1
|
Chhabra A, Hoffmann C, Pérez GA, Korobeinikov AA, Rentsch J, Hümpfer N, Kokwaro L, Gnidovec L, Petrovic A, Wallace JN, Tromm JV, Román-Vendrell C, Johnson EC, Ranković B, Perego E, Volpi T, Fernández-Busnadiego R, Köster S, Rizzoli SO, Ewers H, Morgan JR, Milovanovic D. Condensates of synaptic vesicles and synapsin are molecular beacons for actin sequestering and polymerization. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.07.19.604346. [PMID: 39071264 PMCID: PMC11275919 DOI: 10.1101/2024.07.19.604346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Neuronal communication relies on precisely maintained synaptic vesicle (SV) clusters, which assemble via liquid-liquid phase separation (LLPS). This process requires synapsins, the major synaptic phosphoproteins, which are known to bind actin. The reorganization of SVs, synapsins and actin is a hallmark of synaptic activity, but their interplay is still unclear. Here, we combined the reconstitution approaches, expansion microscopy, super-resolution imaging and cryo-electron tomography to dissect the roles of synapsin-SV condensates in the organization of the presynaptic actin cytoskeleton. Our data indicate that LLPS of synapsin initiates actin polymerization, allowing for SV:synapsin:actin assemblies to facilitate the mesoscale organization of SV clusters along axons mimicking the native presynaptic organization in both lamprey and mammalian synapses. Understanding the relationship between the actin network and synapsin-SVs condensates is an essential building block on a roadmap to unravel how coordinated neurotransmission along the axon enables circuit function and behavior.
Collapse
Affiliation(s)
- Akshita Chhabra
- Laboratory of Molecular Neuroscience, German Center for Neurodegenerative Diseases (DZNE), 10117 Berlin, Germany
- Whitman Center, Marine Biological Laboratory, 02543 Woods Hole, MA, USA
| | - Christian Hoffmann
- Laboratory of Molecular Neuroscience, German Center for Neurodegenerative Diseases (DZNE), 10117 Berlin, Germany
- Whitman Center, Marine Biological Laboratory, 02543 Woods Hole, MA, USA
| | - Gerard Aguilar Pérez
- Laboratory of Molecular Neuroscience, German Center for Neurodegenerative Diseases (DZNE), 10117 Berlin, Germany
- Whitman Center, Marine Biological Laboratory, 02543 Woods Hole, MA, USA
| | - Aleksandr A. Korobeinikov
- Laboratory of Molecular Neuroscience, German Center for Neurodegenerative Diseases (DZNE), 10117 Berlin, Germany
| | - Jakob Rentsch
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Nadja Hümpfer
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Linda Kokwaro
- Laboratory of Molecular Neuroscience, German Center for Neurodegenerative Diseases (DZNE), 10117 Berlin, Germany
- Einstein Center for Neuroscience, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, 10117 Berlin, Germany
| | - Luka Gnidovec
- Laboratory of Molecular Neuroscience, German Center for Neurodegenerative Diseases (DZNE), 10117 Berlin, Germany
| | - Arsen Petrovic
- Institute for Neuropathology, University Medical Center Göttingen, 37073, Göttingen, Germany
| | - Jaqulin N. Wallace
- The Eugene Bell Center for Regenerative Biology and Tissue Engineering, Marine Biological Laboratory, 02543 Woods Hole, MA, USA
| | - Johannes Vincent Tromm
- Laboratory of Molecular Neuroscience, German Center for Neurodegenerative Diseases (DZNE), 10117 Berlin, Germany
- Whitman Center, Marine Biological Laboratory, 02543 Woods Hole, MA, USA
- The Eugene Bell Center for Regenerative Biology and Tissue Engineering, Marine Biological Laboratory, 02543 Woods Hole, MA, USA
| | - Cristina Román-Vendrell
- The Eugene Bell Center for Regenerative Biology and Tissue Engineering, Marine Biological Laboratory, 02543 Woods Hole, MA, USA
| | - Emma C. Johnson
- The Eugene Bell Center for Regenerative Biology and Tissue Engineering, Marine Biological Laboratory, 02543 Woods Hole, MA, USA
| | - Branislava Ranković
- Laboratory of Molecular Neuroscience, German Center for Neurodegenerative Diseases (DZNE), 10117 Berlin, Germany
- Whitman Center, Marine Biological Laboratory, 02543 Woods Hole, MA, USA
| | - Eleonora Perego
- Institute for X-Ray Physics, University of Göttingen, Friedrich-Hund-Platz 1, 37077 Göttingen, Germany
| | - Tommaso Volpi
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT 06520, USA
| | | | - Sarah Köster
- Institute for X-Ray Physics, University of Göttingen, Friedrich-Hund-Platz 1, 37077 Göttingen, Germany
| | - Silvio O. Rizzoli
- Institute for Neuro- and Sensory Physiology, University Medical Center Göttingen, 37073, Göttingen, Germany
| | - Helge Ewers
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Jennifer R. Morgan
- The Eugene Bell Center for Regenerative Biology and Tissue Engineering, Marine Biological Laboratory, 02543 Woods Hole, MA, USA
| | - Dragomir Milovanovic
- Laboratory of Molecular Neuroscience, German Center for Neurodegenerative Diseases (DZNE), 10117 Berlin, Germany
- Whitman Center, Marine Biological Laboratory, 02543 Woods Hole, MA, USA
- Einstein Center for Neuroscience, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, 10117 Berlin, Germany
| |
Collapse
|
2
|
Bansal VA, Tan JM, Soon HR, Zainolabidin N, Saido T, Ch'ng TH. Aβ-driven nuclear pore complex dysfunction alters activation of necroptosis proteins in a mouse model of Alzheimer's disease. eLife 2025; 13:RP92069. [PMID: 40132021 PMCID: PMC11936419 DOI: 10.7554/elife.92069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2025] Open
Abstract
The emergence of Aβ pathology is one of the hallmarks of Alzheimer's disease (AD), but the mechanisms and impact of Aβ in progression of the disease is unclear. The nuclear pore complex (NPC) is a multi-protein assembly in mammalian cells that regulates movement of macromolecules across the nuclear envelope; its function is shown to undergo age-dependent decline during normal aging and is also impaired in multiple neurodegenerative disorders. Yet not much is known about the impact of Aβ on NPC function in neurons. Here, we examined NPC and nucleoporin (NUP) distribution and nucleocytoplasmic transport using a mouse model of AD (AppNL-G-F/NL-G-F) that expresses Aβ in young animals. Our studies revealed that a time-dependent accumulation of intracellular Aβ corresponded with a reduction of NPCs and NUPs in the nuclear envelope which resulted in the degradation of the permeability barrier and inefficient segregation of nucleocytoplasmic proteins, and active transport. As a result of the NPC dysfunction App KI neurons become more vulnerable to inflammation-induced necroptosis - a programmed cell death pathway where the core components are activated via phosphorylation through nucleocytoplasmic shutting. Collectively, our data implicates Aβ in progressive impairment of nuclear pore function and further confirms that the protein complex is vulnerable to disruption in various neurodegenerative diseases and is a potential therapeutic target.
Collapse
Affiliation(s)
| | - Jia Min Tan
- Lee Kong Chian School of Medicine, Nanyang Technological UniversitySingaporeSingapore
- School of Biological Science, Nanyang Technological UniversitySingaporeSingapore
| | - Hui Rong Soon
- Lee Kong Chian School of Medicine, Nanyang Technological UniversitySingaporeSingapore
- School of Biological Science, Nanyang Technological UniversitySingaporeSingapore
| | | | - Takaomi Saido
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical SciencesNagoyaJapan
| | - Toh Hean Ch'ng
- Lee Kong Chian School of Medicine, Nanyang Technological UniversitySingaporeSingapore
- School of Biological Science, Nanyang Technological UniversitySingaporeSingapore
| |
Collapse
|
3
|
Xiao X, Yang A, Zhang H, Park D, Wang Y, Szabo B, Boyden ES, Piatkevich KD. Engineering of Genetically Encoded Bright Near-Infrared Fluorescent Voltage Indicator. Int J Mol Sci 2025; 26:1442. [PMID: 40003908 PMCID: PMC11855178 DOI: 10.3390/ijms26041442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 01/30/2025] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Abstract
Genetically encoded voltage indicators (GEVIs) allow for the cell-type-specific real-time imaging of neuronal membrane potential dynamics, which is essential to understanding neuronal information processing at both cellular and circuit levels. Among GEVIs, near-infrared-shifted GEVIs offer faster kinetics, better tissue penetration, and compatibility with optogenetic tools, enabling all-optical electrophysiology in complex biological contexts. In our previous work, we employed the directed molecular evolution of microbial rhodopsin Archaerhodopsin-3 (Arch-3) in mammalian cells to develop a voltage sensor called Archon1. Archon1 demonstrated excellent membrane localization, signal-to-noise ratio (SNR), sensitivity, kinetics, and photostability, and full compatibility with optogenetic tools. However, Archon1 suffers from low brightness and requires high illumination intensities, which leads to tissue heating and phototoxicity during prolonged imaging. In this study, we aim to improve the brightness of this voltage sensor. We performed random mutation on a bright Archon derivative and identified a novel variant, monArch, which exhibits satisfactory voltage sensitivity (4~5% ΔF/FAP) and a 9-fold increase in basal brightness compared with Archon1. However, it is hindered by suboptimal membrane localization and compromised voltage sensitivity. These challenges underscore the need for continued optimization to achieve an optimal balance of brightness, stability, and functionality in rhodopsin-based voltage sensors.
Collapse
Affiliation(s)
- Xian Xiao
- School of Life Sciences, Westlake University, Hangzhou 310024, China; (X.X.); (H.Z.); (Y.W.)
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, China
| | - Aimei Yang
- McGovern Institute for Brain Research, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA; (A.Y.); (D.P.)
| | - Hanbin Zhang
- School of Life Sciences, Westlake University, Hangzhou 310024, China; (X.X.); (H.Z.); (Y.W.)
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, China
| | - Demian Park
- McGovern Institute for Brain Research, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA; (A.Y.); (D.P.)
| | - Yangdong Wang
- School of Life Sciences, Westlake University, Hangzhou 310024, China; (X.X.); (H.Z.); (Y.W.)
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, China
| | - Balint Szabo
- CellSorter KFT, H-1117 Budapest, Hungary;
- Department of Biological Physics, Eötvös Loránd University (ELTE), H-1053 Budapest, Hungary
| | - Edward S. Boyden
- McGovern Institute for Brain Research, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA; (A.Y.); (D.P.)
- Howard Hughes Medical Institute, Cambridge, MA 01239, USA
- Center for Neurobiological Engineering, K. Lisa Yang Center for Bionics, MIT, Cambridge, MA 01239, USA
- Department of Media Arts and Sciences, MIT, Cambridge, MA 01239, USA
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA 01239, USA
- Department of Biological Engineering, MIT, Cambridge, MA 01239, USA
- Koch Institute, MIT, Cambridge, MA 01239, USA
| | - Kiryl D. Piatkevich
- School of Life Sciences, Westlake University, Hangzhou 310024, China; (X.X.); (H.Z.); (Y.W.)
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, China
| |
Collapse
|
4
|
Shim MS, Liton PB. Time-Lapse Live-Cell Imaging Using Fluorescent Protein Sensors in Outflow Pathway Cells Under Fluid Flow Conditions. Methods Mol Biol 2025; 2858:77-86. [PMID: 39433668 PMCID: PMC11803581 DOI: 10.1007/978-1-0716-4140-8_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2024]
Abstract
The role of shear stress in regulating aqueous humor (AH) outflow and intraocular pressure (IOP) in the trabecular meshwork (TM) and Schlemm's canal (SC) of the eye is an emerging field. Shear stress has been shown to activate mechanosensitive ion channels in TM cells and induce nitric oxide production in SC cells, which can affect outflow resistance and lower IOP. Live-cell imaging using fluorescent protein sensors has provided real-time data to investigate the physiological relationship between fluid flow and shear stress in the outflow pathway cells. The successful application of time-lapse live-cell imaging in primary cultured cells has led to the identification of key cellular and molecular mechanisms involved in regulating AH outflow and IOP, including the role of autophagy and primary cilia as mechanosensors. This chapter presents a detailed protocol for conducting time-lapse live-cell imaging under fluid flow conditions in the outflow pathway cells.
Collapse
Affiliation(s)
- Myoung Sup Shim
- Department of Ophthalmology, Duke University, Durham, NC, USA.
| | - Paloma B Liton
- Department of Ophthalmology, Duke University, Durham, NC, USA
| |
Collapse
|
5
|
Su F, Pfundstein G, Sah S, Zhang S, Keable R, Hagan DW, Sharpe LJ, Clemens KJ, Begg D, Phelps EA, Brown AJ, Leshchyns'ka I, Sytnyk V. Neuronal growth regulator 1 (NEGR1) promotes the synaptic targeting of glutamic acid decarboxylase 65 (GAD65). J Neurochem 2025; 169:e16279. [PMID: 39676071 DOI: 10.1111/jnc.16279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 11/15/2024] [Accepted: 11/20/2024] [Indexed: 12/17/2024]
Abstract
Neuronal growth regulator 1 (NEGR1) is a synaptic plasma membrane localized cell adhesion molecule implicated in a wide spectrum of psychiatric disorders. By RNAseq analysis of the transcriptomic changes in the brain of NEGR1-deficient mice, we found that NEGR1 deficiency affects the expression of the Gad2 gene. We show that glutamic acid decarboxylase 65 (GAD65), the Gad2 - encoded enzyme synthesizing the inhibitory neurotransmitter GABA on synaptic vesicles, accumulates non-synaptically in brains of NEGR1-deficient mice. The density of non-synaptic GAD65 accumulations is also increased in NEGR1 deficient cultured hypothalamic neurons, and this effect is rescued by re-expression of NEGR1. By using a novel biosensor of the plasma membrane attachment of GAD65, we demonstrate that GAD65 attaches to the plasma membrane. NEGR1 promotes palmitoylation-dependent clearance of GAD65 from the plasma membrane and targeting of GAD65 to plasma membrane-derived endocytic vesicles. In NEGR1 deficient cultured hypothalamic neurons, the synaptic and extrasynaptic levels of the plasma membrane attached GAD65 are increased, and the synaptic levels of GABA are reduced. NEGR1-deficient mice are characterized by reduced body weight, lower GABAergic synapse densities in the arcuate nucleus, and blunted responsiveness to the reinforcing effects of food rewards. Our results indicate that abnormalities in synaptic GABA synthesis can contribute to brain disorders associated with abnormal expression of NEGR1 in humans.
