129809 DOI: 10.1083/jcb.202003183] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 02/16/2021] [Accepted: 03/31/2021] [Indexed: 02/06/2023] Open
Abstract
The regulated trafficking of AMPA-type glutamate receptors (AMPARs) from dendritic compartments to the synaptic membrane in response to neuronal activity is a core mechanism for long-term potentiation (LTP). However, the contribution of the microtubule cytoskeleton to this synaptic transport is still unknown. In this work, using electrophysiological, biochemical, and imaging techniques, we have found that one member of the kinesin-3 family of motor proteins, KIF13A, is specifically required for the delivery of AMPARs to the spine surface during LTP induction. Accordingly, KIF13A depletion from hippocampal slices abolishes LTP expression. We also identify the vesicular protein centaurin-α1 as part of a motor transport machinery that is engaged with KIF13A and AMPARs upon LTP induction. Finally, we determine that KIF13A is responsible for the remodeling of Rab11-FIP2 endosomal structures in the dendritic shaft during LTP. Overall, these results identify specific kinesin molecular motors and endosomal transport machinery that catalyzes the dendrite-to-synapse translocation of AMPA receptors during synaptic plasticity.
Collapse
Affiliation(s)
- Yolanda Gutiérrez
- Molecular Neuropathology Unit, Centro de Biología Molecular Severo Ochoa (Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid), Madrid, Spain
| | - Sergio López-García
- Molecular Neuropathology Unit, Centro de Biología Molecular Severo Ochoa (Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid), Madrid, Spain
| | - Argentina Lario
- Molecular Neuropathology Unit, Centro de Biología Molecular Severo Ochoa (Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid), Madrid, Spain
| | - Silvia Gutiérrez-Eisman
- Molecular Neuropathology Unit, Centro de Biología Molecular Severo Ochoa (Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid), Madrid, Spain
| | - Cédric Delevoye
- Institut Curie, Paris Sciences et Lettres Research University, Centre National de la Recherche Scientifique, UMR144, Structure and Membrane Compartments, Paris, France.,Institut Curie, Paris Sciences et Lettres Research University, Centre National de la Recherche Scientifique, UMR144, Cell and Tissue Imaging Facility, Paris, France
| | - José A Esteban
- Molecular Neuropathology Unit, Centro de Biología Molecular Severo Ochoa (Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid), Madrid, Spain
| |
Collapse
|
17
|
Wagner S, Lee C, Rojas L, Specht CG, Rhee J, Brose N, Papadopoulos T. The α3 subunit of GABA A receptors promotes formation of inhibitory synapses in the absence of collybistin. J Biol Chem 2021; 296:100709. [PMID: 33901490 PMCID: PMC8141935 DOI: 10.1016/j.jbc.2021.100709] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 04/14/2021] [Accepted: 04/22/2021] [Indexed: 01/03/2023] Open
Abstract
Signaling at nerve cell synapses is a key determinant of proper brain function, and synaptic defects—or synaptopathies—are at the basis of many neurological and psychiatric disorders. Collybistin (CB), a brain-specific guanine nucleotide exchange factor, is essential for the formation of γ-aminobutyric acidergic (GABAergic) postsynapses in defined regions of the mammalian forebrain, including the hippocampus and basolateral amygdala. This process depends on a direct interaction of CB with the scaffolding protein gephyrin, which leads to the redistribution of gephyrin into submembranous clusters at nascent inhibitory synapses. Strikingly, synaptic clustering of gephyrin and GABAA type A receptors (GABAARs) in several brain regions, including the cerebral cortex and certain thalamic areas, is unperturbed in CB-deficient mice, indicating that the formation of a substantial subset of inhibitory postsynapses must be controlled by gephyrin-interacting proteins other than CB. Previous studies indicated that the α3 subunit of GABAARs (GABAAR-α3) binds directly and with high affinity to gephyrin. Here, we provide evidence (i) that a homooligomeric GABAAR-α3A343W mutant induces the formation of submembranous gephyrin clusters independently of CB in COS-7 cells, (ii) that gephyrin clustering is unaltered in the neuronal subpopulations endogenously expressing the GABAAR-α3 in CB-deficient brains, and (iii) that exogenous expression of GABAAR-α3 partially rescues impaired gephyrin clustering in CB-deficient hippocampal neurons. Our results identify an important role of GABAAR-α3 in promoting gephyrin-mediated and CB-independent formation of inhibitory postsynapses.
Collapse
Affiliation(s)
- Sven Wagner
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - ChoongKu Lee
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Lucia Rojas
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Christian G Specht
- Diseases and Hormones of the Nervous System (DHNS), Inserm U1195, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - JeongSeop Rhee
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Nils Brose
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | | |
Collapse
|
18
|
Zhou L, Kiss E, Demmig R, Kirsch J, Nawrotzki RA, Kuhse J. Binding of gephyrin to microtubules is regulated by its phosphorylation at Ser270. Histochem Cell Biol 2021; 156:5-18. [PMID: 33796945 PMCID: PMC8277605 DOI: 10.1007/s00418-021-01973-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/13/2021] [Indexed: 12/23/2022]
Abstract
Gephyrin is a multifunctional scaffolding protein anchoring glycine- and subtypes of GABA type A- receptors at inhibitory postsynaptic membrane specializations by binding to the microtubule (MT) and/or the actin cytoskeleton. However, the conditions under which gephyrin can bind to MTs and its regulation are currently unknown. Here, we demonstrate that during the purification of MTs from rat brain by sedimentation of polymerized tubulin using high-speed centrifugation a fraction of gephyrin was bound to MTs, whereas gephyrin phosphorylated at the CDK5-dependent site Ser270 was detached from MTs and remained in the soluble protein fraction. Moreover, after collybistin fostered phosphorylation at Ser270 the binding of a recombinant gephyrin to MTs was strongly reduced in co-sedimentation assays. Correspondingly, upon substitution of wild-type gephyrin with recombinant gephyrin carrying alanine mutations at putative CDK5 phosphorylation sites the binding of gephyrin to MTs was increased. Furthermore, the analysis of cultured HEK293T and U2OS cells by immunofluorescence-microscopy disclosed a dispersed and punctuated endogenous gephyrin immunoreactivity co-localizing with MTs which was evidently not phosphorylated at Ser270. Thus, our study provides additional evidence for the binding of gephyrin to MTs in brain tissue and in in vitro cell systems. More importantly, our findings indicate that gephyrin-MT binding is restricted to a specific gephyrin fraction and depicts phosphorylation of gephyrin as a regulatory mechanism of this process by showing that soluble gephyrin detached from MTs can be detected specifically with the mAb7a antibody, which recognizes the Ser270 phosphorylated- version of gephyrin.
Collapse
Affiliation(s)
- Lin Zhou
- Department of Histology and Embryology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Eva Kiss
- Department of Cellular and Molecular Biology, University of Medicine, Pharmacy, Science and Technology "G.E. Palade" of Târgu Mures, Târgu Mures, Romania
| | - Rebecca Demmig
- University of Konstanz, Molecular Genetics, Konstanz, Germany
| | - Joachim Kirsch
- Department of Anatomy and Cell Biology, Institut für Anatomie und Zellbiologie, University of Heidelberg, Lehrstuhl II, Im Neuenheimer Feld 307, 69120, Heidelberg, Germany
| | - Ralph Alexander Nawrotzki
- Department of Anatomy and Cell Biology, Institut für Anatomie und Zellbiologie, University of Heidelberg, Lehrstuhl II, Im Neuenheimer Feld 307, 69120, Heidelberg, Germany
| | - Jochen Kuhse
- Department of Anatomy and Cell Biology, Institut für Anatomie und Zellbiologie, University of Heidelberg, Lehrstuhl II, Im Neuenheimer Feld 307, 69120, Heidelberg, Germany.
| |
Collapse
|
19
|
Parato J, Bartolini F. The microtubule cytoskeleton at the synapse. Neurosci Lett 2021; 753:135850. [PMID: 33775740 DOI: 10.1016/j.neulet.2021.135850] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 03/19/2021] [Accepted: 03/22/2021] [Indexed: 12/13/2022]
Abstract
In neurons, microtubules (MTs) provide routes for transport throughout the cell and structural support for dendrites and axons. Both stable and dynamic MTs are necessary for normal neuronal functions. Research in the last two decades has demonstrated that MTs play additional roles in synaptic structure and function in both pre- and postsynaptic elements. Here, we review current knowledge of the functions that MTs perform in excitatory and inhibitory synapses, as well as in the neuromuscular junction and other specialized synapses, and discuss the implications that this knowledge may have in neurological disease.
Collapse
Affiliation(s)
- Julie Parato
- Columbia University Medical Center, Department of Pathology & Cell Biology, 630 West 168(th)Street, P&S 15-421, NY, NY, 10032, United States; SUNY Empire State College, Department of Natural Sciences, 177 Livingston Street, Brooklyn, NY, 11201, United States
| | - Francesca Bartolini
- Columbia University Medical Center, Department of Pathology & Cell Biology, 630 West 168(th)Street, P&S 15-421, NY, NY, 10032, United States.
| |
Collapse
|
20
|
Bodakuntla S, Janke C, Magiera MM. Tubulin polyglutamylation, a regulator of microtubule functions, can cause neurodegeneration. Neurosci Lett 2021; 746:135656. [PMID: 33482309 DOI: 10.1016/j.neulet.2021.135656] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 01/05/2021] [Accepted: 01/07/2021] [Indexed: 02/07/2023]
Abstract
Neurodegenerative diseases lead to a progressive demise of neuronal functions that ultimately results in neuronal death. Besides a large variety of molecular pathways that have been linked to the degeneration of neurons, dysfunctions of the microtubule cytoskeleton are common features of many human neurodegenerative disorders. Yet, it is unclear whether microtubule dysfunctions are causative, or mere bystanders in the disease progression. A so-far little explored regulatory mechanism of the microtubule cytoskeleton, the posttranslational modifications of tubulin, emerge as candidate mechanisms involved in neuronal dysfunction, and thus, degeneration. Here we review the role of tubulin polyglutamylation, a prominent modification of neuronal microtubules. We discuss the current understanding of how polyglutamylation controls microtubule functions in healthy neurons, and how deregulation of this modification leads to neurodegeneration in mice and humans.
Collapse
Affiliation(s)
- Satish Bodakuntla
- Institut Curie, PSL Research University, CNRS UMR3348, F-91401 Orsay, France; Université Paris-Saclay, CNRS UMR3348, F-91401 Orsay, France
| | - Carsten Janke
- Institut Curie, PSL Research University, CNRS UMR3348, F-91401 Orsay, France; Université Paris-Saclay, CNRS UMR3348, F-91401 Orsay, France.
| | - Maria M Magiera
- Institut Curie, PSL Research University, CNRS UMR3348, F-91401 Orsay, France; Université Paris-Saclay, CNRS UMR3348, F-91401 Orsay, France.
| |
Collapse
|
21
|
Squires KE, Gerber KJ, Tillman MC, Lustberg DJ, Montañez-Miranda C, Zhao M, Ramineni S, Scharer CD, Saha RN, Shu FJ, Schroeder JP, Ortlund EA, Weinshenker D, Dudek SM, Hepler JR. Human genetic variants disrupt RGS14 nuclear shuttling and regulation of LTP in hippocampal neurons. J Biol Chem 2021; 296:100024. [PMID: 33410399 PMCID: PMC7949046 DOI: 10.1074/jbc.ra120.016009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 10/26/2020] [Accepted: 11/04/2020] [Indexed: 12/13/2022] Open
Abstract
The human genome contains vast genetic diversity as naturally occurring coding variants, yet the impact of these variants on protein function and physiology is poorly understood. RGS14 is a multifunctional signaling protein that suppresses synaptic plasticity in dendritic spines of hippocampal neurons. RGS14 also is a nucleocytoplasmic shuttling protein, suggesting that balanced nuclear import/export and dendritic spine localization are essential for RGS14 functions. We identified genetic variants L505R (LR) and R507Q (RQ) located within the nuclear export sequence (NES) of human RGS14. Here we report that RGS14 encoding LR or RQ profoundly impacts protein functions in hippocampal neurons. RGS14 membrane localization is regulated by binding Gαi-GDP, whereas RGS14 nuclear export is regulated by Exportin 1 (XPO1). Remarkably, LR and RQ variants disrupt RGS14 binding to Gαi1-GDP and XPO1, nucleocytoplasmic equilibrium, and capacity to inhibit long-term potentiation (LTP). Variant LR accumulates irreversibly in the nucleus, preventing RGS14 binding to Gαi1, localization to dendritic spines, and inhibitory actions on LTP induction, while variant RQ exhibits a mixed phenotype. When introduced into mice by CRISPR/Cas9, RGS14-LR protein expression was detected predominantly in the nuclei of neurons within hippocampus, central amygdala, piriform cortex, and striatum, brain regions associated with learning and synaptic plasticity. Whereas mice completely lacking RGS14 exhibit enhanced spatial learning, mice carrying variant LR exhibit normal spatial learning, suggesting that RGS14 may have distinct functions in the nucleus independent from those in dendrites and spines. These findings show that naturally occurring genetic variants can profoundly alter normal protein function, impacting physiology in unexpected ways.
Collapse
Affiliation(s)
- Katherine E Squires
- Department of Pharmacology and Chemical Biology, Emory University, Atlanta Georgia, USA
| | - Kyle J Gerber
- Department of Pharmacology and Chemical Biology, Emory University, Atlanta Georgia, USA
| | | | - Daniel J Lustberg
- Department of Human Genetics, Emory University, Atlanta Georgia, USA
| | | | - Meilan Zhao
- National Institute of Environmental Health Sciences, Research Triangle Park, Raleigh North Carolina, USA
| | - Suneela Ramineni
- Department of Pharmacology and Chemical Biology, Emory University, Atlanta Georgia, USA
| | | | - Ramendra N Saha
- Department of Molecular & Cell Biology, University of California-Merced, Merced California, USA
| | - Feng-Jue Shu
- Department of Pharmacology and Chemical Biology, Emory University, Atlanta Georgia, USA
| | - Jason P Schroeder
- Department of Human Genetics, Emory University, Atlanta Georgia, USA
| | - Eric A Ortlund
- Department of Biochemistry, Emory University, Atlanta Georgia, USA
| | - David Weinshenker
- Department of Human Genetics, Emory University, Atlanta Georgia, USA
| | - Serena M Dudek
- National Institute of Environmental Health Sciences, Research Triangle Park, Raleigh North Carolina, USA
| | - John R Hepler
- Department of Pharmacology and Chemical Biology, Emory University, Atlanta Georgia, USA.
| |
Collapse
|
22
|
Lopes AT, Hausrat TJ, Heisler FF, Gromova KV, Lombino FL, Fischer T, Ruschkies L, Breiden P, Thies E, Hermans-Borgmeyer I, Schweizer M, Schwarz JR, Lohr C, Kneussel M. Spastin depletion increases tubulin polyglutamylation and impairs kinesin-mediated neuronal transport, leading to working and associative memory deficits. PLoS Biol 2020; 18:e3000820. [PMID: 32866173 PMCID: PMC7485986 DOI: 10.1371/journal.pbio.3000820] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 09/11/2020] [Accepted: 08/10/2020] [Indexed: 12/21/2022] Open
Abstract
Mutations in the gene encoding the microtubule-severing protein spastin (spastic paraplegia 4 [SPG4]) cause hereditary spastic paraplegia (HSP), associated with neurodegeneration, spasticity, and motor impairment. Complicated forms (complicated HSP [cHSP]) further include cognitive deficits and dementia; however, the etiology and dysfunctional mechanisms of cHSP have remained unknown. Here, we report specific working and associative memory deficits upon spastin depletion in mice. Loss of spastin-mediated severing leads to reduced synapse numbers, accompanied by lower miniature excitatory postsynaptic current (mEPSC) frequencies. At the subcellular level, mutant neurons are characterized by longer microtubules with increased tubulin polyglutamylation levels. Notably, these conditions reduce kinesin-microtubule binding, impair the processivity of kinesin family protein (KIF) 5, and reduce the delivery of presynaptic vesicles and postsynaptic α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors. Rescue experiments confirm the specificity of these results by showing that wild-type spastin, but not the severing-deficient and disease-associated K388R mutant, normalizes the effects at the synaptic, microtubule, and transport levels. In addition, short hairpin RNA (shRNA)-mediated reduction of tubulin polyglutamylation on spastin knockout background normalizes KIF5 transport deficits and attenuates the loss of excitatory synapses. Our data provide a mechanism that connects spastin dysfunction with the regulation of kinesin-mediated cargo transport, synapse integrity, and cognition. This study identifies deficits in working and associative memory in spastin knockout mice, resembling the cognitive deficits described in humans with severe forms of SPG4-type hereditary spastic paraplegia. Mechanistically, the findings suggest that impaired microtubule growth, kinesin motility and cargo delivery of synaptic AMPA receptors may contribute to the etiology of the disease.
Collapse
Affiliation(s)
- André T. Lopes
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Torben J. Hausrat
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Frank F. Heisler
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kira V. Gromova
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Franco L. Lombino
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Timo Fischer
- Division of Neurophysiology, University of Hamburg, Hamburg, Germany
| | - Laura Ruschkies
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Petra Breiden
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Edda Thies
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Irm Hermans-Borgmeyer
- Transgenic Animal Unit, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Michaela Schweizer
- Morphology Unit, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jürgen R. Schwarz
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christian Lohr
- Division of Neurophysiology, University of Hamburg, Hamburg, Germany
| | - Matthias Kneussel
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- * E-mail:
| |
Collapse
|
23
|
Moutin MJ, Bosc C, Peris L, Andrieux A. Tubulin post-translational modifications control neuronal development and functions. Dev Neurobiol 2020; 81:253-272. [PMID: 33325152 PMCID: PMC8246997 DOI: 10.1002/dneu.22774] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 05/26/2020] [Accepted: 07/14/2020] [Indexed: 12/22/2022]
Abstract
Microtubules (MTs) are an essential component of the neuronal cytoskeleton; they are involved in various aspects of neuron development, maintenance, and functions including polarization, synaptic plasticity, and transport. Neuronal MTs are highly heterogeneous due to the presence of multiple tubulin isotypes and extensive post‐translational modifications (PTMs). These PTMs—most notably detyrosination, acetylation, and polyglutamylation—have emerged as important regulators of the neuronal microtubule cytoskeleton. With this review, we summarize what is currently known about the impact of tubulin PTMs on microtubule dynamics, neuronal differentiation, plasticity, and transport as well as on brain function in normal and pathological conditions, in particular during neuro‐degeneration. The main therapeutic approaches to neuro‐diseases based on the modulation of tubulin PTMs are also summarized. Overall, the review indicates how tubulin PTMs can generate a large number of functionally specialized microtubule sub‐networks, each of which is crucial to specific neuronal features.