Collapse
Affiliation(s)
- Feifei Su
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, New South Wales, Australia
| | - Grant Pfundstein
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, New South Wales, Australia
| | - Saroj Sah
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, New South Wales, Australia
| | - Shuyue Zhang
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, New South Wales, Australia
| | - Ryan Keable
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, New South Wales, Australia
| | - D Walker Hagan
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida, USA
| | - Laura J Sharpe
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, New South Wales, Australia
| | - Kelly J Clemens
- School of Psychology, The University of New South Wales, Sydney, New South Wales, Australia
| | - Denovan Begg
- School of Psychology, The University of New South Wales, Sydney, New South Wales, Australia
| | - Edward A Phelps
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida, USA
| | - Andrew J Brown
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, New South Wales, Australia
| | - Iryna Leshchyns'ka
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, New South Wales, Australia
| | - Vladimir Sytnyk
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
6
|
Yokoyama K, Hiraoka Y, Abe Y, Tanaka KF. Visualization of myelin-forming oligodendrocytes in the adult mouse brain. J Neurochem 2025; 169:e16218. [PMID: 39233334 PMCID: PMC11657928 DOI: 10.1111/jnc.16218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 08/10/2024] [Accepted: 08/20/2024] [Indexed: 09/06/2024]
Abstract
Oligodendrocyte (OL) differentiation from oligodendrocyte precursor cells (OPCs) is considered to result in two populations: premyelinating and myelinating OLs. Recent single-cell RNA sequence data subdivided these populations into newly formed (NFOLs), myelin-forming (MFOLs), and mature (MOLs) oligodendrocytes. However, which newly proposed population corresponds to premyelinating or myelinating OLs is unknown. We focused on the NFOL-specific long non-coding oligodendrocyte 1 gene (LncOL1) and sought to label NFOLs under the control of the LncOL1 promoter using a tetracycline-controllable gene induction system. We demonstrated that LncOL1 was expressed by premyelinating OLs and that the MFOL-specific gene, Ctps, was not, indicating that NFOLs correspond to premyelinating OLs and that MFOLs and MOLs correspond to myelinating OLs. We then generated a LncOL1-tTA mouse in which a tetracycline transactivator (tTA) cassette was inserted downstream from the LncOL1 transcription initiation site. By crossing the LncOL1-tTA mice with tetO reporter mice, we generated LncOL1-tTA::tetO-yellow fluorescent protein (YFP) double-transgenic (LncOL1-YFP) mice. Although LncOL1 is non-coding, YFP was detected in LncOL1-YFP mice, indicating successful tTA translation. Unexpectedly, we found that the morphology of LncOL1-tTA-driven YFP+ cells was distinct from that of LncOL1+ premyelinating OLs and that the labeled cells instead appeared as myelinating OLs. We demonstrated from their RNA expression that YFP-labeled OLs were MFOLs, but not MOLs. Using the unique property of delayed YFP induction, we sought to determine whether MFOLs are constantly supplied from OPCs and differentiate into MOLs, or whether MFOLs pause their differentiation and sustain this stage in the adult brain. To achieve this objective, we irradiated adult LncOL1-YFP brains with X-rays to deplete dividing OPCs and their progeny. The irradiation extinguished YFP-labeled OLs, indicating that adult OPCs differentiated into MOLs during a single period. We established a new transgenic mouse line that genetically labels MFOLs, providing a reliable tool for investigating the dynamics of adult oligodendrogenesis.
Collapse
Affiliation(s)
- Kiichi Yokoyama
- Division of Brain SciencesInstitute for Advanced Medical Research, Keio University School of MedicineTokyoJapan
| | - Yuichi Hiraoka
- Laboratory of Molecular NeuroscienceMedical Research Institute, Tokyo Medical and Dental UniversityTokyoJapan
- Laboratory of Genome Editing for Biomedical ResearchMedical Research Institute, Tokyo Medical and Dental UniversityTokyoJapan
| | - Yoshifumi Abe
- Division of Brain SciencesInstitute for Advanced Medical Research, Keio University School of MedicineTokyoJapan
| | - Kenji F. Tanaka
- Division of Brain SciencesInstitute for Advanced Medical Research, Keio University School of MedicineTokyoJapan
| |
Collapse
|
7
|
Yu Z, Fu Q, Qiu T, Yang C, Lu M, Peng Q, Yang J, Hu Z. Role of Rab10 in cocaine-induced behavioral effects is associated with GABAB receptor membrane expression in the nucleus accumbens. Front Pharmacol 2024; 15:1496657. [PMID: 39669198 PMCID: PMC11635607 DOI: 10.3389/fphar.2024.1496657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Accepted: 11/11/2024] [Indexed: 12/14/2024] Open
Abstract
Aim Previous studies have demonstrated that Ras-related GTP-binding protein Rab10 (Rab10) plays a role in psychostimulant-induced behavioral effects. In this study, we showed that Rab10 in the nucleus accumbens (NAc) of male animals affects the development of cocaine-induced behavioral effects, which are associated with the plasma membrane expression of the GABAB heteroreceptor (GABABR). Methods We performed flow cytometry, immunoendocytosis, pHluorin activity analysis, electrophysiology analysis, and open-field testing to explore the role of Rab10 in modulating the membrane expression and function of GABABR and its regulatory effect on cocaine-induced behavioral effects. Results Transcriptomics analysis showed that Rab10 was elevated following acute cocaine treatment. Membrane levels of Rab10 increased within day 1 of the cocaine treatment, subsequently decreasing at later time points. Rab10 deficiency in NAc regions significantly increased cocaine-inhibited membrane GABABR levels and inhibited cocaine-induced hyperlocomotion and behavioral sensitization. In addition, GAD 67 + -expressing neurons from NAc regions treated with cocaine revealed a significant decrease in Rab10 membrane expression. Furthermore, NAc neuron-specific Rab10 knockout resulted in a significant increase in the cocaine-inhibited membrane expression of GABABR, along with increased miniature inhibitory postsynaptic current (mIPSC) amplitude and attenuation of baclofen-amplified Ca2+ influx. Conclusion These results uncover a new mechanism in which Rab10-GABABR signaling may serve as a potential pathway for regulating cocaine-induced behavioral effects.
Collapse
Affiliation(s)
- Zhuoxuan Yu
- The First Clinical Medical College, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Qiang Fu
- Department of Respiration, Department Two, Jiangxi Provincial People’s Hospital, Nanchang, Jiangxi, China
| | - Tianyun Qiu
- Department of Clinical Laboratory, Wuhan Hankou Hospital, Wuhan, Hubei, China
| | - Caidi Yang
- The First Clinical Medical College, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Mingfen Lu
- Department of Pathophysiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Qinghua Peng
- Department of Anesthesiology, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Jianhua Yang
- Department of Physiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Zhenzhen Hu
- Department of Pathophysiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
8
|
Maudes E, Jamet Z, Marmolejo L, Dalmau JO, Groc L. Positive Allosteric Modulation of NMDARs Prevents the Altered Surface Dynamics Caused by Patients' Antibodies. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2024; 11:e200261. [PMID: 38771989 PMCID: PMC11111324 DOI: 10.1212/nxi.0000000000200261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 03/27/2024] [Indexed: 05/23/2024]
Abstract
OBJECTIVES A positive allosteric modulator of the NMDAR, SGE-301, has been shown to reverse the alterations caused by the antibodies of patients with anti-NMDAR encephalitis (NMDARe). However, the mechanisms involved beyond receptor modulation are unclear. In this study, we aimed to investigate how this modulator affects NMDAR membrane dynamics. METHODS Cultured hippocampal neurons were treated with SGE-301 or vehicle, alongside with immunoglobulins G (IgG) from patients with NMDARe or healthy controls. NMDAR surface dynamics were assessed with single-molecule imaging by photoactivated localization microscopy. RESULTS NMDAR trajectories from neurons treated with SGE-301 were less confinement, with increased diffusion coefficients. This effect mainly occurred at synapses because extrasynaptic diffusion and confinement were minimally affected by SGE-301. Treatment with patients' IgG reduced NMDAR surface dynamics and increased their confinement. Remarkably, SGE-301 incubation antagonized patients' IgG effects in both synaptic and extrasynaptic membrane compartments, restoring diffusion and confinement values similar to those from neurons exposed to control IgG. DISCUSSION We demonstrate that SGE-301 upregulates NMDAR surface diffusion and antagonizes the pathogenic effects of patients' IgG on NMDAR membrane organization. These findings suggest a potential therapeutic strategy for NMDARe.
Collapse
Affiliation(s)
- Estibaliz Maudes
- From the Neuroimmunology Program (E.M., L.M., J.O.D.), Fundació Clinic per la Recerca Biomèdiques August Pi i Sunyer (FCRB-IDIBAPS), University of Barcelona, Spain; and University of Bordeaux (Z.J., L.G.), CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, Bordeaux, France
| | - Zoë Jamet
- From the Neuroimmunology Program (E.M., L.M., J.O.D.), Fundació Clinic per la Recerca Biomèdiques August Pi i Sunyer (FCRB-IDIBAPS), University of Barcelona, Spain; and University of Bordeaux (Z.J., L.G.), CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, Bordeaux, France
| | - Laura Marmolejo
- From the Neuroimmunology Program (E.M., L.M., J.O.D.), Fundació Clinic per la Recerca Biomèdiques August Pi i Sunyer (FCRB-IDIBAPS), University of Barcelona, Spain; and University of Bordeaux (Z.J., L.G.), CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, Bordeaux, France
| | - Josep O Dalmau
- From the Neuroimmunology Program (E.M., L.M., J.O.D.), Fundació Clinic per la Recerca Biomèdiques August Pi i Sunyer (FCRB-IDIBAPS), University of Barcelona, Spain; and University of Bordeaux (Z.J., L.G.), CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, Bordeaux, France
| | - Laurent Groc
- From the Neuroimmunology Program (E.M., L.M., J.O.D.), Fundació Clinic per la Recerca Biomèdiques August Pi i Sunyer (FCRB-IDIBAPS), University of Barcelona, Spain; and University of Bordeaux (Z.J., L.G.), CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, Bordeaux, France
| |
Collapse
|
9
|
Giusti SA, Pino NS, Pannunzio C, Ogando MB, Armando NG, Garrett L, Zimprich A, Becker L, Gimeno ML, Lukin J, Merino FL, Pardi MB, Pedroncini O, Di Mauro GC, Durner VG, Fuchs H, de Angelis MH, Patop IL, Turck CW, Deussing JM, Vogt Weisenhorn DM, Jahn O, Kadener S, Hölter SM, Brose N, Giesert F, Wurst W, Marin-Burgin A, Refojo D. A brain-enriched circular RNA controls excitatory neurotransmission and restricts sensitivity to aversive stimuli. SCIENCE ADVANCES 2024; 10:eadj8769. [PMID: 38787942 PMCID: PMC11122670 DOI: 10.1126/sciadv.adj8769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 04/22/2024] [Indexed: 05/26/2024]
Abstract
Circular RNAs (circRNAs) are a large class of noncoding RNAs. Despite the identification of thousands of circular transcripts, the biological significance of most of them remains unexplored, partly because of the lack of effective methods for generating loss-of-function animal models. In this study, we focused on circTulp4, an abundant circRNA derived from the Tulp4 gene that is enriched in the brain and synaptic compartments. By creating a circTulp4-deficient mouse model, in which we mutated the splice acceptor site responsible for generating circTulp4 without affecting the linear mRNA or protein levels, we were able to conduct a comprehensive phenotypic analysis. Our results demonstrate that circTulp4 is critical in regulating neuronal and brain physiology, modulating the strength of excitatory neurotransmission and sensitivity to aversive stimuli. This study provides evidence that circRNAs can regulate biologically relevant functions in neurons, with modulatory effects at multiple levels of the phenotype, establishing a proof of principle for the regulatory role of circRNAs in neural processes.
Collapse
Affiliation(s)
- Sebastian A. Giusti
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)–CONICET–Partner Institute of the Max Planck Society, Buenos Aires, Argentina
- Molecular Neurobiology, Max Planck Institute of Psychiatry, Munich, Germany
| | - Natalia S. Pino
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany
| | - Camila Pannunzio
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)–CONICET–Partner Institute of the Max Planck Society, Buenos Aires, Argentina
| | - Mora B. Ogando
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)–CONICET–Partner Institute of the Max Planck Society, Buenos Aires, Argentina
| | - Natalia G. Armando
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)–CONICET–Partner Institute of the Max Planck Society, Buenos Aires, Argentina
| | - Lillian Garrett
- German Mouse Clinic, Helmholtz Zentrum München, Munich, Germany
| | - Annemarie Zimprich
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany
- German Mouse Clinic, Helmholtz Zentrum München, Munich, Germany
- Chair of Developmental Genetics, Munich School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Lore Becker
- German Mouse Clinic, Helmholtz Zentrum München, Munich, Germany
| | - Maria L. Gimeno
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)–CONICET–Partner Institute of the Max Planck Society, Buenos Aires, Argentina
| | - Jeronimo Lukin
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)–CONICET–Partner Institute of the Max Planck Society, Buenos Aires, Argentina
| | - Florencia L. Merino
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)–CONICET–Partner Institute of the Max Planck Society, Buenos Aires, Argentina
| | - M. Belen Pardi
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)–CONICET–Partner Institute of the Max Planck Society, Buenos Aires, Argentina
| | - Olivia Pedroncini
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)–CONICET–Partner Institute of the Max Planck Society, Buenos Aires, Argentina
| | - Giuliana C. Di Mauro
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)–CONICET–Partner Institute of the Max Planck Society, Buenos Aires, Argentina
| | | | - Helmut Fuchs
- German Mouse Clinic, Helmholtz Zentrum München, Munich, Germany
| | - Martin Hrabe de Angelis
- German Mouse Clinic, Helmholtz Zentrum München, Munich, Germany
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany
| | | | - Christoph W. Turck
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
| | - Jan M. Deussing
- Molecular Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Daniela M. Vogt Weisenhorn
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany
- Chair of Developmental Genetics, Munich School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Olaf Jahn
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Georg-August-University, Göttingen, Germany
| | | | - Sabine M. Hölter
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany
| | - Nils Brose
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Florian Giesert
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany
| | - Wolfgang Wurst
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany
- Chair of Developmental Genetics, Munich School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE) Site Munich, Munich, Germany
| | - Antonia Marin-Burgin
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)–CONICET–Partner Institute of the Max Planck Society, Buenos Aires, Argentina
| | - Damian Refojo
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)–CONICET–Partner Institute of the Max Planck Society, Buenos Aires, Argentina
- Molecular Neurobiology, Max Planck Institute of Psychiatry, Munich, Germany
| |
Collapse
|
10
|
Zhou H, Bi GQ, Liu G. Intracellular magnesium optimizes transmission efficiency and plasticity of hippocampal synapses by reconfiguring their connectivity. Nat Commun 2024; 15:3406. [PMID: 38649706 PMCID: PMC11035601 DOI: 10.1038/s41467-024-47571-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 04/02/2024] [Indexed: 04/25/2024] Open
Abstract
Synapses at dendritic branches exhibit specific properties for information processing. However, how the synapses are orchestrated to dynamically modify their properties, thus optimizing information processing, remains elusive. Here, we observed at hippocampal dendritic branches diverse configurations of synaptic connectivity, two extremes of which are characterized by low transmission efficiency, high plasticity and coding capacity, or inversely. The former favors information encoding, pertinent to learning, while the latter prefers information storage, relevant to memory. Presynaptic intracellular Mg2+ crucially mediates the dynamic transition continuously between the two extreme configurations. Consequently, varying intracellular Mg2+ levels endow individual branches with diverse synaptic computations, thus modulating their ability to process information. Notably, elevating brain Mg2+ levels in aging animals restores synaptic configuration resembling that of young animals, coincident with improved learning and memory. These findings establish intracellular Mg2+ as a crucial factor reconfiguring synaptic connectivity at dendrites, thus optimizing their branch-specific properties in information processing.