Collapse
Affiliation(s)
- Marie-Jo Moutin
- Grenoble Institut Neurosciences, University Grenoble Alpes, Inserm, U1216, CEA, CNRS, Grenoble, France
| | - Christophe Bosc
- Grenoble Institut Neurosciences, University Grenoble Alpes, Inserm, U1216, CEA, CNRS, Grenoble, France
| | - Leticia Peris
- Grenoble Institut Neurosciences, University Grenoble Alpes, Inserm, U1216, CEA, CNRS, Grenoble, France
| | - Annie Andrieux
- Grenoble Institut Neurosciences, University Grenoble Alpes, Inserm, U1216, CEA, CNRS, Grenoble, France
| |
Collapse
|
24
|
Radler MR, Suber A, Spiliotis ET. Spatial control of membrane traffic in neuronal dendrites. Mol Cell Neurosci 2020; 105:103492. [PMID: 32294508 PMCID: PMC7317674 DOI: 10.1016/j.mcn.2020.103492] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 03/24/2020] [Accepted: 04/01/2020] [Indexed: 02/06/2023] Open
Abstract
Neuronal dendrites are highly branched and specialized compartments with distinct structures and secretory organelles (e.g., spines, Golgi outposts), and a unique cytoskeletal organization that includes microtubules of mixed polarity. Dendritic membranes are enriched with proteins, which specialize in the formation and function of the post-synaptic membrane of the neuronal synapse. How these proteins partition preferentially in dendrites, and how they traffic in a manner that is spatiotemporally accurate and regulated by synaptic activity are long-standing questions of neuronal cell biology. Recent studies have shed new insights into the spatial control of dendritic membrane traffic, revealing new classes of proteins (e.g., septins) and cytoskeleton-based mechanisms with dendrite-specific functions. Here, we review these advances by revisiting the fundamental mechanisms that control membrane traffic at the levels of protein sorting and motor-driven transport on microtubules and actin filaments. Overall, dendrites possess unique mechanisms for the spatial control of membrane traffic, which might have specialized and co-evolved with their highly arborized morphology.
Collapse
Affiliation(s)
- Megan R Radler
- Department of Biology, Drexel University, 3245 Chestnut St, Philadelphia, PA 19104, USA
| | - Ayana Suber
- Department of Biology, Drexel University, 3245 Chestnut St, Philadelphia, PA 19104, USA
| | - Elias T Spiliotis
- Department of Biology, Drexel University, 3245 Chestnut St, Philadelphia, PA 19104, USA.
| |
Collapse
|
25
|
Reduced TUBA1A Tubulin Causes Defects in Trafficking and Impaired Adult Motor Behavior. eNeuro 2020; 7:ENEURO.0045-20.2020. [PMID: 32184299 PMCID: PMC7218002 DOI: 10.1523/eneuro.0045-20.2020] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 03/03/2020] [Indexed: 12/30/2022] Open
Abstract
Newly born neurons express high levels of TUBA1A α-tubulin to assemble microtubules for neurite extension and to provide tracks for intracellular transport. In the adult brain, Tuba1a expression decreases dramatically. A mouse that harbors a loss-of-function mutation in the gene encoding TUBA1A (Tuba1aND/+) allows us to ask whether TUBA1A is important for the function of mature neurons. α-Tubulin levels are about half of wild type in juvenile Tuba1aND/+ brains, but are close to normal in older animals. In postnatal day (P)0 cultured neurons, reduced TUBA1A allows for assembly of less microtubules in axons resulting in more pausing during organelle trafficking. While Tuba1aND/+ mouse behavior is indistinguishable from wild-type siblings at weaning, Tuba1aND/+ mice develop adult-onset ataxia. Neurons important for motor function in Tuba1aND/+ remain indistinguishable from wild-type with respect to morphology and number and display no evidence of axon degeneration. Tuba1aND/+ neuromuscular junction (NMJ) synapses are the same size as wild-type before the onset of ataxia, but are reduced in size in older animals. Together, these data indicate that the TUBA1A-rich microtubule tracks that are assembled during development are essential for mature neuron function and maintenance of synapses over time.
Collapse
|
26
|
Hausrat TJ, Radwitz J, Lombino FL, Breiden P, Kneussel M. Alpha- and beta-tubulin isotypes are differentially expressed during brain development. Dev Neurobiol 2020; 81:333-350. [PMID: 32293117 DOI: 10.1002/dneu.22745] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 01/21/2020] [Accepted: 04/08/2020] [Indexed: 12/14/2022]
Abstract
Alpha- and beta-tubulin dimers polymerize into protofilaments that associate laterally to constitute a hollow tube, the microtubule. A dynamic network of interlinking filaments forms the microtubule cytoskeleton, which maintains the structure of cells and is key to various cellular processes including cell division, cell migration, and intracellular transport. Individual microtubules have an identity that depends on the differential integration of specific alpha- and beta-tubulin isotypes and is further specified by a variety of posttranslational modifications (PTMs). It is barely understood to which extent neighboring microtubules differ in their tubulin composition or whether specific tubulin isotypes cluster along the polymer. Furthermore, our knowledge about the spatio-temporal expression patterns of tubulin isotypes is limited, not at least due to the lack of antibodies or antibody cross-reactivities. Here, we asked which alpha- and beta-tubulin mRNAs and proteins are expressed in developing hippocampal neuron cultures and ex vivo brain tissue lysates. Using heterologous expression of GFP-tubulin fusion proteins, we systematically tested antibody-specificities against various tubulin isotypes. Our data provide quantitative information about tubulin expression levels in the mouse brain and classify tubulin isotypes during pre- and postnatal development.
Collapse
Affiliation(s)
- Torben J Hausrat
- Department of Molecular Neurogenetics, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jennifer Radwitz
- Department of Molecular Neurogenetics, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Franco L Lombino
- Department of Molecular Neurogenetics, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Petra Breiden
- Department of Molecular Neurogenetics, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Matthias Kneussel
- Department of Molecular Neurogenetics, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
27
|
The tubulin code and its role in controlling microtubule properties and functions. Nat Rev Mol Cell Biol 2020; 21:307-326. [PMID: 32107477 DOI: 10.1038/s41580-020-0214-3] [Citation(s) in RCA: 488] [Impact Index Per Article: 97.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/14/2020] [Indexed: 02/07/2023]
Abstract
Microtubules are core components of the eukaryotic cytoskeleton with essential roles in cell division, shaping, motility and intracellular transport. Despite their functional heterogeneity, microtubules have a highly conserved structure made from almost identical molecular building blocks: the tubulin proteins. Alternative tubulin isotypes and a variety of post-translational modifications control the properties and functions of the microtubule cytoskeleton, a concept known as the 'tubulin code'. Here we review the current understanding of the molecular components of the tubulin code and how they impact microtubule properties and functions. We discuss how tubulin isotypes and post-translational modifications control microtubule behaviour at the molecular level and how this translates into physiological functions at the cellular and organism levels. We then go on to show how fine-tuning of microtubule function by some tubulin modifications can affect homeostasis and how perturbation of this fine-tuning can lead to a range of dysfunctions, many of which are linked to human disease.
Collapse
|
28
|
Bodakuntla S, Schnitzler A, Villablanca C, Gonzalez-Billault C, Bieche I, Janke C, Magiera MM. Tubulin polyglutamylation is a general traffic-control mechanism in hippocampal neurons. J Cell Sci 2020; 133:jcs241802. [PMID: 31932508 DOI: 10.1242/jcs.241802] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 12/23/2019] [Indexed: 08/31/2023] Open
Abstract
Neurons are highly complex cells that heavily rely on intracellular transport to distribute a range of functionally essential cargoes within the cell. Post-translational modifications of tubulin are emerging as mechanisms for regulating microtubule functions, but their impact on neuronal transport is only marginally understood. Here, we have systematically studied the impact of post-translational polyglutamylation on axonal transport. In cultured hippocampal neurons, deletion of a single deglutamylase, CCP1 (also known as AGTPBP1), is sufficient to induce abnormal accumulation of polyglutamylation, i.e. hyperglutamylation. We next investigated how hyperglutamylation affects axonal transport of a range of functionally different neuronal cargoes: mitochondria, lysosomes, LAMP1 endosomes and BDNF vesicles. Strikingly, we found a reduced motility for all these cargoes, suggesting that polyglutamylation could act as a regulator of cargo transport in neurons. This, together with the recent discovery that hyperglutamylation induces neurodegeneration, makes it likely that perturbed neuronal trafficking could be one of the central molecular causes underlying this novel type of degeneration.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Satish Bodakuntla
- Institut Curie, PSL Research University, CNRS UMR3348, F-91405 Orsay, France
- Université Paris Sud, Université Paris-Saclay, CNRS UMR3348, F-91405 Orsay, France
| | - Anne Schnitzler
- Institut Curie, PSL Research University, Department of Genetics, F-75005 Paris, France
| | - Cristopher Villablanca
- Center for Geroscience, Brain Health and Metabolism (GERO), Santiago 7800003, Chile
- Department of Biology, Faculty of Sciences, University of Chile, Santiago 7800003, Chile
| | - Christian Gonzalez-Billault
- Center for Geroscience, Brain Health and Metabolism (GERO), Santiago 7800003, Chile
- Department of Biology, Faculty of Sciences, University of Chile, Santiago 7800003, Chile
| | - Ivan Bieche
- Institut Curie, PSL Research University, Department of Genetics, F-75005 Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, F-75005 Paris, France
| | - Carsten Janke
- Institut Curie, PSL Research University, CNRS UMR3348, F-91405 Orsay, France
- Université Paris Sud, Université Paris-Saclay, CNRS UMR3348, F-91405 Orsay, France
| | - Maria M Magiera
- Institut Curie, PSL Research University, CNRS UMR3348, F-91405 Orsay, France
- Université Paris Sud, Université Paris-Saclay, CNRS UMR3348, F-91405 Orsay, France
| |
Collapse
|
29
|
Bodakuntla S, Magiera MM, Janke C. Measuring the Impact of Tubulin Posttranslational Modifications on Axonal Transport. Methods Mol Biol 2020; 2101:353-370. [PMID: 31879913 DOI: 10.1007/978-1-0716-0219-5_20] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Axonal transport is a process essential for neuronal function and survival that takes place on the cellular highways-the microtubules. It requires three major components: the microtubules that serve as tracks for the transport, the motor proteins that drive the movement, and the transported cargoes with their adaptor proteins. Axonal transport could be controlled by tubulin posttranslational modifications, which by decorating specific microtubule tracks could determine the specificity of cargo delivery inside neurons. However, it appears that the effects of tubulin modifications on transport can be rather subtle, and might thus be easily overlooked depending on which parameter of the transport process is analyzed. Here we propose an analysis paradigm that allows detecting rather subtle alterations in neuronal transport, as induced for instance by accumulation of posttranslational polyglutamylation. Analyzing mitochondria movements in axons, we found that neither the average speed nor the distance traveled were affected by hyperglutamylation, but we detected an about 50% reduction of the overall motility, suggesting that polyglutamylation controls the efficiency of mitochondria transport in axons. Our protocol can readily be expanded to the analysis of the impact of other tubulin modifications on the transport of a range of different neuronal cargoes.
Collapse
Affiliation(s)
- Satish Bodakuntla
- Institut Curie, PSL Research University, CNRS UMR3348, Centre Universitaire, Orsay, France.,Université Paris Sud, Université Paris-Saclay, CNRS UMR3348, Orsay, France
| | - Maria M Magiera
- Institut Curie, PSL Research University, CNRS UMR3348, Centre Universitaire, Orsay, France. .,Université Paris Sud, Université Paris-Saclay, CNRS UMR3348, Orsay, France.
| | - Carsten Janke
- Institut Curie, PSL Research University, CNRS UMR3348, Centre Universitaire, Orsay, France. .,Université Paris Sud, Université Paris-Saclay, CNRS UMR3348, Orsay, France.
| |
Collapse
|
30
|
Brown CA, Del Corsso C, Zoidl C, Donaldson LW, Spray DC, Zoidl G. Tubulin-Dependent Transport of Connexin-36 Potentiates the Size and Strength of Electrical Synapses. Cells 2019; 8:E1146. [PMID: 31557934 PMCID: PMC6829524 DOI: 10.3390/cells8101146] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 09/17/2019] [Accepted: 09/20/2019] [Indexed: 12/12/2022] Open
Abstract
Connexin-36 (Cx36) electrical synapses strengthen transmission in a calcium/calmodulin (CaM)/calmodulin-dependent kinase II (CaMKII)-dependent manner similar to a mechanism whereby the N-methyl-D-aspartate (NMDA) receptor subunit NR2B facilitates chemical transmission. Since NR2B-microtubule interactions recruit receptors to the cell membrane during plasticity, we hypothesized an analogous modality for Cx36. We determined that Cx36 binding to tubulin at the carboxy-terminal domain was distinct from Cx43 and NR2B by binding a motif overlapping with the CaM and CaMKII binding motifs. Dual patch-clamp recordings demonstrated that pharmacological interference of the cytoskeleton and deleting the binding motif at the Cx36 carboxyl-terminal (CT) reversibly abolished Cx36 plasticity. Mechanistic details of trafficking to the gap-junction plaque (GJP) were probed pharmacologically and through mutational analysis, all of which affected GJP size and formation between cell pairs. Lys279, Ile280, and Lys281 positions were particularly critical. This study demonstrates that tubulin-dependent transport of Cx36 potentiates synaptic strength by delivering channels to GJPs, reinforcing the role of protein transport at chemical and electrical synapses to fine-tune communication between neurons.
Collapse
Affiliation(s)
- Cherie A Brown
- Department of Biology, York University, Toronto, ON M3J 1P3, Canada.
| | - Cristiane Del Corsso
- Department of Biophysics and Physiology, Federal University of Rio de Janeiro-RJ, Rio de Janeiro 21941-901, Brazil.
| | - Christiane Zoidl
- Department of Biology, York University, Toronto, ON M3J 1P3, Canada.
| | - Logan W Donaldson
- Department of Biology, York University, Toronto, ON M3J 1P3, Canada.
| | - David C Spray
- Department of Neuroscience, Albert Einstein College, Bronx, NY 10461, USA.
- Department of Medicine, Albert Einstein College, Bronx, NY 10461, USA.
| | - Georg Zoidl
- Department of Biology, York University, Toronto, ON M3J 1P3, Canada.
- Department of Psychology, York University, Toronto, ON M3J 1P3, Canada.
| |
Collapse
|
31
|
Groeneweg FL, Trattnig C, Kuhse J, Nawrotzki RA, Kirsch J. Gephyrin: a key regulatory protein of inhibitory synapses and beyond. Histochem Cell Biol 2018; 150:489-508. [DOI: 10.1007/s00418-018-1725-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/07/2018] [Indexed: 12/26/2022]
|
32
|
Kasaragod VB, Schindelin H. Structure-Function Relationships of Glycine and GABA A Receptors and Their Interplay With the Scaffolding Protein Gephyrin. Front Mol Neurosci 2018; 11:317. [PMID: 30258351 PMCID: PMC6143783 DOI: 10.3389/fnmol.2018.00317] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 08/16/2018] [Indexed: 12/03/2022] Open
Abstract
Glycine and γ-aminobutyric acid (GABA) are the major determinants of inhibition in the central nervous system (CNS). These neurotransmitters target glycine and GABAA receptors, respectively, which both belong to the Cys-loop superfamily of pentameric ligand-gated ion channels (pLGICs). Interactions of the neurotransmitters with the cognate receptors result in receptor opening and a subsequent influx of chloride ions, which, in turn, leads to hyperpolarization of the membrane potential, thus counteracting excitatory stimuli. The majority of glycine receptors and a significant fraction of GABAA receptors (GABAARs) are recruited and anchored to the post-synaptic membrane by the central scaffolding protein gephyrin. This ∼93 kDa moonlighting protein is structurally organized into an N-terminal G-domain (GephG) connected to a C-terminal E-domain (GephE) via a long unstructured linker. Both inhibitory neurotransmitter receptors interact via a short peptide motif located in the large cytoplasmic loop located in between transmembrane helices 3 and 4 (TM3-TM4) of the receptors with a universal receptor-binding epitope residing in GephE. Gephyrin engages in nearly identical interactions with the receptors at the N-terminal end of the peptide motif, and receptor-specific interaction toward the C-terminal region of the peptide. In addition to its receptor-anchoring function, gephyrin also interacts with a rather large collection of macromolecules including different cytoskeletal elements, thus acting as central scaffold at inhibitory post-synaptic specializations. Dysfunctions in receptor-mediated or gephyrin-mediated neurotransmission have been identified in various severe neurodevelopmental disorders. Although biochemical, cellular and electrophysiological studies have helped to understand the physiological and pharmacological roles of the receptors, recent high resolution structures of the receptors have strengthened our understanding of the receptors and their gating mechanisms. Besides that, multiple crystal structures of GephE in complex with receptor-derived peptides have shed light into receptor clustering by gephyrin at inhibitory post-synapses. This review will highlight recent biochemical and structural insights into gephyrin and the GlyRs as well as GABAA receptors, which provide a deeper understanding of the molecular machinery mediating inhibitory neurotransmission.
Collapse
Affiliation(s)
- Vikram B Kasaragod
- Institute of Structural Biology, Rudolf Virchow Center for Experimental Biomedicine, University of Würzburg, Würzburg, Germany
| | - Hermann Schindelin
- Institute of Structural Biology, Rudolf Virchow Center for Experimental Biomedicine, University of Würzburg, Würzburg, Germany
| |
Collapse
|
33
|
Bressloff PC, Maclaurin JN. Stochastic Hybrid Systems in Cellular Neuroscience. JOURNAL OF MATHEMATICAL NEUROSCIENCE 2018; 8:12. [PMID: 30136005 PMCID: PMC6104574 DOI: 10.1186/s13408-018-0067-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 08/05/2018] [Indexed: 06/08/2023]
Abstract
We review recent work on the theory and applications of stochastic hybrid systems in cellular neuroscience. A stochastic hybrid system or piecewise deterministic Markov process involves the coupling between a piecewise deterministic differential equation and a time-homogeneous Markov chain on some discrete space. The latter typically represents some random switching process. We begin by summarizing the basic theory of stochastic hybrid systems, including various approximation schemes in the fast switching (weak noise) limit. In subsequent sections, we consider various applications of stochastic hybrid systems, including stochastic ion channels and membrane voltage fluctuations, stochastic gap junctions and diffusion in randomly switching environments, and intracellular transport in axons and dendrites. Finally, we describe recent work on phase reduction methods for stochastic hybrid limit cycle oscillators.