Collapse
Affiliation(s)
- Hang Zhou
- Faculty of Life and Health Sciences, Shenzhen University of Advanced Technology, Shenzhen, 518107, China.
- Interdisciplinary Center for Brain Information, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| | - Guo-Qiang Bi
- Faculty of Life and Health Sciences, Shenzhen University of Advanced Technology, Shenzhen, 518107, China
- Interdisciplinary Center for Brain Information, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- Shenzhen-Hong Kong Institute of Brain Science, Shenzhen, 518055, China
- Hefei National Laboratory for Physical Sciences at the Microscale, and School of Life Sciences, University of Science and Technology of China, Hefei, 230031, China
| | - Guosong Liu
- School of Medicine, Tsinghua University, Beijing, 100084, China.
- NeuroCentria Inc., Walnut Creek, CA, 94596, USA.
| |
Collapse
|
11
|
Moreno-Aguilera M, Neher AM, Mendoza MB, Dodel M, Mardakheh FK, Ortiz R, Gallego C. KIS counteracts PTBP2 and regulates alternative exon usage in neurons. eLife 2024; 13:e96048. [PMID: 38597390 PMCID: PMC11045219 DOI: 10.7554/elife.96048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 04/09/2024] [Indexed: 04/11/2024] Open
Abstract
Alternative RNA splicing is an essential and dynamic process in neuronal differentiation and synapse maturation, and dysregulation of this process has been associated with neurodegenerative diseases. Recent studies have revealed the importance of RNA-binding proteins in the regulation of neuronal splicing programs. However, the molecular mechanisms involved in the control of these splicing regulators are still unclear. Here, we show that KIS, a kinase upregulated in the developmental brain, imposes a genome-wide alteration in exon usage during neuronal differentiation in mice. KIS contains a protein-recognition domain common to spliceosomal components and phosphorylates PTBP2, counteracting the role of this splicing factor in exon exclusion. At the molecular level, phosphorylation of unstructured domains within PTBP2 causes its dissociation from two co-regulators, Matrin3 and hnRNPM, and hinders the RNA-binding capability of the complex. Furthermore, KIS and PTBP2 display strong and opposing functional interactions in synaptic spine emergence and maturation. Taken together, our data uncover a post-translational control of splicing regulators that link transcriptional and alternative exon usage programs in neuronal development.
Collapse
Affiliation(s)
| | - Alba M Neher
- Molecular Biology Institute of Barcelona (IBMB), CSICBarcelonaSpain
| | - Mónica B Mendoza
- Molecular Biology Institute of Barcelona (IBMB), CSICBarcelonaSpain
| | - Martin Dodel
- Barts Cancer Institute, Queen Mary University of LondonLondonUnited Kingdom
| | - Faraz K Mardakheh
- Barts Cancer Institute, Queen Mary University of LondonLondonUnited Kingdom
| | - Raúl Ortiz
- Molecular Biology Institute of Barcelona (IBMB), CSICBarcelonaSpain
| | - Carme Gallego
- Molecular Biology Institute of Barcelona (IBMB), CSICBarcelonaSpain
| |
Collapse
|
12
|
Park J, Evangelopoulos M, Vasher MK, Kudruk S, Ramani N, Mayer V, Solivan AC, Lee A, Mirkin CA. Enhancing Endosomal Escape and Gene Regulation Activity for Spherical Nucleic Acids. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2306902. [PMID: 37932003 PMCID: PMC10947971 DOI: 10.1002/smll.202306902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 10/19/2023] [Indexed: 11/08/2023]
Abstract
The therapeutic potential of small interfering RNAs (siRNAs) is limited by their poor stability and low cellular uptake. When formulated as spherical nucleic acids (SNAs), siRNAs are resistant to nuclease degradation and enter cells without transfection agents with enhanced activity compared to their linear counterparts; however, the gene silencing activity of SNAs is limited by endosomal entrapment, a problem that impacts many siRNA-based nanoparticle constructs. To increase cytosolic delivery, SNAs are formulated using calcium chloride (CaCl2 ) instead of the conventionally used sodium chloride (NaCl). The divalent calcium (Ca2+ ) ions remain associated with the multivalent SNA and have a higher affinity for SNAs compared to their linear counterparts. Importantly, confocal microscopy studies show a 22% decrease in the accumulation of CaCl2 -salted SNAs within the late endosomes compared to NaCl-salted SNAs, indicating increased cytosolic delivery. Consistent with this finding, CaCl2 -salted SNAs comprised of siRNA and antisense DNA all exhibit enhanced gene silencing activity (up to 20-fold), compared to NaCl-salted SNAs regardless of sequence or cell line (U87-MG and SK-OV-3) studied. Moreover, CaCl2 -salted SNA-based forced intercalation probes show improved cytosolic mRNA detection.
Collapse
Affiliation(s)
- Jungsoo Park
- Interdisciplinary Biological Sciences Graduate Program, Northwestern University, Evanston, Illinois, 60208, United States
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois, 60208, United States
| | - Michael Evangelopoulos
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois, 60208, United States
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois, 60208, United States
| | - Matthew K. Vasher
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois, 60208, United States
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois, 60208, United States
| | - Sergej Kudruk
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois, 60208, United States
- Department of Chemistry, Northwestern University, Evanston, Illinois, 60208, United States
| | - Namrata Ramani
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois, 60208, United States
- Department of Material Sciences and Engineering, Northwestern University, Evanston, Illinois, 60208, United States
| | - Vinzenz Mayer
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois, 60208, United States
- Department of Chemistry, Northwestern University, Evanston, Illinois, 60208, United States
| | - Alexander C. Solivan
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois, 60208, United States
- Department of Chemistry, Northwestern University, Evanston, Illinois, 60208, United States
| | - Andrew Lee
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois, 60208, United States
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, Illinois, 60208
| | - Chad A. Mirkin
- Interdisciplinary Biological Sciences Graduate Program, Northwestern University, Evanston, Illinois, 60208, United States
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois, 60208, United States
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois, 60208, United States
- Department of Chemistry, Northwestern University, Evanston, Illinois, 60208, United States
- Department of Material Sciences and Engineering, Northwestern University, Evanston, Illinois, 60208, United States
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, Illinois, 60208
| |
Collapse
|
13
|
Jiang ZF, Xuan LN, Sun XW, Liu SB, Yin J. Knockdown of SIK3 in the CA1 Region can Reduce Seizure Susceptibility in Mice by Inhibiting Decreases in GABA AR α1 Expression. Mol Neurobiol 2024; 61:1404-1416. [PMID: 37715891 DOI: 10.1007/s12035-023-03630-2] [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: 03/07/2023] [Accepted: 08/30/2023] [Indexed: 09/18/2023]
Abstract
Imbalance between excitation and inhibition is an important cause of epilepsy. Salt-inducible kinase 1 (SIK1) gene mutation can cause epilepsy. In this study, we first found that the expression of SIK3 is increased after epilepsy. Furthermore, the role of SIK3 in epilepsy was explored. In cultured hippocampal neurons, we used Pterosin B, a selective SIK3 inhibitor that can inhibit epileptiform discharges induced by the convulsant drug cyclothiazide (a positive allosteric modulator of AMPA receptors, CTZ). Knockdown of SIK3 inhibited epileptiform discharges and increased the amplitude of miniature inhibitory postsynaptic currents (mIPSCs). In mice, knockdown of SIK3 reduced epilepsy susceptibility in a pentylenetetrazole (a GABAA receptor antagonist, PTZ) acute kindling experiment and increased the expression of GABAA receptor α1. In conclusion, our results suggest that blockade or knockdown of SIK3 can inhibit epileptiform discharges and that SIK3 has the potential to be a novel target for epilepsy treatment.
Collapse
Affiliation(s)
- Zhen-Fu Jiang
- Dalian Medical University, Dalian, 116044, Liaoning, China.
- Department of Neurosurgery, the Second Affiliated Hospital of Dalian Medical University, 467 Zhongshan Road, Shahekou, Dalian, 116023, Liaoning, China.
| | - Li-Na Xuan
- Dalian Medical University, Dalian, 116044, Liaoning, China
| | - Xiao-Wan Sun
- East China Normal University, Shanghai, 200241, China
| | - Shao-Bo Liu
- Dalian Medical University, Dalian, 116044, Liaoning, China
| | - Jian Yin
- Dalian Medical University, Dalian, 116044, Liaoning, China.
- Department of Neurosurgery, the Second Affiliated Hospital of Dalian Medical University, 467 Zhongshan Road, Shahekou, Dalian, 116023, Liaoning, China.
| |
Collapse
|
14
|
Chao YW, Lee YL, Tseng CS, Wang LUH, Hsia KC, Chen H, Fustin JM, Azeem S, Chang TT, Chen CY, Kung FC, Hsueh YP, Huang YS, Chao HW. Improved CaP Nanoparticles for Nucleic Acid and Protein Delivery to Neural Primary Cultures and Stem Cells. ACS NANO 2024; 18:4822-4839. [PMID: 38285698 PMCID: PMC10867895 DOI: 10.1021/acsnano.3c09608] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 01/21/2024] [Accepted: 01/23/2024] [Indexed: 01/31/2024]
Abstract
Efficiently delivering exogenous materials into primary neurons and neural stem cells (NSCs) has long been a challenge in neurobiology. Existing methods have struggled with complex protocols, unreliable reproducibility, high immunogenicity, and cytotoxicity, causing a huge conundrum and hindering in-depth analyses. Here, we establish a cutting-edge method for transfecting primary neurons and NSCs, named teleofection, by a two-step process to enhance the formation of biocompatible calcium phosphate (CaP) nanoparticles. Teleofection enables both nucleic acid and protein transfection into primary neurons and NSCs, eliminating the need for specialized skills and equipment. It can easily fine-tune transfection efficiency by adjusting the incubation time and nanoparticle quantity, catering to various experimental requirements. Teleofection's versatility allows for the delivery of different cargos into the same cell culture, whether simultaneously or sequentially. This flexibility proves invaluable for long-term studies, enabling the monitoring of neural development and synapse plasticity. Moreover, teleofection ensures the consistent and robust expression of delivered genes, facilitating molecular and biochemical investigations. Teleofection represents a significant advancement in neurobiology, which has promise to transcend the limitations of current gene delivery methods. It offers a user-friendly, cost-effective, and reproducible approach for researchers, potentially revolutionizing our understanding of brain function and development.
Collapse
Affiliation(s)
- Yu-Wen Chao
- Department
of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110301, Taiwan
- Graduate
Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110301, Taiwan
| | - Yen-Lurk Lee
- Institute
of Molecular Biology, Academia Sinica, Taipei 115201, Taiwan
- Institute
of Biomedical Sciences, Academia Sinica, Taipei 115201, Taiwan
| | - Ching-San Tseng
- Department
of Anatomy, School of Medicine, China Medical
University, Taichung 40402, Taiwan
| | - Lily Ueh-Hsi Wang
- Institute
of Molecular Biology, Academia Sinica, Taipei 115201, Taiwan
| | - Kuo-Chiang Hsia
- Institute
of Molecular Biology, Academia Sinica, Taipei 115201, Taiwan
| | - Huatao Chen
- Department
of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
- Key
Laboratory of Animal Biotechnology of the Ministry of Agriculture
and Rural Affairs, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Jean-Michel Fustin
- The
University of Manchester, Faculty of Biology, Medicine and Health, Oxford Road, Manchester M13 9PL, U.K.
| | - Sayma Azeem
- Institute
of Biomedical Sciences, Academia Sinica, Taipei 115201, Taiwan
- Taiwan
International Graduate Program in Interdisciplinary Neuroscience, National Yang-Ming Chao-Tung University and Academia
Sinica, Taipei 115201, Taiwan
| | - Tzu-Tung Chang
- Institute
of Biomedical Sciences, Academia Sinica, Taipei 115201, Taiwan
| | - Chiung-Ya Chen
- Institute
of Molecular Biology, Academia Sinica, Taipei 115201, Taiwan
| | - Fan-Che Kung
- Institute
of Biomedical Sciences, Academia Sinica, Taipei 115201, Taiwan
| | - Yi-Ping Hsueh
- Institute
of Molecular Biology, Academia Sinica, Taipei 115201, Taiwan
| | - Yi-Shuian Huang
- Institute
of Biomedical Sciences, Academia Sinica, Taipei 115201, Taiwan
- Taiwan
International Graduate Program in Interdisciplinary Neuroscience, National Yang-Ming Chao-Tung University and Academia
Sinica, Taipei 115201, Taiwan
- Institute
of Molecular Medicine, College of Medicine, National Taiwan University, Taipei 10002, Taiwan
| | - Hsu-Wen Chao
- Department
of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110301, Taiwan
- Graduate
Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110301, Taiwan
- Department
of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| |
Collapse
|
15
|
Lilis P, Al Kabbani MA, Zempel H. Optimized Calcium-Phosphate-Based Co-transfection of Tau and tdTomato into Human iPSC-Derived Neurons for the Study of Intracellular Distribution of Wild-type and Mutant Human Tau. Methods Mol Biol 2024; 2754:551-560. [PMID: 38512689 DOI: 10.1007/978-1-0716-3629-9_32] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2024]
Abstract
The study of Tau protein in disease-relevant neuronal cells in culture requires efficient delivery systems for transfection of exogenous Tau and also modulators and interactors of Tau. Transfection of cultivated cells using calcium phosphate precipitation is a simple and cost-effective approach, also for difficult-to-transfect and sensitive cells such as primary neurons. Because of its low cell toxicity and ease of use, the Ca2+-phosphate transfection method is one of the most widely used gene transfer procedures in neuroscience. However, Ca2+-phosphate transfection efficacy in neurons is poor, often in the range of 1-5%, limiting its use in functional investigations. Here, we outline our improved Ca2+-phosphate transfection methodology for human iPSC-derived neurons that yields a reasonable efficiency (20-30% for bright volume markers) without apparent effects on cell health. We have used it to introduce wild-type and mutant human Tau with and without co-transfection of a volume marker (used here: tdTomato). In sum, our procedure can deliver neuronal genes (e.g., MAPT) using typical eukaryotic expression vectors (e.g., using CMV promoter) and is optimized for transfection of human iPSC-derived neurons.