Collapse
|
34
|
Schaefer N, Roemer V, Janzen D, Villmann C. Impaired Glycine Receptor Trafficking in Neurological Diseases. Front Mol Neurosci 2018; 11:291. [PMID: 30186111 PMCID: PMC6110938 DOI: 10.3389/fnmol.2018.00291] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 08/02/2018] [Indexed: 12/21/2022] Open
Abstract
Ionotropic glycine receptors (GlyRs) enable fast synaptic neurotransmission in the adult spinal cord and brainstem. The inhibitory GlyR is a transmembrane glycine-gated chloride channel. The immature GlyR protein undergoes various processing steps, e.g., folding, assembly, and maturation while traveling from the endoplasmic reticulum to and through the Golgi apparatus, where post-translational modifications, e.g., glycosylation occur. The mature receptors are forward transported via microtubules to the cellular surface and inserted into neuronal membranes followed by synaptic clustering. The normal life cycle of a receptor protein includes further processes like internalization, recycling, and degradation. Defects in GlyR life cycle, e.g., impaired protein maturation and degradation have been demonstrated to underlie pathological mechanisms of various neurological diseases. The neurological disorder startle disease is caused by glycinergic dysfunction mainly due to missense mutations in genes encoding GlyR subunits (GLRA1 and GLRB). In vitro studies have shown that most recessive forms of startle disease are associated with impaired receptor biogenesis. Another neurological disease with a phenotype similar to startle disease is a special form of stiff-person syndrome (SPS), which is most probably due to the development of GlyR autoantibodies. Binding of GlyR autoantibodies leads to enhanced receptor internalization. Here we focus on the normal life cycle of GlyRs concentrating on assembly and maturation, receptor trafficking, post-synaptic integration and clustering, and GlyR internalization/recycling/degradation. Furthermore, this review highlights findings on impairment of these processes under disease conditions such as disturbed neuronal ER-Golgi trafficking as the major pathomechanism for recessive forms of human startle disease. In SPS, enhanced receptor internalization upon autoantibody binding to the GlyR has been shown to underlie the human pathology. In addition, we discuss how the existing mouse models of startle disease increased our current knowledge of GlyR trafficking routes and function. This review further illuminates receptor trafficking of GlyR variants originally identified in startle disease patients and explains changes in the life cycle of GlyRs in patients with SPS with respect to structural and functional consequences at the receptor level.
Collapse
Affiliation(s)
- Natascha Schaefer
- Institute for Clinical Neurobiology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Vera Roemer
- Institute for Clinical Neurobiology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Dieter Janzen
- Institute for Clinical Neurobiology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Carmen Villmann
- Institute for Clinical Neurobiology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| |
Collapse
|
35
|
Ribeiro EA, Salery M, Scarpa JR, Calipari ES, Hamilton PJ, Ku SM, Kronman H, Purushothaman I, Juarez B, Heshmati M, Doyle M, Lardner C, Burek D, Strat A, Pirpinias S, Mouzon E, Han MH, Neve RL, Bagot RC, Kasarskis A, Koo JW, Nestler EJ. Transcriptional and physiological adaptations in nucleus accumbens somatostatin interneurons that regulate behavioral responses to cocaine. Nat Commun 2018; 9:3149. [PMID: 30089879 PMCID: PMC6082848 DOI: 10.1038/s41467-018-05657-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Accepted: 07/12/2018] [Indexed: 11/20/2022] Open
Abstract
The role of somatostatin interneurons in nucleus accumbens (NAc), a key brain reward region, remains poorly understood due to the fact that these cells account for < 1% of NAc neurons. Here, we use optogenetics, electrophysiology, and RNA-sequencing to characterize the transcriptome and functioning of NAc somatostatin interneurons after repeated exposure to cocaine. We find that the activity of somatostatin interneurons regulates behavioral responses to cocaine, with repeated cocaine reducing the excitability of these neurons. Repeated cocaine also induces transcriptome-wide changes in gene expression within NAc somatostatin interneurons. We identify the JUND transcription factor as a key regulator of cocaine action and confirmed, by use of viral-mediated gene transfer, that JUND activity in somatostatin interneurons influences behavioral responses to cocaine. Our results identify alterations in NAc induced by cocaine in a sparse population of somatostatin interneurons, and illustrate the value of studying brain diseases using cell type-specific whole transcriptome RNA-sequencing.
Collapse
Affiliation(s)
- Efrain A Ribeiro
- Department of Neuroscience, Friedman Brain Institute, New York, 10029, NY, USA
| | - Marine Salery
- Department of Neuroscience, Friedman Brain Institute, New York, 10029, NY, USA
| | - Joseph R Scarpa
- Department of Genetics and Genomic Science, Icahn Institute of Genomics and Multiscale Biology, New York, 10029, NY, USA
| | - Erin S Calipari
- Department of Neuroscience, Friedman Brain Institute, New York, 10029, NY, USA
| | - Peter J Hamilton
- Department of Neuroscience, Friedman Brain Institute, New York, 10029, NY, USA
| | - Stacy M Ku
- Department of Neuroscience, Friedman Brain Institute, New York, 10029, NY, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, 10029, NY, USA
| | - Hope Kronman
- Department of Neuroscience, Friedman Brain Institute, New York, 10029, NY, USA
| | | | - Barbara Juarez
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, 10029, NY, USA
| | - Mitra Heshmati
- Department of Neuroscience, Friedman Brain Institute, New York, 10029, NY, USA
| | - Marie Doyle
- Department of Neuroscience, Friedman Brain Institute, New York, 10029, NY, USA
| | - Casey Lardner
- Department of Neuroscience, Friedman Brain Institute, New York, 10029, NY, USA
| | - Dominicka Burek
- Department of Neuroscience, Friedman Brain Institute, New York, 10029, NY, USA
| | - Ana Strat
- Department of Neuroscience, Friedman Brain Institute, New York, 10029, NY, USA
| | - Stephen Pirpinias
- Department of Neuroscience, Friedman Brain Institute, New York, 10029, NY, USA
| | - Ezekiell Mouzon
- Department of Neuroscience, Friedman Brain Institute, New York, 10029, NY, USA
| | - Ming-Hu Han
- Department of Neuroscience, Friedman Brain Institute, New York, 10029, NY, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, 10029, NY, USA
| | - Rachael L Neve
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Rosemary C Bagot
- Department of Neuroscience, Friedman Brain Institute, New York, 10029, NY, USA
- Department of Psychology, McGill University, Québec, H3A 1B1, Montreal, Canada
| | - Andrew Kasarskis
- Department of Genetics and Genomic Science, Icahn Institute of Genomics and Multiscale Biology, New York, 10029, NY, USA
| | - Ja Wook Koo
- Department of Neuroscience, Friedman Brain Institute, New York, 10029, NY, USA
- Department of Neural Development and Disease, Korea Brain Research Institute, Daegu, 41068, Republic of Korea
| | - Eric J Nestler
- Department of Neuroscience, Friedman Brain Institute, New York, 10029, NY, USA.
| |
Collapse
|
36
|
Magiera MM, Singh P, Gadadhar S, Janke C. Tubulin Posttranslational Modifications and Emerging Links to Human Disease. Cell 2018; 173:1323-1327. [PMID: 29856952 DOI: 10.1016/j.cell.2018.05.018] [Citation(s) in RCA: 124] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
Tubulin posttranslational modifications are currently emerging as important regulators of the microtubule cytoskeleton and thus have a strong potential to be implicated in a number of disorders. Here, we review the latest advances in understanding the physiological roles of tubulin modifications and their links to a variety of pathologies.
Collapse
Affiliation(s)
- Maria M Magiera
- Institut Curie, PSL Research University, CNRS UMR3348, Orsay, France; Université Paris Sud, Université Paris-Saclay, CNRS UMR3348, Orsay, France.
| | - Puja Singh
- Institut Curie, PSL Research University, CNRS UMR3348, Orsay, France; Université Paris Sud, Université Paris-Saclay, CNRS UMR3348, Orsay, France
| | - Sudarshan Gadadhar
- Institut Curie, PSL Research University, CNRS UMR3348, Orsay, France; Université Paris Sud, Université Paris-Saclay, CNRS UMR3348, Orsay, France
| | - Carsten Janke
- Institut Curie, PSL Research University, CNRS UMR3348, Orsay, France; Université Paris Sud, Université Paris-Saclay, CNRS UMR3348, Orsay, France.
| |
Collapse
|
37
|
Kimura Y, Tsutsumi K, Konno A, Ikegami K, Hameed S, Kaneko T, Kaplan OI, Teramoto T, Fujiwara M, Ishihara T, Blacque OE, Setou M. Environmental responsiveness of tubulin glutamylation in sensory cilia is regulated by the p38 MAPK pathway. Sci Rep 2018; 8:8392. [PMID: 29849065 PMCID: PMC5976657 DOI: 10.1038/s41598-018-26694-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 05/16/2018] [Indexed: 01/09/2023] Open
Abstract
Glutamylation is a post-translational modification found on tubulin that can alter the interaction between microtubules (MTs) and associated proteins. The molecular mechanisms regulating tubulin glutamylation in response to the environment are not well understood. Here, we show that in the sensory cilia of Caenorhabditis elegans, tubulin glutamylation is upregulated in response to various signals such as temperature, osmolality, and dietary conditions. Similarly, tubulin glutamylation is modified in mammalian photoreceptor cells following light adaptation. A tubulin glutamate ligase gene ttll-4, which is essential for tubulin glutamylation of axonemal MTs in sensory cilia, is activated by p38 MAPK. Amino acid substitution of TTLL-4 has revealed that a Thr residue (a putative MAPK-phosphorylation site) is required for enhancement of tubulin glutamylation. Intraflagellar transport (IFT), a bidirectional trafficking system specifically observed along axonemal MTs, is required for the formation, maintenance, and function of sensory cilia. Measurement of the velocity of IFT particles revealed that starvation accelerates IFT, which was also dependent on the Thr residue of TTLL-4. Similarly, starvation-induced attenuation of avoidance behaviour from high osmolality conditions was also dependent on ttll-4. Our data suggest that a novel evolutionarily conserved regulatory system exists for tubulin glutamylation in sensory cilia in response to the environment.