Collapse
Affiliation(s)
- Panagiotis Lilis
- Institute for Human Genetics & Center for Molecular Medicine Cologne, University Hospital of Cologne, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Mohamed Aghyad Al Kabbani
- Institute for Human Genetics & Center for Molecular Medicine Cologne, University Hospital of Cologne, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Hans Zempel
- Institute for Human Genetics & Center for Molecular Medicine Cologne, University Hospital of Cologne, Faculty of Medicine, University of Cologne, Cologne, Germany.
| |
Collapse
|
16
|
Hoffmann C, Rentsch J, Tsunoyama TA, Chhabra A, Aguilar Perez G, Chowdhury R, Trnka F, Korobeinikov AA, Shaib AH, Ganzella M, Giannone G, Rizzoli SO, Kusumi A, Ewers H, Milovanovic D. Synapsin condensation controls synaptic vesicle sequestering and dynamics. Nat Commun 2023; 14:6730. [PMID: 37872159 PMCID: PMC10593750 DOI: 10.1038/s41467-023-42372-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 10/09/2023] [Indexed: 10/25/2023] Open
Abstract
Neuronal transmission relies on the regulated secretion of neurotransmitters, which are packed in synaptic vesicles (SVs). Hundreds of SVs accumulate at synaptic boutons. Despite being held together, SVs are highly mobile, so that they can be recruited to the plasma membrane for their rapid release during neuronal activity. However, how such confinement of SVs corroborates with their motility remains unclear. To bridge this gap, we employ ultrafast single-molecule tracking (SMT) in the reconstituted system of native SVs and in living neurons. SVs and synapsin 1, the most highly abundant synaptic protein, form condensates with liquid-like properties. In these condensates, synapsin 1 movement is slowed in both at short (i.e., 60-nm) and long (i.e., several hundred-nm) ranges, suggesting that the SV-synapsin 1 interaction raises the overall packing of the condensate. Furthermore, two-color SMT and super-resolution imaging in living axons demonstrate that synapsin 1 drives the accumulation of SVs in boutons. Even the short intrinsically-disordered fragment of synapsin 1 was sufficient to restore the native SV motility pattern in synapsin triple knock-out animals. Thus, synapsin 1 condensation is sufficient to guarantee reliable confinement and motility of SVs, allowing for the formation of mesoscale domains of SVs at synapses in vivo.
Collapse
Affiliation(s)
- Christian Hoffmann
- Laboratory of Molecular Neuroscience, German Center for Neurodegenerative Diseases (DZNE), 10117, Berlin, Germany
| | - Jakob Rentsch
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195, Berlin, Germany
| | - Taka A Tsunoyama
- Membrane Cooperativity Unit, Okinawa Institute of Science and Technology Graduate University (OIST); Onna-son, Okinawa, 904-0495, Japan
| | - Akshita Chhabra
- Laboratory of Molecular Neuroscience, German Center for Neurodegenerative Diseases (DZNE), 10117, Berlin, Germany
| | - Gerard Aguilar Perez
- Laboratory of Molecular Neuroscience, German Center for Neurodegenerative Diseases (DZNE), 10117, Berlin, Germany
| | - Rajdeep Chowdhury
- University Medical Center Göttingen, Institute for Neuro- and Sensory Physiology, Germany; Biostructural Imaging of Neurodegeneration (BIN) Center, Göttingen, Germany; Excellence Cluster Multiscale Bioimaging, 37073, Göttingen, Germany
| | - Franziska Trnka
- Laboratory of Molecular Neuroscience, German Center for Neurodegenerative Diseases (DZNE), 10117, Berlin, Germany
| | - Aleksandr A Korobeinikov
- Laboratory of Molecular Neuroscience, German Center for Neurodegenerative Diseases (DZNE), 10117, Berlin, Germany
| | - Ali H Shaib
- University Medical Center Göttingen, Institute for Neuro- and Sensory Physiology, Germany; Biostructural Imaging of Neurodegeneration (BIN) Center, Göttingen, Germany; Excellence Cluster Multiscale Bioimaging, 37073, Göttingen, Germany
| | - Marcelo Ganzella
- Department of Neurobiology, Max Planck Institute for Multidisciplinary Sciences, 37077, Göttingen, Germany
| | - Gregory Giannone
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, UMR 5297, F-33000, Bordeaux, France
| | - Silvio O Rizzoli
- University Medical Center Göttingen, Institute for Neuro- and Sensory Physiology, Germany; Biostructural Imaging of Neurodegeneration (BIN) Center, Göttingen, Germany; Excellence Cluster Multiscale Bioimaging, 37073, Göttingen, Germany
| | - Akihiro Kusumi
- Membrane Cooperativity Unit, Okinawa Institute of Science and Technology Graduate University (OIST); Onna-son, Okinawa, 904-0495, Japan
| | - Helge Ewers
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195, Berlin, Germany
| | - Dragomir Milovanovic
- Laboratory of Molecular Neuroscience, German Center for Neurodegenerative Diseases (DZNE), 10117, Berlin, Germany.
| |
Collapse
|
17
|
Zhu WH, Yang XX, Gou XZ, Fu SM, Chen JH, Gao F, Shen Y, Bi DL, Tang AH. Nanoscale reorganisation of synaptic proteins in Alzheimer's disease. Neuropathol Appl Neurobiol 2023; 49:e12924. [PMID: 37461203 DOI: 10.1111/nan.12924] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 05/30/2023] [Accepted: 06/24/2023] [Indexed: 08/31/2023]
Abstract
AIMS Synaptic strength depends strongly on the subsynaptic organisation of presynaptic transmitter release and postsynaptic receptor densities, and their alterations are expected to underlie pathologies. Although synaptic dysfunctions are common pathogenic traits of Alzheimer's disease (AD), it remains unknown whether synaptic protein nano-organisation is altered in AD. Here, we systematically characterised the alterations in the subsynaptic organisation in cellular and mouse models of AD. METHODS We used immunostaining and super-resolution stochastic optical reconstruction microscopy imaging to quantitatively examine the synaptic protein nano-organisation in both Aβ1-42-treated neuronal cultures and cortical sections from a mouse model of AD, APP23 mice. RESULTS We found that Aβ1-42-treatment of cultured hippocampal neurons decreased the synaptic retention of postsynaptic scaffolds and receptors and disrupted their nanoscale alignment to presynaptic transmitter release sites. In cortical sections, we found that while GluA1 receptors in wild-type mice were organised in subsynaptic nanoclusters with high local densities, receptors in APP23 mice distributed more homogeneously within synapses. This reorganisation, together with the reduced overall receptor density, led to reduced glutamatergic synaptic transmission. Meanwhile, the transsynaptic alignment between presynaptic release-guiding RIM1/2 and postsynaptic scaffolding protein PSD-95 was reduced in APP23 mice. Importantly, these reorganisations were progressive with age and were more pronounced in synapses in close vicinity of Aβ plaques with dense cores. CONCLUSIONS Our study revealed a spatiotemporal-specific reorganisation of synaptic nanostructures in AD and identifies dense-core amyloid plaques as the major local inductor in APP23 mice.
Collapse
Affiliation(s)
- Wang-Hui Zhu
- 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, China
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, China
| | - Xiao-Xu Yang
- 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, China
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Sciences and Technology of China, Hefei, China
- Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei, China
| | - Xu-Zhuo Gou
- 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, China
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, China
| | - Shu-Mei 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, China
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Sciences and Technology of China, Hefei, China
- Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei, China
| | - Jia-Hui Chen
- 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, China
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, China
- Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei, China
| | - Feng Gao
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Sciences and Technology of China, Hefei, China
| | - Yong Shen
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Sciences and Technology of China, Hefei, China
- Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei, China
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, China
| | - Dan-Lei Bi
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, China
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Sciences and Technology of China, Hefei, China
- Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei, China
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 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, China
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, China
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Sciences and Technology of China, Hefei, China
- Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei, China
| |
Collapse
|
18
|
Lu MF, Fu Q, Qiu TY, Yang JH, Peng QH, Hu ZZ. The CaMKII-dependent phosphorylation of GABA B receptors in the nucleus accumbens was involved in cocaine-induced behavioral sensitization in rats. CNS Neurosci Ther 2023; 29:1345-1356. [PMID: 36756679 PMCID: PMC10068462 DOI: 10.1111/cns.14107] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 01/16/2023] [Accepted: 01/18/2023] [Indexed: 02/10/2023] Open
Abstract
BACKGROUND Previous studies have established that the regulation of prolonged, distal neuronal inhibition by the GABAB heteroreceptor (GABAB R) is determined by its stability, and hence residence time, on the plasma membrane. AIMS Here, we show that GABAB R in the nucleus accumbens (NAc) of rats affects the development of cocaine-induced behavioral sensitization by mediating its perinucleus internalization and membrane expression. MATERIALS & METHODS By immunofluorescent labeling, flow cytometry analysis, Co-immunoprecipitation and open field test, we measured the role of Ca2+ /calmodulin-dependent protein kinase II (CaMKII) to the control of GABAB R membrane anchoring and cocaine induced-behavioral sensitization. RESULTS Repeated cocaine treatment in rats (15 mg/kg) significantly decreases membrane levels of GABAB1 R and GABAB2 R in the NAc after day 3, 5 and 7. The membrane fluorescence and protein levels of GABAB R was also decreased in NAc GAD67 + neurons post cocaine (1 μM) treatment after 5 min. Moreover, the majority of internalized GABAB1 Rs exhibited perinuclear localization, a decrease in GABAB1 R-pHluroin signals was observed in cocaine-treated NAc neurons. By contrast, membrane expression of phosphorylated CaMKII (pCaMKII) post cocaine treatment was significantly increased after day 1, 3, 5 and 7. Baclofen blocked the cocaine induced behavioral sensitization via inhibition of cocaine enhanced-pCaMKII-GABAB1 R interaction. CONCLUSION These findings reveal a new mechanism by which pCaMKII-GABAB R signaling can promote psychostimulant-induced behavioral sensitization.
Collapse
Affiliation(s)
- Ming F Lu
- Department of Pathophysiology, College of Basic Medicine, Nanchang University, Nanchang, Jiangxi, China.,Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology, Department of Pathology, Schools of Basic Medical Sciences and Pharmaceutical Sciences, Nanchang University, Nanchang, Jiangxi, China
| | - Qiang Fu
- Department of Respiration, Department Two, Jiangxi Provincial People's Hospital, Nanchang, Jiangxi, China
| | - Tian Y Qiu
- Department of Pathophysiology, College of Basic Medicine, Nanchang University, Nanchang, Jiangxi, China.,Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology, Department of Pathology, Schools of Basic Medical Sciences and Pharmaceutical Sciences, Nanchang University, Nanchang, Jiangxi, China
| | - Jian H Yang
- Department of Physiology, College of Basic Medicine, Nanchang University, Nanchang, Jiangxi, China
| | - Qing H Peng
- Department of Anesthesiology, The First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi, China
| | - Zhen Z Hu
- Department of Pathophysiology, College of Basic Medicine, Nanchang University, Nanchang, Jiangxi, China.,Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology, Department of Pathology, Schools of Basic Medical Sciences and Pharmaceutical Sciences, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
19
|
Yu J, Wang G, Chen Z, Wan L, Zhou J, Cai J, Liu X, Wang Y. Deficit of PKHD1L1 in the dentate gyrus increases seizure susceptibility in mice. Hum Mol Genet 2023; 32:506-519. [PMID: 36067019 DOI: 10.1093/hmg/ddac220] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/28/2022] [Accepted: 08/30/2022] [Indexed: 01/24/2023] Open
Abstract
Epilepsy is a chronic neurological disorder featuring recurrent, unprovoked seizures, which affect more than 65 million people worldwide. Here, we discover that the PKHD1L1, which is encoded by polycystic kidney and hepatic disease1-like 1 (Pkhd1l1), wildly distributes in neurons in the central nervous system (CNS) of mice. Disruption of PKHD1L1 in the dentate gyrus region of the hippocampus leads to increased susceptibility to pentylenetetrazol-induced seizures in mice. The disturbance of PKHD1L1 leads to the overactivation of the mitogen-activated protein kinase (MAPK)/extracellular regulated kinase (ERK)-Calpain pathway, which is accompanied by remarkable degradation of cytoplasmic potassium chloride co-transporter 2 (KCC2) level together with the impaired expression and function of membrane KCC2. However, the reduction of membrane KCC2 is associated with the damaged inhibitory ability of the vital GABA receptors, which ultimately leads to the significantly increased susceptibility to epileptic seizures. Our data, thus, indicate for the first time that Pkhd1l1, a newly discovered polycystic kidney disease (PKD) association gene, is required in neurons to maintain neuronal excitability by regulation of KCC2 expression in CNS. A new mechanism of the clinical association between genetic PKD and seizures has been built, which could be a potential therapeutic target for treating PKD-related seizures.
Collapse
Affiliation(s)
- Jiangning Yu
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institute of Biological Science, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Guoxiang Wang
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institute of Biological Science, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Zhiyun Chen
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institute of Biological Science, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Li Wan
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institute of Biological Science, Zhongshan Hospital, Fudan University, Shanghai 200032, China.,Rehabilitation Center, Shenzhen Second People's Hospital/the First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen 518035, China
| | - Jing Zhou
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institute of Biological Science, Zhongshan Hospital, Fudan University, Shanghai 200032, China.,Rehabilitation Center, Shenzhen Second People's Hospital/the First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen 518035, China
| | - Jingyi Cai
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institute of Biological Science, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xu Liu
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institute of Biological Science, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yun Wang
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institute of Biological Science, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
20
|
Michaluk P, Rusakov DA. Monitoring cell membrane recycling dynamics of proteins using whole-cell fluorescence recovery after photobleaching of pH-sensitive genetic tags. Nat Protoc 2022; 17:3056-3079. [PMID: 36064755 DOI: 10.1038/s41596-022-00732-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Accepted: 06/07/2022] [Indexed: 11/08/2022]
Abstract
Population behavior of signaling molecules on the cell surface is key to their adaptive function. Live imaging of proteins tagged with fluorescent molecules has been an essential tool in understanding this behavior. Typically, genetic or chemical tags are used to target molecules present throughout the cell, whereas antibody-based tags label the externally exposed molecular domains only. Both approaches could potentially overlook the intricate process of in-out membrane recycling in which target molecules appear or disappear on the cell surface. This limitation is overcome by using a pH-sensitive fluorescent tag, such as Super-Ecliptic pHluorin (SEP), because its emission depends on whether it resides inside or outside the cell. Here we focus on the main glial glutamate transporter GLT1 and describe a genetic design that equips GLT1 molecules with SEP without interfering with the transporter's main function. Expressing GLT1-SEP in astroglia in cultures or in hippocampal slices enables monitoring of the real-time dynamics of the cell-surface and cytosolic fractions of the transporter in living cells. Whole-cell fluorescence recovery after photobleaching and quantitative image-kinetic analysis of the resulting time-lapse images enables assessment of the rate of GLT1-SEP recycling on the cell surface, a fundamental trafficking parameter unattainable previously. The present protocol takes 15-20 d to set up cell preparations, and 2-3 d to carry out live cell experiments and data analyses. The protocol can be adapted to study different membrane molecules of interest, particularly those proteins whose lifetime on the cell surface is critical to their adaptive function.
Collapse
Affiliation(s)
- Piotr Michaluk
- UCL Queen Square Institute of Neurology, University College London, London, UK.
- BRAINCITY, Laboratory of Neurobiology, Nencki Institute of Experimental Biology PAS, Warsaw, Poland.
| | - Dmitri A Rusakov
- UCL Queen Square Institute of Neurology, University College London, London, UK.
| |
Collapse
|
21
|
Keable R, Hu S, Pfundstein G, Kozlova I, Su F, Du X, Yang H, Gunnersen J, Schachner M, Leshchyns'ka I, Sytnyk V. The BACE1-generated C-terminal fragment of the neural cell adhesion molecule 2 (NCAM2) promotes BACE1 targeting to Rab11-positive endosomes. Cell Mol Life Sci 2022; 79:555. [PMID: 36251052 PMCID: PMC9576659 DOI: 10.1007/s00018-022-04575-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 09/16/2022] [Accepted: 09/27/2022] [Indexed: 11/08/2022]
Abstract
Beta-site amyloid precursor protein cleaving enzyme 1 (BACE1), also known as β-secretase, is an aspartic protease. The sorting of this enzyme into Rab11-positive recycling endosomes regulates the BACE1-mediated cleavage of its substrates, however, the mechanisms underlying this targeting remain poorly understood. The neural cell adhesion molecule 2 (NCAM2) is a substrate of BACE1. We show that BACE1 cleaves NCAM2 in cultured hippocampal neurons and NCAM2-transfected CHO cells. The C-terminal fragment of NCAM2 that comprises the intracellular domain and a small portion of NCAM2’s extracellular domain, associates with BACE1. This association is not affected in cells with inhibited endocytosis, indicating that the interaction of NCAM2 and BACE1 precedes the targeting of BACE1 from the cell surface to endosomes. In neurons and CHO cells, this fragment and BACE1 co-localize in Rab11-positive endosomes. Overexpression of full-length NCAM2 or a recombinant NCAM2 fragment containing the transmembrane and intracellular domains but lacking the extracellular domain leads to an increase in BACE1 levels in these organelles. In NCAM2-deficient neurons, the levels of BACE1 are increased at the cell surface and reduced in intracellular organelles. These effects are correlated with increased levels of the soluble extracellular domain of BACE1 in the brains of NCAM2-deficient mice, suggesting increased shedding of BACE1 from the cell surface. Of note, shedding of the extracellular domain of Sez6, a protein cleaved exclusively by BACE1, is reduced in NCAM2-deficient animals. These results indicate that the BACE1-generated fragment of NCAM2 regulates BACE1 activity by promoting the targeting of BACE1 to Rab11-positive endosomes.
Collapse
Affiliation(s)
- Ryan Keable
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, NSW, 2052, Australia
| | - Shangfeng Hu
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, NSW, 2052, Australia
| | - Grant Pfundstein
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, NSW, 2052, Australia
| | - Irina Kozlova
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, NSW, 2052, Australia
| | - Feifei Su
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, NSW, 2052, Australia
| | - Ximing Du
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, NSW, 2052, Australia
| | - Hongyuan Yang
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, NSW, 2052, Australia
| | - Jenny Gunnersen
- Department of Anatomy and Physiology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Melitta Schachner
- Department of Cell Biology and Neuroscience, Keck Center for Collaborative Neuroscience, Rutgers University, Piscataway, NJ, 08554, USA
| | - Iryna Leshchyns'ka
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, NSW, 2052, Australia
| | - Vladimir Sytnyk
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, NSW, 2052, Australia.
| |
Collapse
|
22
|
Sharma G, Banerjee S. Activity-regulated E3 ubiquitin ligase TRIM47 modulates excitatory synapse development. Front Mol Neurosci 2022; 15:943980. [PMID: 36211980 PMCID: PMC9532517 DOI: 10.3389/fnmol.2022.943980] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 08/29/2022] [Indexed: 11/30/2022] Open
Abstract
The Ubiquitin Proteasome System (UPS) has been shown to regulate neuronal development and synapse formation. Activity-dependent regulation of E3 ligase, a component of the UPS that targets specific proteins for proteasome-mediated degradation, is emerging as a pivotal player for the establishment of functional synapses. Here, we identified TRIM47 as a developmentally regulated E3 ligase that is expressed in rat hippocampus during the temporal window of synapse formation. We have demonstrated that the expression of TRIM47 is regulated by the glutamate-induced synaptic activity of hippocampal neurons in culture. In addition, the activity-dependent enhancement of TRIM47 expression is recapitulated following the object location test, a hippocampus-dependent spatial memory paradigm. We observed that this enhancement of TRIM47 expression requires NMDA receptor activation. The knockdown of TRIM47 leads to an enhancement of spine density without affecting dendritic complexity. Furthermore, we observed an increase in excitatory synapse development upon loss of TRIM47 function. Comprehensively, our study identified an activity-regulated E3 ligase that drives excitatory synapse formation in hippocampal neurons.
Collapse
|
23
|
Hawkins S, Namboori SC, Tariq A, Blaker C, Flaxman C, Dey NS, Henley P, Randall A, Rosa A, Stanton LW, Bhinge A. Upregulation of β-catenin due to loss of miR-139 contributes to motor neuron death in amyotrophic lateral sclerosis. Stem Cell Reports 2022; 17:1650-1665. [PMID: 35750046 PMCID: PMC9287677 DOI: 10.1016/j.stemcr.2022.05.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 05/25/2022] [Accepted: 05/26/2022] [Indexed: 01/12/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by the loss of motor neurons (MNs). There are no effective treatments and patients usually die within 2-5 years of diagnosis. Emerging commonalities between familial and sporadic cases of this complex multifactorial disorder include disruption to RNA processing and cytoplasmic inclusion bodies containing TDP-43 and/or FUS protein aggregates. Both TDP-43 and FUS have been implicated in RNA processing functions, including microRNA biogenesis, transcription, and splicing. In this study, we explore the misexpression of microRNAs in an iPSC-based disease model of FUS ALS. We identify the downregulation of miR-139, an MN-enriched microRNA, in FUS and sporadic ALS MN. We discover that miR-139 downregulation leads to the activation of canonical WNT signaling and demonstrate that the WNT transcriptional mediator β-catenin is a major driver of MN degeneration in ALS. Our results highlight the importance of homeostatic RNA networks in ALS.
Collapse
Affiliation(s)
- Sophie Hawkins
- College of Medicine and Health, University of Exeter, Exeter EX1 2LU, UK; Living Systems Institute, University of Exeter, Exeter EX4 4QD, UK
| | - Seema C Namboori
- College of Medicine and Health, University of Exeter, Exeter EX1 2LU, UK; Living Systems Institute, University of Exeter, Exeter EX4 4QD, UK
| | - Ammarah Tariq
- Living Systems Institute, University of Exeter, Exeter EX4 4QD, UK; Biosciences, University of Exeter, Exeter EX4 4QD, UK
| | - Catherine Blaker
- College of Medicine and Health, University of Exeter, Exeter EX1 2LU, UK
| | - Christine Flaxman
- College of Medicine and Health, University of Exeter, Exeter EX1 2LU, UK
| | - Nidhi S Dey
- York Biomedical Research Institute, Hull York Medical School, University of York, York YO10 5DD, UK
| | - Peter Henley
- College of Medicine and Health, University of Exeter, Exeter EX1 2LU, UK
| | - Andrew Randall
- College of Medicine and Health, University of Exeter, Exeter EX1 2LU, UK
| | - Alessandro Rosa
- Department of Biology and Biotechnologies Charles Darwin, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy; Center for Life Nano- & Neuro-Science, Fondazione Istituto Italiano di Tecnologia (IIT), 00161 Rome, Italy
| | - Lawrence W Stanton
- Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Doha, Qatar
| | - Akshay Bhinge
- College of Medicine and Health, University of Exeter, Exeter EX1 2LU, UK; Living Systems Institute, University of Exeter, Exeter EX4 4QD, UK.
| |
Collapse
|
24
|
Elmasri M, Hunter DW, Winchester G, Bates EE, Aziz W, Van Der Does DM, Karachaliou E, Sakimura K, Penn AC. Common synaptic phenotypes arising from diverse mutations in the human NMDA receptor subunit GluN2A. Commun Biol 2022; 5:174. [PMID: 35228668 PMCID: PMC8885697 DOI: 10.1038/s42003-022-03115-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 01/31/2022] [Indexed: 02/06/2023] Open
Abstract
Dominant mutations in the human gene GRIN2A, encoding NMDA receptor (NMDAR) subunit GluN2A, make a significant and growing contribution to the catalogue of published single-gene epilepsies. Understanding the disease mechanism in these epilepsy patients is complicated by the surprising diversity of effects that the mutations have on NMDARs. Here we have examined the cell-autonomous effect of five GluN2A mutations, 3 loss-of-function and 2 gain-of-function, on evoked NMDAR-mediated synaptic currents (NMDA-EPSCs) in CA1 pyramidal neurons in cultured hippocampal slices. Despite the mutants differing in their functional incorporation at synapses, prolonged NMDA-EPSC current decays (with only marginal changes in charge transfer) were a common effect for both gain- and loss-of-function mutants. Modelling NMDA-EPSCs with mutant properties in a CA1 neuron revealed that the effect of GRIN2A mutations can lead to abnormal temporal integration and spine calcium dynamics during trains of concerted synaptic activity. Investigations beyond establishing the molecular defects of GluN2A mutants are much needed to understand their impact on synaptic transmission. The cell-autonomous effect of five severe loss- or gain-of-function GluN2A (NMDA receptor) mutations is assessed on evoked NMDAR-mediated synaptic currents in CA1 pyramidal neurons in cultured mouse hippocampal slices. Data and modelling suggest that mutant-like NMDA-EPSCs can lead to abnormal temporal summation and spine calcium dynamics.
Collapse
Affiliation(s)
- Marwa Elmasri
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton, BN1 9QG, UK
| | - Daniel William Hunter
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton, BN1 9QG, UK
| | - Giles Winchester
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton, BN1 9QG, UK
| | - Ella Emine Bates
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton, BN1 9QG, UK
| | - Wajeeha Aziz
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton, BN1 9QG, UK
| | | | - Eirini Karachaliou
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton, BN1 9QG, UK
| | - Kenji Sakimura
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, Niigata, 951-8585, Japan
| | - Andrew Charles Penn
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton, BN1 9QG, UK.
| |
Collapse
|
25
|
Zheng R, Du Y, Wang X, Liao T, Zhang Z, Wang N, Li X, Shen Y, Shi L, Luo J, Xia J, Wang Z, Xu J. KIF2C regulates synaptic plasticity and cognition in mice through dynamic microtubule depolymerization. eLife 2022; 11:e72483. [PMID: 35138249 PMCID: PMC8828051 DOI: 10.7554/elife.72483] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 02/01/2022] [Indexed: 11/13/2022] Open
Abstract
Dynamic microtubules play a critical role in cell structure and function. In nervous system, microtubules are the major route for cargo protein trafficking and they specially extend into and out of synapses to regulate synaptic development and plasticity. However, the detailed depolymerization mechanism that regulates dynamic microtubules in synapses and dendrites is still unclear. In this study, we find that KIF2C, a dynamic microtubule depolymerization protein without known function in the nervous system, plays a pivotal role in the structural and functional plasticity of synapses and regulates cognitive function in mice. Through its microtubule depolymerization capability, KIF2C regulates microtubule dynamics in dendrites, and regulates microtubule invasion of spines in neurons in a neuronal activity-dependent manner. Using RNAi knockdown and conditional knockout approaches, we showed that KIF2C regulates spine morphology and synaptic membrane expression of AMPA receptors. Moreover, KIF2C deficiency leads to impaired excitatory transmission, long-term potentiation, and altered cognitive behaviors in mice. Collectively, our study explores a novel function of KIF2C in the nervous system and provides an important regulatory mechanism on how activity-dependent microtubule dynamic regulates synaptic plasticity and cognition behaviors.
Collapse
Affiliation(s)
- Rui Zheng
- Department of Neurobiology and Department of Rehabilitation of the Children’s Hospital, Zhejiang University School of MedicineHangzhouChina
- NHC and CAMS Key Laboratory of Medical Neurobiology, Ministry of Education Frontier Science Center for Brain Research and Brain Machine Integration, School of Brain Science and Brain Medicine, Zhejiang UniversityHangzhouChina
| | - Yonglan Du
- Department of Neurobiology and Department of Rehabilitation of the Children’s Hospital, Zhejiang University School of MedicineHangzhouChina
- NHC and CAMS Key Laboratory of Medical Neurobiology, Ministry of Education Frontier Science Center for Brain Research and Brain Machine Integration, School of Brain Science and Brain Medicine, Zhejiang UniversityHangzhouChina
| | - Xintai Wang
- NHC and CAMS Key Laboratory of Medical Neurobiology, Ministry of Education Frontier Science Center for Brain Research and Brain Machine Integration, School of Brain Science and Brain Medicine, Zhejiang UniversityHangzhouChina
| | - Tailin Liao
- Department of Neurobiology and Department of Rehabilitation of the Children’s Hospital, Zhejiang University School of MedicineHangzhouChina
- NHC and CAMS Key Laboratory of Medical Neurobiology, Ministry of Education Frontier Science Center for Brain Research and Brain Machine Integration, School of Brain Science and Brain Medicine, Zhejiang UniversityHangzhouChina
| | - Zhe Zhang
- Department of Neurobiology and Department of Rehabilitation of the Children’s Hospital, Zhejiang University School of MedicineHangzhouChina
- NHC and CAMS Key Laboratory of Medical Neurobiology, Ministry of Education Frontier Science Center for Brain Research and Brain Machine Integration, School of Brain Science and Brain Medicine, Zhejiang UniversityHangzhouChina
| | - Na Wang
- Department of Neurobiology and Department of Rehabilitation of the Children’s Hospital, Zhejiang University School of MedicineHangzhouChina
- NHC and CAMS Key Laboratory of Medical Neurobiology, Ministry of Education Frontier Science Center for Brain Research and Brain Machine Integration, School of Brain Science and Brain Medicine, Zhejiang UniversityHangzhouChina
| | - Xiumao Li
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
| | - Ying Shen
- Department of Physiology and Department of Neurology of First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
| | - Lei Shi
- JNU-HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, Jinan UniversityGuanzhouChina
| | - Jianhong Luo
- Department of Neurobiology and Department of Rehabilitation of the Children’s Hospital, Zhejiang University School of MedicineHangzhouChina
- NHC and CAMS Key Laboratory of Medical Neurobiology, Ministry of Education Frontier Science Center for Brain Research and Brain Machine Integration, School of Brain Science and Brain Medicine, Zhejiang UniversityHangzhouChina
| | - Jun Xia
- Division of Life Science and The Brain and Intelligence Research Institute, The Hong Kong University of Science and TechnologyHong KongChina
| | - Ziyi Wang
- Innovative Institute of Basic Medical Sciences of Zhejiang University (Yuhang)HangzhouChina
| | - Junyu Xu
- Department of Neurobiology and Department of Rehabilitation of the Children’s Hospital, Zhejiang University School of MedicineHangzhouChina
- NHC and CAMS Key Laboratory of Medical Neurobiology, Ministry of Education Frontier Science Center for Brain Research and Brain Machine Integration, School of Brain Science and Brain Medicine, Zhejiang UniversityHangzhouChina
| |
Collapse
|
26
|
López-Hernández T, Takenaka KI, Mori Y, Kongpracha P, Nagamori S, Haucke V, Takamori S. Clathrin-independent endocytic retrieval of SV proteins mediated by the clathrin adaptor AP-2 at mammalian central synapses. eLife 2022; 11:e71198. [PMID: 35014951 PMCID: PMC8752090 DOI: 10.7554/elife.71198] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 12/28/2021] [Indexed: 12/25/2022] Open
Abstract
Neurotransmission is based on the exocytic fusion of synaptic vesicles (SVs) followed by endocytic membrane retrieval and the reformation of SVs. Conflicting models have been proposed regarding the mechanisms of SV endocytosis, most notably clathrin/adaptor protein complex 2 (AP-2)-mediated endocytosis and clathrin-independent ultrafast endocytosis. Partitioning between these pathways has been suggested to be controlled by temperature and stimulus paradigm. We report on the comprehensive survey of six major SV proteins to show that SV endocytosis in mouse hippocampal neurons at physiological temperature occurs independent of clathrin while the endocytic retrieval of a subset of SV proteins including the vesicular transporters for glutamate and GABA depend on sorting by the clathrin adaptor AP-2. Our findings highlight a clathrin-independent role of the clathrin adaptor AP-2 in the endocytic retrieval of select SV cargos from the presynaptic cell surface and suggest a revised model for the endocytosis of SV membranes at mammalian central synapses.