Collapse
Affiliation(s)
- Yoshishige Kimura
- Department of Cellular & Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, Shizuoka, 431-3192, Japan
- Department of Liberal Arts and Sciences, Kanagawa University of Human Services, 1-10-1 Heisei-cho, Yokosuka, Kanagawa, 238-8522, Japan
| | - Koji Tsutsumi
- Department of Cellular & Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, Shizuoka, 431-3192, Japan
- Division of Cell Biology, Department of Biosciences, School of Science, Kitasato University, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa, 252-0373, Japan
| | - Alu Konno
- Department of Cellular & Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, Shizuoka, 431-3192, Japan
| | - Koji Ikegami
- Department of Cellular & Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, Shizuoka, 431-3192, Japan
- Department of Anatomy and Developmental Biology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8553, Japan
| | - Saira Hameed
- Department of Cellular & Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, Shizuoka, 431-3192, Japan
| | - Tomomi Kaneko
- Department of Liberal Arts and Sciences, Kanagawa University of Human Services, 1-10-1 Heisei-cho, Yokosuka, Kanagawa, 238-8522, Japan
| | - Oktay Ismail Kaplan
- School of Biomolecular and Biomedical Science, University College Dublin, Belfield, Dublin, 4, Ireland
- Abdullah Gul Universitesi, Doga Bilimleri Fakultesi, Sumer Kampusu, 38090, Kocasinan, Kayseri, Turkey
| | - Takayuki Teramoto
- Department of Biology, Faculty of Sciences, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Manabi Fujiwara
- Department of Biology, Faculty of Sciences, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Takeshi Ishihara
- Department of Biology, Faculty of Sciences, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Oliver E Blacque
- School of Biomolecular and Biomedical Science, University College Dublin, Belfield, Dublin, 4, Ireland
| | - Mitsutoshi Setou
- Department of Cellular & Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, Shizuoka, 431-3192, Japan.
- Department of Systems Molecular Anatomy, Institute for Medical Photonics Research, Preeminent Medical Photonics Education & Research Center, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, Shizuoka, 431-3192, Japan.
- The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan.
- Department of Anatomy, The University of Hong Kong, Hong Kong, China.
- Division of Neural Systematics, National Institute for Physiological Sciences, 38 Nishigonaka Myodaiji, Okazaki, Aichi, 444-8585, Japan.
| |
Collapse
|
38
|
The tubulin code in neuronal polarity. Curr Opin Neurobiol 2018; 51:95-102. [PMID: 29554585 DOI: 10.1016/j.conb.2018.03.001] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Revised: 02/28/2018] [Accepted: 03/01/2018] [Indexed: 11/22/2022]
Abstract
Cells depend on the asymmetric distribution of their components for homeostasis, differentiation and movement. In no other cell type is this requirement more critical than in the neuron where complex structures are generated during process growth and elaboration and cargo is transported over distances several thousand times the cell body diameter. Microtubules act both as dynamic structural elements and as tracks for intracellular transport. Microtubules are mosaic polymers containing multiple tubulin isoforms functionalized with abundant posttranslational modifications that are asymmetrically distributed in neurons. An increasing body of evidence supports the hypothesis that the combinatorial information expressed through tubulin genetic and chemical diversity controls microtubule dynamics, mechanics and interactions with microtubule effectors and thus constitutes a 'tubulin code'. Here we give a brief overview of tubulin isoform usage and posttranslational modifications in the neuron, and highlight recent progress in understanding the molecular mechanisms of the tubulin code.
Collapse
|
39
|
Qu X, Yuan FN, Corona C, Pasini S, Pero ME, Gundersen GG, Shelanski ML, Bartolini F. Stabilization of dynamic microtubules by mDia1 drives Tau-dependent Aβ 1-42 synaptotoxicity. J Cell Biol 2017; 216:3161-3178. [PMID: 28877993 PMCID: PMC5626542 DOI: 10.1083/jcb.201701045] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 06/15/2017] [Accepted: 07/26/2017] [Indexed: 01/17/2023] Open
Abstract
Oligomeric Amyloid β1-42 (Aβ) plays a crucial synaptotoxic role in Alzheimer's disease, and hyperphosphorylated tau facilitates Aβ toxicity. The link between Aβ and tau, however, remains controversial. In this study, we find that in hippocampal neurons, Aβ acutely induces tubulin posttranslational modifications (PTMs) and stabilizes dynamic microtubules (MTs) by reducing their catastrophe frequency. Silencing or acute inhibition of the formin mDia1 suppresses these activities and corrects the synaptotoxicity and deficits of axonal transport induced by Aβ. We explored the mechanism of rescue and found that stabilization of dynamic MTs promotes tau-dependent loss of dendritic spines and tau hyperphosphorylation. Collectively, these results uncover a novel role for mDia1 in Aβ-mediated synaptotoxicity and demonstrate that inhibition of MT dynamics and accumulation of PTMs are driving factors for the induction of tau-mediated neuronal damage.
Collapse
Affiliation(s)
- Xiaoyi Qu
- Department of Pathology, Anatomy and Cell Biology, Columbia University, New York, NY
| | - Feng Ning Yuan
- Department of Pathology, Anatomy and Cell Biology, Columbia University, New York, NY
| | - Carlo Corona
- Department of Pathology, Anatomy and Cell Biology, Columbia University, New York, NY
| | - Silvia Pasini
- Department of Pathology, Anatomy and Cell Biology, Columbia University, New York, NY
| | - Maria Elena Pero
- Department of Pathology, Anatomy and Cell Biology, Columbia University, New York, NY.,Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Naples, Italy
| | - Gregg G Gundersen
- Department of Pathology, Anatomy and Cell Biology, Columbia University, New York, NY
| | - Michael L Shelanski
- Department of Pathology, Anatomy and Cell Biology, Columbia University, New York, NY
| | - Francesca Bartolini
- Department of Pathology, Anatomy and Cell Biology, Columbia University, New York, NY
| |
Collapse
|
40
|
Axonal transport deficits in multiple sclerosis: spiraling into the abyss. Acta Neuropathol 2017; 134:1-14. [PMID: 28315956 PMCID: PMC5486629 DOI: 10.1007/s00401-017-1697-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Revised: 03/13/2017] [Accepted: 03/14/2017] [Indexed: 12/16/2022]
Abstract
The transport of mitochondria and other cellular components along the axonal microtubule cytoskeleton plays an essential role in neuronal survival. Defects in this system have been linked to a large number of neurological disorders. In multiple sclerosis (MS) and associated models such as experimental autoimmune encephalomyelitis (EAE), alterations in axonal transport have been shown to exist before neurodegeneration occurs. Genome-wide association (GWA) studies have linked several motor proteins to MS susceptibility, while neuropathological studies have shown accumulations of proteins and organelles suggestive for transport deficits. A reduced effectiveness of axonal transport can lead to neurodegeneration through inhibition of mitochondrial motility, disruption of axoglial interaction or prevention of remyelination. In MS, demyelination leads to dysregulation of axonal transport, aggravated by the effects of TNF-alpha, nitric oxide and glutamate on the cytoskeleton. The combined effect of all these pathways is a vicious cycle in which a defective axonal transport system leads to an increase in ATP consumption through loss of membrane organization and a reduction in available ATP through inhibition of mitochondrial transport, resulting in even further inhibition of transport. The persistent activity of this positive feedback loop contributes to neurodegeneration in MS.
Collapse
|
41
|
Abstract
Microtubules are key cytoskeletal elements of all eukaryotic cells and are assembled of evolutionarily conserved α-tubulin-β-tubulin heterodimers. Despite their uniform structure, microtubules fulfill a large diversity of functions. A regulatory mechanism to control the specialization of the microtubule cytoskeleton is the 'tubulin code', which is generated by (i) expression of different α- and β-tubulin isotypes, and by (ii) post-translational modifications of tubulin. In this Cell Science at a Glance article and the accompanying poster, we provide a comprehensive overview of the molecular components of the tubulin code, and discuss the mechanisms by which these components contribute to the generation of functionally specialized microtubules.