Collapse
Affiliation(s)
| | - Koh-ichiro Takenaka
- Laboratory of Neural Membrane Biology, Graduate School of Brain Science, Doshisha UniversityKyotoJapan
| | - Yasunori Mori
- Laboratory of Neural Membrane Biology, Graduate School of Brain Science, Doshisha UniversityKyotoJapan
| | - Pornparn Kongpracha
- Department of Laboratory Medicine, The Jikei University School of MedicineTokyoJapan
| | - Shushi Nagamori
- Department of Laboratory Medicine, The Jikei University School of MedicineTokyoJapan
| | - Volker Haucke
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP)BerlinGermany
| | - Shigeo Takamori
- Laboratory of Neural Membrane Biology, Graduate School of Brain Science, Doshisha UniversityKyotoJapan
| |
Collapse
|
27
|
Fenno LE, Levy R, Yizhar O. Molecular Optimization of Rhodopsin-Based Tools for Neuroscience Applications. Methods Mol Biol 2022; 2501:289-310. [PMID: 35857234 DOI: 10.1007/978-1-0716-2329-9_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
There is no question that genetically encoded tools have revolutionized neuroscience. These include optically modulated tools for writing-in (optogenetics) and reading-out (calcium, voltage, and neurotransmitter indicators) neural activity as well as precision expression of these reagents using virally mediated delivery. With few exceptions, these powerful approaches are derived from naturally occurring molecules that are sourced from diverse organisms that span all kingdoms of life. Successful expression of genetic tools in standard neuroscience model organisms requires optimizing gene structure, taking into account differences in both protein translation and trafficking. Myriad approaches have resolved these two challenges, resulting in order-of-magnitude increases in functional expression. In this chapter, we focus on synthesizing prior experience in successfully enabling the transition of genes across kingdoms with a goal of facilitating the production of the next generation of molecular tools for neuroscience. We then provide a detailed protocol that allows expression and testing of novel genetically encoded tools in mammalian cell lines and primary cultured neurons.
Collapse
Affiliation(s)
- Lief E Fenno
- Departments of Psychiatry and Neuroscience, University of Texas Austin Dell Medical School, Austin, TX, USA
| | - Rivka Levy
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Ofer Yizhar
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
28
|
Riera-Tur I, Schäfer T, Hornburg D, Mishra A, da Silva Padilha M, Fernández-Mosquera L, Feigenbutz D, Auer P, Mann M, Baumeister W, Klein R, Meissner F, Raimundo N, Fernández-Busnadiego R, Dudanova I. Amyloid-like aggregating proteins cause lysosomal defects in neurons via gain-of-function toxicity. Life Sci Alliance 2021; 5:5/3/e202101185. [PMID: 34933920 PMCID: PMC8711852 DOI: 10.26508/lsa.202101185] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 12/02/2021] [Accepted: 12/03/2021] [Indexed: 01/02/2023] Open
Abstract
Using cryo-ET, cell biology, and proteomics, this study shows that aggregating proteins impair the autophagy-lysosomal pathway in neurons by sequestering a subunit of the AP-3 adaptor complex. The autophagy-lysosomal pathway is impaired in many neurodegenerative diseases characterized by protein aggregation, but the link between aggregation and lysosomal dysfunction remains poorly understood. Here, we combine cryo-electron tomography, proteomics, and cell biology studies to investigate the effects of protein aggregates in primary neurons. We use artificial amyloid-like β-sheet proteins (β proteins) to focus on the gain-of-function aspect of aggregation. These proteins form fibrillar aggregates and cause neurotoxicity. We show that late stages of autophagy are impaired by the aggregates, resulting in lysosomal alterations reminiscent of lysosomal storage disorders. Mechanistically, β proteins interact with and sequester AP-3 μ1, a subunit of the AP-3 adaptor complex involved in protein trafficking to lysosomal organelles. This leads to destabilization of the AP-3 complex, missorting of AP-3 cargo, and lysosomal defects. Restoring AP-3μ1 expression ameliorates neurotoxicity caused by β proteins. Altogether, our results highlight the link between protein aggregation, lysosomal impairments, and neurotoxicity.
Collapse
Affiliation(s)
- Irene Riera-Tur
- Department of Molecules-Signaling-Development, Max Planck Institute of Neurobiology, Martinsried, Germany.,Molecular Neurodegeneration Group, Max Planck Institute of Neurobiology, Martinsried, Germany
| | - Tillman Schäfer
- Department of Molecular Structural Biology, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Daniel Hornburg
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany.,Experimental Systems Immunology Group, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Archana Mishra
- Department of Molecules-Signaling-Development, Max Planck Institute of Neurobiology, Martinsried, Germany
| | - Miguel da Silva Padilha
- Department of Molecules-Signaling-Development, Max Planck Institute of Neurobiology, Martinsried, Germany.,Molecular Neurodegeneration Group, Max Planck Institute of Neurobiology, Martinsried, Germany
| | - Lorena Fernández-Mosquera
- The William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Dennis Feigenbutz
- Department of Molecules-Signaling-Development, Max Planck Institute of Neurobiology, Martinsried, Germany.,Molecular Neurodegeneration Group, Max Planck Institute of Neurobiology, Martinsried, Germany
| | - Patrick Auer
- Department of Molecules-Signaling-Development, Max Planck Institute of Neurobiology, Martinsried, Germany.,Molecular Neurodegeneration Group, Max Planck Institute of Neurobiology, Martinsried, Germany
| | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Wolfgang Baumeister
- Department of Molecular Structural Biology, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Rüdiger Klein
- Department of Molecules-Signaling-Development, Max Planck Institute of Neurobiology, Martinsried, Germany
| | - Felix Meissner
- Experimental Systems Immunology Group, Max Planck Institute of Biochemistry, Martinsried, Germany.,Department of Systems Immunology and Proteomics, Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Nuno Raimundo
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA, USA
| | - Rubén Fernández-Busnadiego
- Department of Molecular Structural Biology, Max Planck Institute of Biochemistry, Martinsried, Germany .,Institute of Neuropathology, University Medical Center Goettingen, Goettingen, Germany.,Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Goettingen, Goettingen, Germany
| | - Irina Dudanova
- Department of Molecules-Signaling-Development, Max Planck Institute of Neurobiology, Martinsried, Germany .,Molecular Neurodegeneration Group, Max Planck Institute of Neurobiology, Martinsried, Germany
| |
Collapse
|
29
|
Bloxham W, Brinks D, Kheifets S, Cohen AE. Linearly polarized excitation enhances signals from fluorescent voltage indicators. Biophys J 2021; 120:5333-5342. [PMID: 34710379 DOI: 10.1016/j.bpj.2021.10.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 10/10/2021] [Accepted: 10/20/2021] [Indexed: 10/20/2022] Open
Abstract
Voltage imaging in cells requires high-speed recording of small fluorescent signals, often leading to low signal/noise ratios. Because voltage indicators are membrane bound, their orientations are partially constrained by the plane of the membrane. We explored whether tuning the linear polarization of excitation light could enhance voltage indicator fluorescence. We tested a panel of dye- and protein-based voltage indicators in mammalian cells. The dye BeRST1 showed a 73% increase in brightness between the least and most favorable polarizations. The protein-based reporter ASAP1 showed a 22% increase in brightness, and QuasAr3 showed a 14% increase in brightness. In very thin neurites expressing QuasAr3, improvements were anomalously large, with a 170% increase in brightness between polarization parallel versus perpendicular to the dendrite. Signal/noise ratios of optically recorded action potentials were increased by up to 50% in neurites expressing QuasAr3. These results demonstrate that polarization control can be a facile means to enhance signals from fluorescent voltage indicators, particularly in thin neurites or in high-background environments.
Collapse
Affiliation(s)
- William Bloxham
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts; Department of Physics, Harvard University, Cambridge, Massachusetts; Physics of Living Systems, Department of Physics, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Daan Brinks
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts; Department of Imaging Physics, Delft University of Technology, Delft, the Netherlands; Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Simon Kheifets
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts
| | - Adam E Cohen
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts; Department of Physics, Harvard University, Cambridge, Massachusetts.
| |
Collapse
|
30
|
Gray ME, Johnson ZR, Modak D, Tamilselvan E, Tyska MJ, Sotomayor M. Heterophilic and homophilic cadherin interactions in intestinal intermicrovillar links are species dependent. PLoS Biol 2021; 19:e3001463. [PMID: 34871294 PMCID: PMC8691648 DOI: 10.1371/journal.pbio.3001463] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 12/21/2021] [Accepted: 10/30/2021] [Indexed: 11/19/2022] Open
Abstract
Enterocytes are specialized epithelial cells lining the luminal surface of the small intestine that build densely packed arrays of microvilli known as brush borders. These microvilli drive nutrient absorption and are arranged in a hexagonal pattern maintained by intermicrovillar links formed by 2 nonclassical members of the cadherin superfamily of calcium-dependent cell adhesion proteins: protocadherin-24 (PCDH24, also known as CDHR2) and the mucin-like protocadherin (CDHR5). The extracellular domains of these proteins are involved in heterophilic and homophilic interactions important for intermicrovillar function, yet the structural determinants of these interactions remain unresolved. Here, we present X-ray crystal structures of the PCDH24 and CDHR5 extracellular tips and analyze their species-specific features relevant for adhesive interactions. In parallel, we use binding assays to identify the PCDH24 and CDHR5 domains involved in both heterophilic and homophilic adhesion for human and mouse proteins. Our results suggest that homophilic and heterophilic interactions involving PCDH24 and CDHR5 are species dependent with unique and distinct minimal adhesive units.
Collapse
Affiliation(s)
- Michelle E. Gray
- Ohio State Biochemistry Program, The Ohio State University, Columbus, Ohio, United States of America
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio, United States of America
| | - Zachary R. Johnson
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio, United States of America
| | - Debadrita Modak
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio, United States of America
| | - Elakkiya Tamilselvan
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio, United States of America
- Biophysics Program, The Ohio State University, Columbus, Ohio, United States of America
| | - Matthew J. Tyska
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Marcos Sotomayor
- Ohio State Biochemistry Program, The Ohio State University, Columbus, Ohio, United States of America
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio, United States of America
- Biophysics Program, The Ohio State University, Columbus, Ohio, United States of America
- * E-mail:
| |
Collapse
|
31
|
Primary cilia and the reciprocal activation of AKT and SMAD2/3 regulate stretch-induced autophagy in trabecular meshwork cells. Proc Natl Acad Sci U S A 2021; 118:2021942118. [PMID: 33753495 DOI: 10.1073/pnas.2021942118] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Activation of autophagy is one of the responses elicited by high intraocular pressure (IOP) and mechanical stretch in trabecular meshwork (TM) cells. However, the mechanosensor and the molecular mechanisms by which autophagy is induced by mechanical stretch in these or other cell types is largely unknown. Here, we have investigated the mechanosensor and downstream signaling pathway that regulate cyclic mechanical stretch (CMS)-induced autophagy in TM cells. We report that primary cilia act as a mechanosensor for CMS-induced autophagy and identified a cross-regulatory talk between AKT1 and noncanonical SMAD2/3 signaling as critical components of primary cilia-mediated activation of autophagy by mechanical stretch. Furthermore, we demonstrated the physiological significance of our findings in ex vivo perfused eyes. Removal of primary cilia disrupted the homeostatic IOP compensatory response and prevented the increase in LC3-II protein levels in response to elevated pressure challenge, strongly supporting a role of primary cilia-mediated autophagy in regulating IOP homeostasis.
Collapse
|
32
|
Liu T, Li Q, Yang S, Zhao T, Lin J, Ju T, Wen Z. CNTs-CaP/chitosan-coated AZ91D magnesium alloy extract promoted rat dorsal root ganglia neuron growth via activating ERK signalling pathway. Cell Biochem Funct 2021; 39:908-920. [PMID: 34296452 DOI: 10.1002/cbf.3662] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 06/24/2021] [Accepted: 06/29/2021] [Indexed: 01/19/2023]
Abstract
Increasing attention has been paid on the application of biodegradable materials such as magnesium and its alloys in neuron repair. AZ91D magnesium alloy coated with carbon nanotubes (CNTs) and/or calcium phosphate (CaP)/chitosan (CS) was fabricated in this study. To evaluate the bioactivity of these AZ91D-based composites, the extracts were prepared by immersing samples in modified simulated body fluid (m-SBF) for 0, 2, 8, 16, 24, 34, 44, 60, or 90 days. Immunofluorescence staining for neuronal class III β-tubulin (TUJ1) revealed that both CNTs-CaP/CS-AZ91D and CaP/CS-AZ91D extracts promoted axon outgrowth of dorsal root ganglia (DRG) neurons, accompanied with increased expression of phosphorylated focal adhesion kinase (p-FAK) and growth associated protein-43 (GAP-43). Besides, the extracts increased the expression and the release of neurotrophic factors including nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF). ERK signalling was activated in DRG neurons after treating with either CNTs-CaP/CS-AZ91D or CaP/CS-AZ91D extracts, and its inhibition with U0126 counteracted the beneficial effects of these extracts on DRG neuron. Overall, the extracts from these AZ91D-based composites might promote DRG neuron growth via activating ERK signalling pathway. Notably, CNTs-CaP/CS-AZ91D extracts showed a better promoting effect on neuron growth than CaP/CS-AZ91D. Assessment of ion elements showed that the addition of CNTs coating enhanced magnesium corrosion resistance and reduced the deposition of calcium and phosphorus on the surface of CaP/CS-AZ91D alloy. These findings demonstrate that CNTs-CaP/CS-AZ91D likely provide a more suitable environment for neuron growth, which suggests a potential implantable biomaterial for the treatment of nerve injury. SIGNIFICANCE: AZ91D magnesium alloy coated with carbon nanotubes (CNTs) and/or calcium phosphate (CaP)/chitosan (CS) was fabricated and their immersion extracts were prepared using modified simulated body fluid in this study. Both extracts from CNTs-CaP/CS and CaP/CS-coated AZ91D magnesium alloy promotes rat dorsal root ganglia (DRG) neuron growth via activating ERK signalling pathway. Notably, the addition of CNTs improves the performance of CaP/CS-AZ91D. For the first time, our research demonstrates that CNTs-CaP/CS-AZ91D likely provide a suitable environment for neuron growth, suggesting these AZ91D-based composites as potential implantable biomaterials for the treatment of nerve injury.
Collapse
Affiliation(s)
- Tingjiao Liu
- Department of Neurology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qianqian Li
- Department of Neurology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shanshan Yang
- Department of Neurology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Tingting Zhao
- Department of Neurology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jinghan Lin
- Department of Neurology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Ting Ju
- Department of Neurology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhaohui Wen
- Department of Neurology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
33
|
Mendoza MB, Gutierrez S, Ortiz R, Moreno DF, Dermit M, Dodel M, Rebollo E, Bosch M, Mardakheh FK, Gallego C. The elongation factor eEF1A2 controls translation and actin dynamics in dendritic spines. Sci Signal 2021; 14:14/691/eabf5594. [PMID: 34257105 DOI: 10.1126/scisignal.abf5594] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Synaptic plasticity involves structural modifications in dendritic spines that are modulated by local protein synthesis and actin remodeling. Here, we investigated the molecular mechanisms that connect synaptic stimulation to these processes. We found that the phosphorylation of isoform-specific sites in eEF1A2-an essential translation elongation factor in neurons-is a key modulator of structural plasticity in dendritic spines. Expression of a nonphosphorylatable eEF1A2 mutant stimulated mRNA translation but reduced actin dynamics and spine density. By contrast, a phosphomimetic eEF1A2 mutant exhibited decreased association with F-actin and was inactive as a translation elongation factor. Activation of metabotropic glutamate receptor signaling triggered transient dissociation of eEF1A2 from its regulatory guanine exchange factor (GEF) protein in dendritic spines in a phosphorylation-dependent manner. We propose that eEF1A2 establishes a cross-talk mechanism that coordinates translation and actin dynamics during spine remodeling.
Collapse
Affiliation(s)
- Mònica B Mendoza
- Molecular Biology Institute of Barcelona (IBMB), CSIC, Catalonia 08028, Spain
| | - Sara Gutierrez
- Molecular Biology Institute of Barcelona (IBMB), CSIC, Catalonia 08028, Spain
| | - Raúl Ortiz
- Molecular Biology Institute of Barcelona (IBMB), CSIC, Catalonia 08028, Spain
| | - David F Moreno
- Molecular Biology Institute of Barcelona (IBMB), CSIC, Catalonia 08028, Spain
| | - Maria Dermit
- Centre for Cancer Cell and Molecular Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse square, London EC1M 6BQ, UK
| | - Martin Dodel
- Centre for Cancer Cell and Molecular Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse square, London EC1M 6BQ, UK
| | - Elena Rebollo
- Molecular Biology Institute of Barcelona (IBMB), CSIC, Catalonia 08028, Spain
| | - Miquel Bosch
- Department of Basic Sciences, Universitat Internacional de Catalunya (UIC-Barcelona), Sant Cugat del Vallès 08195, Spain.,Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona 08028, Spain
| | - Faraz K Mardakheh
- Centre for Cancer Cell and Molecular Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse square, London EC1M 6BQ, UK
| | - Carme Gallego
- Molecular Biology Institute of Barcelona (IBMB), CSIC, Catalonia 08028, Spain.
| |
Collapse
|
34
|
Gonzalez Sabater V, Rigby M, Burrone J. Voltage-Gated Potassium Channels Ensure Action Potential Shape Fidelity in Distal Axons. J Neurosci 2021; 41:5372-5385. [PMID: 34001627 PMCID: PMC8221596 DOI: 10.1523/jneurosci.2765-20.2021] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 03/29/2021] [Accepted: 04/01/2021] [Indexed: 11/21/2022] Open
Abstract
The initiation and propagation of the action potential (AP) along an axon allows neurons to convey information rapidly and across distant sites. Although AP properties have typically been characterized at the soma and proximal axon, knowledge of the propagation of APs toward distal axonal domains of mammalian CNS neurons remains limited. We used genetically encoded voltage indicators (GEVIs) to image APs with submillisecond temporal resolution simultaneously at different locations along the long axons of dissociated hippocampal neurons from rat embryos of either sex. We found that APs became sharper and showed remarkable fidelity as they traveled toward distal axons, even during a high-frequency train. Blocking voltage-gated potassium channels (Kv) with 4-AP resulted in an increase in AP width in all compartments, which was stronger at distal locations and exacerbated during AP trains. We conclude that the higher levels of Kv channel activity in distal axons serve to sustain AP fidelity, conveying a reliable digital signal to presynaptic boutons.SIGNIFICANCE STATEMENT The AP represents the electrical signal carried along axons toward distant presynaptic boutons where it culminates in the release of neurotransmitters. The nonlinearities involved in this process are such that small changes in AP shape can result in large changes in neurotransmitter release. Since axons are remarkably long structures, any distortions that APs suffer along the way have the potential to translate into a significant modulation of synaptic transmission, particularly in distal domains. To avoid these issues, distal axons have ensured that signals are kept remarkably constant and insensitive to modulation during a train, despite the long distances traveled. Here, we uncover the mechanisms that allow distal axonal domains to provide a reliable and faithful digital signal to presynaptic terminals.
Collapse
Affiliation(s)
- Victoria Gonzalez Sabater
- MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, United Kingdom
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, United Kingdom
| | - Mark Rigby
- MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, United Kingdom
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, United Kingdom
| | - Juan Burrone
- MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, United Kingdom
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, United Kingdom
| |
Collapse
|
35
|
Beilina A, Bonet-Ponce L, Kumaran R, Kordich JJ, Ishida M, Mamais A, Kaganovich A, Saez-Atienzar S, Gershlick DC, Roosen DA, Pellegrini L, Malkov V, Fell MJ, Harvey K, Bonifacino JS, Moore DJ, Cookson MR. The Parkinson's Disease Protein LRRK2 Interacts with the GARP Complex to Promote Retrograde Transport to the trans-Golgi Network. Cell Rep 2021; 31:107614. [PMID: 32375042 PMCID: PMC7315779 DOI: 10.1016/j.celrep.2020.107614] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 01/17/2020] [Accepted: 04/13/2020] [Indexed: 12/11/2022] Open
Abstract
Mutations in Leucine-rich repeat kinase 2 (LRRK2) cause Parkinson's disease (PD). However, the precise function of LRRK2 remains unclear. We report an interaction between LRRK2 and VPS52, a subunit of the Golgi-associated retrograde protein (GARP) complex that identifies a function of LRRK2 in regulating membrane fusion at the trans-Golgi network (TGN). At the TGN, LRRK2 further interacts with the Golgi SNAREs VAMP4 and Syntaxin-6 and acts as a scaffolding platform that stabilizes the GARP-SNAREs complex formation. Therefore, LRRK2 influences both retrograde and post-Golgi trafficking pathways in a manner dependent on its GTP binding and kinase activity. This action is exaggerated by mutations associated with Parkinson's disease and can be blocked by kinase inhibitors. Disruption of GARP sensitizes dopamine neurons to mutant LRRK2 toxicity in C. elegans, showing that these pathways are interlinked in vivo and suggesting a link in PD.
Collapse
Affiliation(s)
- Alexandra Beilina
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20814, USA
| | - Luis Bonet-Ponce
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20814, USA
| | - Ravindran Kumaran
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20814, USA
| | - Jennifer J Kordich
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - Morié Ishida
- Cell Biology and Neurobiology Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20814, USA
| | - Adamantios Mamais
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20814, USA
| | - Alice Kaganovich
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20814, USA
| | - Sara Saez-Atienzar
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20814, USA
| | - David C Gershlick
- Cell Biology and Neurobiology Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20814, USA
| | - Dorien A Roosen
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20814, USA; School of Pharmacy, University of Reading, Whiteknights, Reading, RG6 6AP, UK
| | - Laura Pellegrini
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20814, USA; Department of Pharmacology, UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX, UK
| | - Vlad Malkov
- Merck & Co., Inc., 33 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Matthew J Fell
- Merck & Co., Inc., 33 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Kirsten Harvey
- Department of Pharmacology, UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX, UK
| | - Juan S Bonifacino
- Cell Biology and Neurobiology Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20814, USA
| | - Darren J Moore
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - Mark R Cookson
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20814, USA.
| |
Collapse
|
36
|
Behrendt L, Hoischen C, Kaether C. Disease-causing mutated ATLASTIN 3 is excluded from distal axons and reduces axonal autophagy. Neurobiol Dis 2021; 155:105400. [PMID: 34019998 DOI: 10.1016/j.nbd.2021.105400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 04/27/2021] [Accepted: 05/16/2021] [Indexed: 10/21/2022] Open
Abstract
Mutations in the ER-network forming GTPase atlastin3 (ATL3) can cause axon degeneration of sensory neurons by not fully understood mechanisms. We here show that the hereditary sensory and autonomous neuropathy (HSAN)-causing ATL3 Y192C or P338R are excluded from distal axons by a barrier at the axon initial segment (AIS). This barrier is selective for mutated ATL3, but not wildtype ATL3 or unrelated ER-membrane proteins. Actin-depolymerization partially restores the transport of ATL3 Y192C into distal axons. The results point to the existence of a selective diffusion barrier in the ER membrane at the AIS, analogous to the AIS-based barriers for plasma membrane and cytosolic proteins. Functionally, the absence of ATL3 at the distal axon reduces axonal autophagy and the ER network deformation in the soma causes a reduction in axonal lysosomes. Both could contribute to axonal degeneration and eventually to HSAN.
Collapse
Affiliation(s)
- Laura Behrendt
- Leibniz-Institut für Alternsforschung-Fritz-Lipmann-Institut, 07745 Jena, Germany
| | - Christian Hoischen
- Leibniz-Institut für Alternsforschung-Fritz-Lipmann-Institut, 07745 Jena, Germany
| | - Christoph Kaether
- Leibniz-Institut für Alternsforschung-Fritz-Lipmann-Institut, 07745 Jena, Germany.
| |
Collapse
|
37
|
Ryanodine receptor-mediated Ca 2+ release and atlastin-2 GTPase activity contribute to IP 3-induced dendritic Ca 2+ signals in primary hippocampal neurons. Cell Calcium 2021; 96:102399. [PMID: 33812310 DOI: 10.1016/j.ceca.2021.102399] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 03/17/2021] [Accepted: 03/18/2021] [Indexed: 12/19/2022]
Abstract
Neuronal Ca2+ signals are fundamental for synaptic transmission and activity-dependent changes in gene expression. Voltage-gated Ca2+ channels and N-methyl-d-aspartate receptors play major roles in mediating external Ca2+ entry during action potential firing and glutamatergic activity. Additionally, the inositol-1,4,5-trisphosphate receptor (IP3R) and the ryanodine receptor (RyR) channels expressed in the endoplasmic reticulum (ER) also contribute to the generation of Ca2+ signals in response to neuronal activity. The ER forms a network that pervades the entire neuronal volume, allowing intracellular Ca2+ release in dendrites, soma and presynaptic boutons. Despite its unique morphological features, the contributions of ER structure and of ER-shaping proteins such as atlastin - an ER enriched GTPase that mediates homotypic ER tubule fusion - to the generation of Ca2+ signals in dendrites remains unreported. Here, we investigated the contribution of RyR-mediated Ca2+ release to IP3-generated Ca2+ signals in dendrites of cultured hippocampal neurons. We also employed GTPase activity-deficient atlastin-2 (ATL2) mutants to evaluate the potential role of atlastin on Ca2+ signaling and ER-resident Ca2+ channel distribution. We found that pharmacological suppression of RyR channel activity increased the rising time and reduced the magnitude and propagation of IP3-induced Ca2+ signals. Additionally, ATL2 mutants induced specific ER morphological alterations, delayed the onset and increased the rising time of IP3-evoked Ca2+ signals, and caused RyR2 and IP3R1 aggregation and RyR2 redistribution. These results indicate that both RyR and ATL2 activity regulate IP3-induced Ca2+ signal dynamics through RyR-mediated Ca2+-induced Ca2+ release, ER shaping and RyR2 distribution.
Collapse
|
38
|
Dong F, Mao J, Chen M, Yoon J, Mao Y. Schizophrenia risk ZNF804A interacts with its associated proteins to modulate dendritic morphology and synaptic development. Mol Brain 2021; 14:12. [PMID: 33446247 PMCID: PMC7809827 DOI: 10.1186/s13041-021-00729-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 01/01/2021] [Indexed: 11/30/2022] Open
Abstract
Schizophrenia (SZ) is a devastating brain disease that affects about 1% of world population. Among the top genetic associations, zinc finger protein 804A (ZNF804A) gene encodes a zinc finger protein, associated with SZ and biolar disorder (BD). Copy number variants (CNVs) of ZNF804A have been observed in patients with autism spectrum disorders (ASDs), anxiety disorder, and BD, suggesting that ZNF804A is a dosage sensitive gene for brain development. However, its molecular functions have not been fully determined. Our previous interactomic study revealed that ZNF804A interacts with multiple proteins to control protein translation and neural development. ZNF804A is localized in the cytoplasm and neurites in the human cortex and is expressed in various types of neurons, including pyramidal, dopaminergic, GABAergic, and Purkinje neurons in mouse brain. To further examine the effect of gene dosage of ZNF804A on neurite morphology, both knockdown and overexpression of ZNF804A in primary neuronal cells significantly attenuate dendritic complex and spine formation. To determine the factors mediating these phenotypes, interestingly, three binding proteins of ZNF804A, galectin 1 (LGALS1), fasciculation and elongation protein zeta 1 (FEZ1) and ribosomal protein SA (RPSA), show different effects on reversing the deficits. LGALS1 and FEZ1 stimulate neurite outgrowth at basal level but RPSA shows no effect. Intriguingly, LGALS1 but not FEZ1, reverses the neurite outgrowth deficits induced by ZNF804A knockdown. However, FEZ1 and RPSA but not LGALS1, can ameliorate ZNF804A overexpression-mediated dendritic abnormalities. Thus, our results uncover a critical post-mitotic role of ZNF804A in neurite and synaptic development relevant to neurodevelopmental pathologies.