Collapse
Affiliation(s)
- Sudarshan Gadadhar
- Institut Curie, PSL Research University, CNRS UMR3348, Orsay F-91405, France.,Université Paris Sud, Université Paris-Saclay, CNRS UMR3348, Orsay F-91405, France
| | - Satish Bodakuntla
- Institut Curie, PSL Research University, CNRS UMR3348, Orsay F-91405, France.,Université Paris Sud, Université Paris-Saclay, CNRS UMR3348, Orsay F-91405, France
| | - Kathiresan Natarajan
- Institut Curie, PSL Research University, CNRS UMR3348, Orsay F-91405, France.,Université Paris Sud, Université Paris-Saclay, CNRS UMR3348, Orsay F-91405, France
| | - Carsten Janke
- Institut Curie, PSL Research University, CNRS UMR3348, Orsay F-91405, France .,Université Paris Sud, Université Paris-Saclay, CNRS UMR3348, Orsay F-91405, France
| |
Collapse
|
42
|
Mechanisms of Chromosome Congression during Mitosis. BIOLOGY 2017; 6:biology6010013. [PMID: 28218637 PMCID: PMC5372006 DOI: 10.3390/biology6010013] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Revised: 01/07/2017] [Accepted: 01/28/2017] [Indexed: 12/13/2022]
Abstract
Chromosome congression during prometaphase culminates with the establishment of a metaphase plate, a hallmark of mitosis in metazoans. Classical views resulting from more than 100 years of research on this topic have attempted to explain chromosome congression based on the balance between opposing pulling and/or pushing forces that reach an equilibrium near the spindle equator. However, in mammalian cells, chromosome bi-orientation and force balance at kinetochores are not required for chromosome congression, whereas the mechanisms of chromosome congression are not necessarily involved in the maintenance of chromosome alignment after congression. Thus, chromosome congression and maintenance of alignment are determined by different principles. Moreover, it is now clear that not all chromosomes use the same mechanism for congressing to the spindle equator. Those chromosomes that are favorably positioned between both poles when the nuclear envelope breaks down use the so-called "direct congression" pathway in which chromosomes align after bi-orientation and the establishment of end-on kinetochore-microtubule attachments. This favors the balanced action of kinetochore pulling forces and polar ejection forces along chromosome arms that drive chromosome oscillatory movements during and after congression. The other pathway, which we call "peripheral congression", is independent of end-on kinetochore microtubule-attachments and relies on the dominant and coordinated action of the kinetochore motors Dynein and Centromere Protein E (CENP-E) that mediate the lateral transport of peripheral chromosomes along microtubules, first towards the poles and subsequently towards the equator. How the opposite polarities of kinetochore motors are regulated in space and time to drive congression of peripheral chromosomes only now starts to be understood. This appears to be regulated by position-dependent phosphorylation of both Dynein and CENP-E and by spindle microtubule diversity by means of tubulin post-translational modifications. This so-called "tubulin code" might work as a navigation system that selectively guides kinetochore motors with opposite polarities along specific spindle microtubule populations, ultimately leading to the congression of peripheral chromosomes. We propose an integrated model of chromosome congression in mammalian cells that depends essentially on the following parameters: (1) chromosome position relative to the spindle poles after nuclear envelope breakdown; (2) establishment of stable end-on kinetochore-microtubule attachments and bi-orientation; (3) coordination between kinetochore- and arm-associated motors; and (4) spatial signatures associated with post-translational modifications of specific spindle microtubule populations. The physiological consequences of abnormal chromosome congression, as well as the therapeutic potential of inhibiting chromosome congression are also discussed.
Collapse
|
43
|
Vu HT, Akatsu H, Hashizume Y, Setou M, Ikegami K. Increase in α-tubulin modifications in the neuronal processes of hippocampal neurons in both kainic acid-induced epileptic seizure and Alzheimer's disease. Sci Rep 2017; 7:40205. [PMID: 28067280 PMCID: PMC5220350 DOI: 10.1038/srep40205] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 12/05/2016] [Indexed: 12/31/2022] Open
Abstract
Neurodegeneration includes acute changes and slow-developing alterations, both of which partly involve common cellular machinery. During neurodegeneration, neuronal processes are impaired along with dysregulated post-translational modifications (PTMs) of cytoskeletal proteins. In neuronal processes, tubulin undergoes unique PTMs including a branched form of modification called glutamylation and loss of the C-terminal tyrosine residue and the penultimate glutamic acid residue forming Δ2-tubulin. Here, we investigated the state of two PTMs, glutamylation and Δ2 form, in both acute and slow-developing neurodegenerations, using a newly generated monoclonal antibody, DTE41, which had 2-fold higher affinity to glutamylated Δ2-tubulin, than to unmodified Δ2-tubulin. DTE41 recognised glutamylated Δ2-tubulin preferentially in immunostaining than in enzyme-linked immunosorbent assay and immunoblotting. In normal mouse brain, DTE41 stained molecular layer of the cerebellum as well as synapse-rich regions in pyramidal neurons of the cerebral cortex. In kainic acid-induced epileptic seizure, DTE41-labelled signals were increased in the hippocampal CA3 region, especially in the stratum lucidum. In the hippocampi of post-mortem patients with Alzheimer's disease, intensities of DTE41 staining were increased in mossy fibres in the CA3 region as well as in apical dendrites of the pyramidal neurons. Our findings indicate that glutamylation on Δ2-tubulin is increased in both acute and slow-developing neurodegeneration.
Collapse
Affiliation(s)
- Hang Thi Vu
- Department of Cellular and Molecular Anatomy, and International Mass Imaging Center, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Hiroyasu Akatsu
- Choju Medical Institute, Fukushimura Hospital, Toyohashi, Japan
- Department of Medicine for Aging in Place and Community-Based Medical Education, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | | | - Mitsutoshi Setou
- Department of Cellular and Molecular Anatomy, and International Mass Imaging Center, Hamamatsu University School of Medicine, Hamamatsu, Japan
- Department of Systems Molecular Anatomy, Preeminent Medical Photonics Education and Research Center, Hamamatsu University School of Medicine, Hamamatsu, Japan
- Department of Anatomy, The University of Hong Kong, Hong Kong, China
- Division of Neural Systematics, National Institute for Physiological Sciences, Okazaki, Japan
- Riken Center for Molecular Imaging Science, Kobe, Japan
| | - Koji Ikegami
- Department of Cellular and Molecular Anatomy, and International Mass Imaging Center, Hamamatsu University School of Medicine, Hamamatsu, Japan
| |
Collapse
|
44
|
Gornstein EL, Schwarz TL. Neurotoxic mechanisms of paclitaxel are local to the distal axon and independent of transport defects. Exp Neurol 2016; 288:153-166. [PMID: 27894788 DOI: 10.1016/j.expneurol.2016.11.015] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 11/22/2016] [Accepted: 11/23/2016] [Indexed: 11/15/2022]
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a dose-limiting side effect of paclitaxel and other chemotherapeutic agents. Paclitaxel binds and stabilizes microtubules, but the cellular mechanisms that underlie paclitaxel's neurotoxic effects are not well understood. We therefore used primary cultures of adult murine dorsal root ganglion neurons, the cell type affected in patients, to examine leading hypotheses to explain paclitaxel neurotoxicity. We address the role of microtubule hyperstabilization and its downstream effects. Paclitaxel administered at 10-50nM for 1-3days induced retraction bulbs at the tips of axons and arrested axon growth without triggering axon fragmentation or cell death. By correlating the toxic effects and microtubule stabilizing activity of structurally different microtubule stabilizing compounds, we confirmed that microtubule hyperstabilization, rather than an off-target effect, is the likely primary cause of paclitaxel neurotoxicity. We examined potential downstream consequences of microtubule hyperstabilization and found that changes in levels of tubulin posttranslational modifications, although present after paclitaxel exposure, are not implicated in the paclitaxel neurotoxicity we observed in the cultures. Additionally, defects in axonal transport were not implicated as an early, causative mechanism of paclitaxel's toxic effects on dorsal root ganglion neurons. By using microfluidic chambers to selectively treat different parts of the axon with paclitaxel, we found that the distal axon was primarily vulnerable to paclitaxel, indicating that paclitaxel acts directly on the distal axon to induce degenerative effects. Together, our findings point to local effects of microtubule hyperstabilization on the distal-most portion of the axon as an early mediator of paclitaxel neurotoxicity. Because sensory neurons have a unique and ongoing requirement for distal growth in order to reinnervate the epidermis as it turns over, we propose that the ability of paclitaxel to arrest their growth accounts for the selective vulnerability of sensory neurons to paclitaxel neurotoxicity.
Collapse
Affiliation(s)
- Erica L Gornstein
- The F.M. Kirby Neurobiology Center, Children's Hospital Boston, Boston, MA 02115, USA; Biological and Biomedical Sciences Program, Harvard Medical School, Boston, MA 02115, USA
| | - Thomas L Schwarz
- The F.M. Kirby Neurobiology Center, Children's Hospital Boston, Boston, MA 02115, USA.
| |
Collapse
|
45
|
Alvarez FJ. Gephyrin and the regulation of synaptic strength and dynamics at glycinergic inhibitory synapses. Brain Res Bull 2016; 129:50-65. [PMID: 27612963 DOI: 10.1016/j.brainresbull.2016.09.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 08/23/2016] [Accepted: 09/05/2016] [Indexed: 01/23/2023]
Abstract
Glycinergic synapses predominate in brainstem and spinal cord where they modulate motor and sensory processing. Their postsynaptic mechanisms have been considered rather simple because they lack a large variety of glycine receptor isoforms and have relatively simple postsynaptic densities at the ultrastructural level. However, this simplicity is misleading being their postsynaptic regions regulated by a variety of complex mechanisms controlling the efficacy of synaptic inhibition. Early studies suggested that glycinergic inhibitory strength and dynamics depend largely on structural features rather than on molecular complexity. These include regulation of the number of postsynaptic glycine receptors, their localization and the amount of co-localized GABAA receptors and GABA-glycine co-transmission. These properties we now know are under the control of gephyrin. Gephyrin is the first postsynaptic scaffolding protein ever discovered and it was recently found to display a large degree of variation and regulation by splice variants, posttranslational modifications, intracellular trafficking and interactions with the underlying cytoskeleton. Many of these mechanisms are governed by converging excitatory activity and regulate gephyrin oligomerization and receptor binding, the architecture of the postsynaptic density (and by extension the whole synaptic complex), receptor retention and stability. These newly uncovered molecular mechanisms define the size and number of gephyrin postsynaptic regions and the numbers and proportions of glycine and GABAA receptors contained within. All together, they control the emergence of glycinergic synapses of different strength and temporal properties to best match the excitatory drive received by each individual neuron or local dendritic compartment.
Collapse
Affiliation(s)
- Francisco J Alvarez
- Department of Physiology, Emory University, Atlanta, GA 30322-3110, United States.
| |
Collapse
|
46
|
Biphasic Alteration of the Inhibitory Synapse Scaffold Protein Gephyrin in Early and Late Stages of an Alzheimer Disease Model. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:2279-91. [DOI: 10.1016/j.ajpath.2016.05.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 04/20/2016] [Accepted: 05/10/2016] [Indexed: 11/22/2022]
|
47
|
Barisic M, Maiato H. The Tubulin Code: A Navigation System for Chromosomes during Mitosis. Trends Cell Biol 2016; 26:766-775. [PMID: 27344407 DOI: 10.1016/j.tcb.2016.06.001] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 05/27/2016] [Accepted: 06/02/2016] [Indexed: 10/25/2022]
Abstract
Before chromosomes segregate during mitosis in metazoans, they align at the cell equator by a process known as chromosome congression. This is in part mediated by the coordinated activities of kinetochore motors with opposite directional preferences that transport peripheral chromosomes along distinct spindle microtubule populations. Because spindle microtubules are all made from the same α/β-tubulin heterodimers, a critical longstanding question has been how chromosomes are guided to specific locations during mitosis. This implies the existence of spatial cues/signals on specific spindle microtubules that are read by kinetochore motors on chromosomes and ultimately indicate the way towards the equator. Here, we discuss the emerging concept that tubulin post-translational modifications (PTMs), as part of the so-called tubulin code, work as a navigation system for kinetochore-based chromosome motility during early mitosis.
Collapse
Affiliation(s)
- Marin Barisic
- Danish Cancer Society Research Center, Cell Division Laboratory, Strandboulevarden 49, 2100 Copenhagen, Denmark.
| | - Helder Maiato
- Chromosome Instability and Dynamics Laboratory, Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; Instituto de Investigação e Inovação em Saúde - i3S, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; Cell Division Unit, Department of Experimental Biology, Faculdade de Medicina, Universidade do Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal.
| |
Collapse
|
48
|
Langlhofer G, Villmann C. The Intracellular Loop of the Glycine Receptor: It's not all about the Size. Front Mol Neurosci 2016; 9:41. [PMID: 27330534 PMCID: PMC4891346 DOI: 10.3389/fnmol.2016.00041] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2016] [Accepted: 05/17/2016] [Indexed: 11/15/2022] Open
Abstract
The family of Cys-loop receptors (CLRs) shares a high degree of homology and sequence identity. The overall structural elements are highly conserved with a large extracellular domain (ECD) harboring an α-helix and 10 β-sheets. Following the ECD, four transmembrane domains (TMD) are connected by intracellular and extracellular loop structures. Except the TM3–4 loop, their length comprises 7–14 residues. The TM3–4 loop forms the largest part of the intracellular domain (ICD) and exhibits the most variable region between all CLRs. The ICD is defined by the TM3–4 loop together with the TM1–2 loop preceding the ion channel pore. During the last decade, crystallization approaches were successful for some members of the CLR family. To allow crystallization, the intracellular loop was in most structures replaced by a short linker present in prokaryotic CLRs. Therefore, no structural information about the large TM3–4 loop of CLRs including the glycine receptors (GlyRs) is available except for some basic stretches close to TM3 and TM4. The intracellular loop has been intensively studied with regard to functional aspects including desensitization, modulation of channel physiology by pharmacological substances, posttranslational modifications, and motifs important for trafficking. Furthermore, the ICD interacts with scaffold proteins enabling inhibitory synapse formation. This review focuses on attempts to define structural and functional elements within the ICD of GlyRs discussed with the background of protein-protein interactions and functional channel formation in the absence of the TM3–4 loop.
Collapse
Affiliation(s)
- Georg Langlhofer
- Institute of Clinical Neurobiology, University of Würzburg Würzburg, Germany
| | - Carmen Villmann
- Institute of Clinical Neurobiology, University of Würzburg Würzburg, Germany
| |
Collapse
|
49
|
Deprez F, Pallotto M, Vogt F, Grabiec M, Virtanen MA, Tyagarajan SK, Panzanelli P, Fritschy JM. Postsynaptic gephyrin clustering controls the development of adult-born granule cells in the olfactory bulb. J Comp Neurol 2016; 523:1998-2016. [PMID: 25772192 DOI: 10.1002/cne.23776] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Revised: 03/06/2015] [Accepted: 03/09/2015] [Indexed: 01/01/2023]
Abstract
In adult rodent olfactory bulb, GABAergic signaling regulates migration, differentiation, and synaptic integration of newborn granule cells (GCs), migrating from the subventricular zone. Here we show that these effects depend on the formation of a postsynaptic scaffold organized by gephyrin-the main scaffolding protein of GABAergic synapses, which anchors receptors and signaling molecules to the postsynaptic density-and are regulated by the phosphorylation status of gephyrin. Using lentiviral vectors to selectively transfect adult-born GCs, we observed that overexpression of the phospho-deficient gephyrin mutant eGFP-gephyrin(S270A), which facilitates the formation of supernumerary GABAergic synapses in vitro, favors dendritic branching and the formation of transient GABAergic synapses on spines, identified by the presence of α2-GABAA Rs. In contrast, overexpression of the dominant-negative eGFP-gephyrin(L2B) (a chimera that is enzymatically active but clustering defective), curtailed dendritic growth, spine formation, and long-term survival of GCs, pointing to the essential role of gephyrin cluster formation for its function. We could exclude any gephyrin overexpression artifacts, as GCs infected with eGFP-gephyrin were comparable to those infected with eGFP alone. The opposite effects induced by the two gephyrin mutant constructs indicate that the gephyrin scaffold at GABAergic synapses orchestrates signaling cascades acting on the cytoskeleton to regulate neuronal growth and synapse formation. Specifically, gephyrin phosphorylation emerges as a novel mechanism regulating morphological differentiation and long-term survival of adult-born olfactory bulb neurons.
Collapse
Affiliation(s)
- Francine Deprez
- University of Zurich, Institute of Pharmacology and Toxicology, 8057, Zurich, Switzerland.,Neuroscience Center Zurich, ETH and University of Zurich, Switzerland
| | - Marta Pallotto
- Circuit Dynamics and Connectivity Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Fabia Vogt
- University of Zurich, Institute of Pharmacology and Toxicology, 8057, Zurich, Switzerland
| | - Marta Grabiec
- University of Zurich, Institute of Pharmacology and Toxicology, 8057, Zurich, Switzerland
| | - Mari A Virtanen
- Department of Neurosciences Fondamentales CMU, University of Geneva, 1211, Geneva, Switzerland
| | - Shiva K Tyagarajan
- University of Zurich, Institute of Pharmacology and Toxicology, 8057, Zurich, Switzerland.,Neuroscience Center Zurich, ETH and University of Zurich, Switzerland
| | - Patrizia Panzanelli
- University of Turin, Department of Neuroscience Rita Levi Montalcini, Turin, Italy
| | - Jean-Marc Fritschy
- University of Zurich, Institute of Pharmacology and Toxicology, 8057, Zurich, Switzerland.,Neuroscience Center Zurich, ETH and University of Zurich, Switzerland
| |
Collapse
|
50
|
Lenz M, Galanis C, Müller-Dahlhaus F, Opitz A, Wierenga CJ, Szabó G, Ziemann U, Deller T, Funke K, Vlachos A. Repetitive magnetic stimulation induces plasticity of inhibitory synapses. Nat Commun 2016; 7:10020. [PMID: 26743822 PMCID: PMC4729863 DOI: 10.1038/ncomms10020] [Citation(s) in RCA: 127] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 10/26/2015] [Indexed: 12/21/2022] Open
Abstract
Repetitive transcranial magnetic stimulation (rTMS) is used as a therapeutic tool in neurology and psychiatry. While repetitive magnetic stimulation (rMS) has been shown to induce plasticity of excitatory synapses, it is unclear whether rMS can also modify structural and functional properties of inhibitory inputs. Here we employed 10-Hz rMS of entorhinohippocampal slice cultures to study plasticity of inhibitory neurotransmission on CA1 pyramidal neurons. Our experiments reveal a rMS-induced reduction in GABAergic synaptic strength (2-4 h after stimulation), which is Ca(2+)-dependent and accompanied by the remodelling of postsynaptic gephyrin scaffolds. Furthermore, we present evidence that 10-Hz rMS predominantly acts on dendritic, but not somatic inhibition. Consistent with this finding, a reduction in clustered gephyrin is detected in CA1 stratum radiatum of rTMS-treated anaesthetized mice. These results disclose that rTMS induces coordinated Ca(2+)-dependent structural and functional changes of specific inhibitory postsynapses on principal neurons.
Collapse
Affiliation(s)
- Maximilian Lenz
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe-University, Frankfurt/Main 60590, Germany
| | - Christos Galanis
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe-University, Frankfurt/Main 60590, Germany
| | - Florian Müller-Dahlhaus
- Department of Neurology and Stroke and Hertie Institute for Clinical Brain Research, Eberhard-Karls-University, Tübingen 72076, Germany
| | - Alexander Opitz
- Center for Biomedical Imaging and Neuromodulation, Nathan Kline Institute for Psychiatric Research, Orangeburg, New York 10962, USA.,Center for the Developing Brain, Child Mind Institute, New York, New York 10022, USA
| | - Corette J Wierenga
- Division of Cell Biology, Faculty of Science, Utrecht University, Utrecht 3584 CH, The Netherlands
| | - Gábor Szabó
- Laboratory of Molecular Biology and Genetics, Institute of Experimental Medicine, Budapest H1083, Hungary
| | - Ulf Ziemann
- Department of Neurology and Stroke and Hertie Institute for Clinical Brain Research, Eberhard-Karls-University, Tübingen 72076, Germany
| | - Thomas Deller
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe-University, Frankfurt/Main 60590, Germany
| | - Klaus Funke
- Department of Neurophysiology, Medical Faculty, Ruhr-University, Bochum 44780, Germany
| | - Andreas Vlachos
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe-University, Frankfurt/Main 60590, Germany
| |
Collapse
|