Collapse
Affiliation(s)
- Fengping Dong
- Department of Biology, Pennsylvania State University, 214 Life Sciences Building, University Park, PA, 16802, USA
| | - Joseph Mao
- Department of Biology, Pennsylvania State University, 214 Life Sciences Building, University Park, PA, 16802, USA
| | - Miranda Chen
- Department of Biology, Pennsylvania State University, 214 Life Sciences Building, University Park, PA, 16802, USA
| | - Joy Yoon
- Department of Biology, Pennsylvania State University, 214 Life Sciences Building, University Park, PA, 16802, USA
| | - Yingwei Mao
- Department of Biology, Pennsylvania State University, 214 Life Sciences Building, University Park, PA, 16802, USA.
| |
Collapse
|
39
|
Qian Y, Cosio DMO, Piatkevich KD, Aufmkolk S, Su WC, Celiker OT, Schohl A, Murdock MH, Aggarwal A, Chang YF, Wiseman PW, Ruthazer ES, Boyden ES, Campbell RE. Improved genetically encoded near-infrared fluorescent calcium ion indicators for in vivo imaging. PLoS Biol 2020; 18:e3000965. [PMID: 33232322 PMCID: PMC7723245 DOI: 10.1371/journal.pbio.3000965] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 12/08/2020] [Accepted: 10/29/2020] [Indexed: 12/15/2022] Open
Abstract
Near-infrared (NIR) genetically encoded calcium ion (Ca2+) indicators (GECIs) can provide advantages over visible wavelength fluorescent GECIs in terms of reduced phototoxicity, minimal spectral cross talk with visible light excitable optogenetic tools and fluorescent probes, and decreased scattering and absorption in mammalian tissues. Our previously reported NIR GECI, NIR-GECO1, has these advantages but also has several disadvantages including lower brightness and limited fluorescence response compared to state-of-the-art visible wavelength GECIs, when used for imaging of neuronal activity. Here, we report 2 improved NIR GECI variants, designated NIR-GECO2 and NIR-GECO2G, derived from NIR-GECO1. We characterized the performance of the new NIR GECIs in cultured cells, acute mouse brain slices, and Caenorhabditis elegans and Xenopus laevis in vivo. Our results demonstrate that NIR-GECO2 and NIR-GECO2G provide substantial improvements over NIR-GECO1 for imaging of neuronal Ca2+ dynamics. This study describes improved genetically encoded near-infrared fluorescent calcium ion indicators, demonstrating that they enable robust detection of neuronal activity in cultured cells, rodent brain slices, Caenorhabditis elegans, and Xenopus laevis.
Collapse
Affiliation(s)
- Yong Qian
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada
- Departments of Biological Engineering, Media Arts and Sciences, Brain and Cognitive Sciences, McGovern Institute, Koch Institute, Center for Neurobiological Engineering, MIT, and Howard Hughes Medical Institute, Cambridge, Massachusetts, United States of America
| | - Danielle M. Orozco Cosio
- Departments of Biological Engineering, Media Arts and Sciences, Brain and Cognitive Sciences, McGovern Institute, Koch Institute, Center for Neurobiological Engineering, MIT, and Howard Hughes Medical Institute, Cambridge, Massachusetts, United States of America
| | - Kiryl D. Piatkevich
- Departments of Biological Engineering, Media Arts and Sciences, Brain and Cognitive Sciences, McGovern Institute, Koch Institute, Center for Neurobiological Engineering, MIT, and Howard Hughes Medical Institute, Cambridge, Massachusetts, United States of America
- School of Life Sciences, Westlake University, Hangzhou, China
| | - Sarah Aufmkolk
- Montreal Neurological Institute-Hospital, Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
- Department of Chemistry, McGill University, Montreal, Quebec, Canada
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Wan-Chi Su
- LumiSTAR Biotechnology, Nangang District, Taipei City, Taiwan
| | - Orhan T. Celiker
- Departments of Biological Engineering, Media Arts and Sciences, Brain and Cognitive Sciences, McGovern Institute, Koch Institute, Center for Neurobiological Engineering, MIT, and Howard Hughes Medical Institute, Cambridge, Massachusetts, United States of America
| | - Anne Schohl
- Montreal Neurological Institute-Hospital, Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Mitchell H. Murdock
- Departments of Biological Engineering, Media Arts and Sciences, Brain and Cognitive Sciences, McGovern Institute, Koch Institute, Center for Neurobiological Engineering, MIT, and Howard Hughes Medical Institute, Cambridge, Massachusetts, United States of America
| | - Abhi Aggarwal
- Howard Hughes Medical Institute, Janelia Research Campus, Ashburn, Virginia, United States of America
| | - Yu-Fen Chang
- LumiSTAR Biotechnology, Nangang District, Taipei City, Taiwan
| | - Paul W. Wiseman
- Department of Chemistry, McGill University, Montreal, Quebec, Canada
- Department of Physics, McGill University, Montreal, Quebec, Canada
| | - Edward S. Ruthazer
- Montreal Neurological Institute-Hospital, Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Edward S. Boyden
- Departments of Biological Engineering, Media Arts and Sciences, Brain and Cognitive Sciences, McGovern Institute, Koch Institute, Center for Neurobiological Engineering, MIT, and Howard Hughes Medical Institute, Cambridge, Massachusetts, United States of America
| | - Robert E. Campbell
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada
- Department of Chemistry, The University of Tokyo, Tokyo, Japan
- * E-mail:
| |
Collapse
|
40
|
Komuro H, Yamazoe M, Nozaki K, Nagai A, Sasano T. Cardiomyocyte uptake mechanism of a hydroxyapatite nanoparticle mediated gene delivery system. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2020; 11:1685-1692. [PMID: 33194517 PMCID: PMC7653333 DOI: 10.3762/bjnano.11.150] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 10/06/2020] [Indexed: 06/11/2023]
Abstract
Gene therapy has been explored as a future alternative for treating heart disease. Among several gene delivery systems aimed at penetrating specific target cells, we focused on safe and non-viral gene delivery materials with a high transfection efficiency. Although various techniques have been developed, the mechanisms underlying the cellular uptake of gene delivery materials have not yet been sufficiently studied in cardiomyocytes. The aim of this study was to determine how hydroxyapatite (HAp) nanoparticles contribute to the delivery of plasmid DNA (pDNA) into cardiomyocytes. We fabricated HAp nanoparticles using the water-in-oil (W/O) emulsion method and used these nanoparticles as the delivery vector for transfecting cardiomyocyte-derived HL-1 cells. HAp exhibited particles on the nanoscale and with a low cytotoxicity in HL-1 cells. The transfection assay performed with several endocytosis inhibitors suggested that the HAp/pDNA complexes were internalized by HL-1 cells through macropinocytosis. Furthermore, this HL-1 cell uptake was generated in response to HAp stimulation. Thus, HAp is a positive regulator of macropinocytosis in HL-1 cells and a good system for gene delivery in cardiomyocytes.
Collapse
Affiliation(s)
- Hiroaki Komuro
- Department of Cardiovascular Physiology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo, Tokyo 113-8510, Japan
- Department of Cardiovascular Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo, Tokyo 113-8510, Japan
| | - Masahiro Yamazoe
- Department of Bio-informational Pharmacology, Medical Research Institute, Tokyo Medical and Dental University, Bunkyo, Tokyo 113-8510, Japan
| | - Kosuke Nozaki
- Department of Restorative Sciences, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo, Tokyo 113-8510, Japan
| | - Akiko Nagai
- Department of Anatomy, School of Dentistry, Aichi Gakuin University, Chikusa, Nagoya 464-8650, Japan
| | - Tetsuo Sasano
- Department of Cardiovascular Physiology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo, Tokyo 113-8510, Japan
- Department of Cardiovascular Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo, Tokyo 113-8510, Japan
| |
Collapse
|
41
|
Monakhov MV, Matlashov ME, Colavita M, Song C, Shcherbakova DM, Antic SD, Verkhusha VV, Knöpfel T. Screening and Cellular Characterization of Genetically Encoded Voltage Indicators Based on Near-Infrared Fluorescent Proteins. ACS Chem Neurosci 2020; 11:3523-3531. [PMID: 33063984 DOI: 10.1021/acschemneuro.0c00046] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
We developed genetically encoded voltage indicators using a transmembrane voltage-sensing domain and bright near-infrared fluorescent proteins derived from bacterial phytochromes. These new voltage indicators are excited by 640 nm light and emission is measured at 670 nm, allowing imaging in the near-infrared tissue transparency window. The spectral properties of our new indicators permit seamless voltage imaging with simultaneous blue-green light optogenetic actuator activation as well as simultaneous voltage-calcium imaging when paired with green calcium indicators. Iterative optimizations led to a fluorescent probe, here termed nirButterfly, which reliably reports neuronal activities including subthreshold membrane potential depolarization and hyperpolarization as well as spontaneous spiking or electrically- and optogenetically evoked action potentials. This enables largely improved all-optical causal interrogations of physiology.
Collapse
Affiliation(s)
- Mikhail V Monakhov
- Institute for Systems Genomics, Stem Cell Institute, Department of Neuroscience, UConn Health, Farmington, Connecticut 06030, United States
- Department of Anatomy and Structural Biology and Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, New York 10461, United States
| | - Mikhail E Matlashov
- Department of Anatomy and Structural Biology and Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, New York 10461, United States
| | - Michelangelo Colavita
- Laboratory for Neuronal Circuit Dynamics, Imperial College London, London W12 0NN, U.K
| | - Chenchen Song
- Laboratory for Neuronal Circuit Dynamics, Imperial College London, London W12 0NN, U.K
| | - Daria M Shcherbakova
- Department of Anatomy and Structural Biology and Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, New York 10461, United States
| | - Srdjan D Antic
- Institute for Systems Genomics, Stem Cell Institute, Department of Neuroscience, UConn Health, Farmington, Connecticut 06030, United States
| | - Vladislav V Verkhusha
- Department of Anatomy and Structural Biology and Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, New York 10461, United States
| | - Thomas Knöpfel
- Laboratory for Neuronal Circuit Dynamics, Imperial College London, London W12 0NN, U.K
| |
Collapse
|
42
|
Aguilar R, Bustos FJ, Nardocci G, van Zundert B, Montecino M. Epigenetic silencing of the osteoblast-lineage gene program during hippocampal maturation. J Cell Biochem 2020; 122:367-384. [PMID: 33135214 DOI: 10.1002/jcb.29865] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 10/15/2020] [Accepted: 10/21/2020] [Indexed: 12/11/2022]
Abstract
Accumulating evidence indicates that epigenetic control of gene expression plays a significant role during cell lineage commitment and subsequent cell fate maintenance. Here, we assess epigenetic mechanisms operating in the rat brain that mediate silencing of genes that are expressed during early and late stages of osteogenesis. We report that repression of the osteoblast master regulator Sp7 in embryonic (E18) hippocampus is mainly mediated through the Polycomb complex PRC2 and its enzymatic product H3K27me3. During early postnatal (P10), juvenile (P30), and adult (P90) hippocampal stages, the repressive H3K27me3 mark is progressively replaced by nucleosome enrichment and increased CpG DNA methylation at the Sp7 gene promoter. In contrast, silencing of the late bone phenotypic Bglap gene in the hippocampus is PRC2-independent and accompanied by strong CpG methylation from E18 through postnatal and adult stages. Forced ectopic expression of the primary master regulator of osteogenesis Runx2 in embryonic hippocampal neurons activates the expression of its downstream target Sp7 gene. Moreover, transcriptomic analyses show that several genes associated with the mesenchymal-osteogenic lineages are transcriptionally activated in these hippocampal cells that express Runx2 and Sp7. This effect is accompanied by a loss in neuronal properties, including a significant reduction in secondary processes at the dendritic arbor and reduced expression of critical postsynaptic genes like PSD95. Together, our results reveal a developmental progression in epigenetic control mechanisms that repress the expression of the osteogenic program in hippocampal neurons at embryonic, postnatal, and adult stages.
Collapse
Affiliation(s)
- Rodrigo Aguilar
- Faculty of Medicine and Faculty of Life Sciences, Institute of Biomedical Sciences, Universidad Andres Bello, Santiago, Chile.,FONDAP Center for Genome Regulation, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Fernando J Bustos
- Faculty of Medicine and Faculty of Life Sciences, Institute of Biomedical Sciences, Universidad Andres Bello, Santiago, Chile
| | - Gino Nardocci
- Faculty of Medicine and Faculty of Life Sciences, Institute of Biomedical Sciences, Universidad Andres Bello, Santiago, Chile
| | - Brigitte van Zundert
- Faculty of Medicine and Faculty of Life Sciences, Institute of Biomedical Sciences, Universidad Andres Bello, Santiago, Chile.,CARE Biomedical Research Center, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Martin Montecino
- Faculty of Medicine and Faculty of Life Sciences, Institute of Biomedical Sciences, Universidad Andres Bello, Santiago, Chile.,FONDAP Center for Genome Regulation, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
43
|
Shemetov AA, Monakhov MV, Zhang Q, Canton-Josh JE, Kumar M, Chen M, Matlashov ME, Li X, Yang W, Nie L, Shcherbakova DM, Kozorovitskiy Y, Yao J, Ji N, Verkhusha VV. A near-infrared genetically encoded calcium indicator for in vivo imaging. Nat Biotechnol 2020; 39:368-377. [PMID: 33106681 PMCID: PMC7956128 DOI: 10.1038/s41587-020-0710-1] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 09/18/2020] [Indexed: 12/18/2022]
Abstract
While calcium imaging has become a mainstay of modern neuroscience, the spectral properties of current fluorescent calcium indicators limit deep tissue imaging as well as simultaneous use with other probes. Using two monomeric near-infrared fluorescent proteins, we engineered a near-infrared FRET-based genetically encoded calcium indicator (iGECI). iGECI exhibits high brightness, high photostability, and up to 600% increase in fluorescence response to calcium. In dissociated neurons, iGECI detects spontaneous neuronal activity, and electrically and optogenetically induced firing. We validated iGECI performance up to a depth of almost 400 μm in acute brain slices using one-photon light-sheet imaging. Applying hybrid photoacoustic and fluorescence microscopy, we simultaneously monitored neuronal and hemodynamic activities in the mouse brain through an intact skull, with ~3 μm lateral and ~25–50 μm axial resolution. Using two-photon imaging, we detected evoked and spontaneous neuronal activity in the mouse visual cortex, with fluorescence changes of up to 25%. iGECI allows biosensors and optogenetic actuators to be multiplexed without spectral crosstalk.
Collapse
Affiliation(s)
- Anton A Shemetov
- Department of Anatomy and Structural Biology and Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA.,Autonomous Therapeutics, Inc., New York, NY, USA
| | - Mikhail V Monakhov
- Department of Anatomy and Structural Biology and Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Qinrong Zhang
- Department of Physics, University of California, Berkeley, Berkeley, CA, USA
| | - Jose Ernesto Canton-Josh
- Department of Neurobiology, Weinberg School of Arts and Sciences, Northwestern University, Evanston, IL, USA
| | - Manish Kumar
- Department of Neurobiology, Weinberg School of Arts and Sciences, Northwestern University, Evanston, IL, USA
| | - Maomao Chen
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.,Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, China
| | - Mikhail E Matlashov
- Department of Anatomy and Structural Biology and Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Xuan Li
- Department of Anesthesiology, Duke University, Durham, NC, USA
| | - Wei Yang
- Department of Anesthesiology, Duke University, Durham, NC, USA
| | - Liming Nie
- Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, China.,Department of Radiology and Optical Imaging Laboratory, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Daria M Shcherbakova
- Department of Anatomy and Structural Biology and Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Yevgenia Kozorovitskiy
- Department of Neurobiology, Weinberg School of Arts and Sciences, Northwestern University, Evanston, IL, USA.,Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA
| | - Junjie Yao
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Na Ji
- Department of Physics, University of California, Berkeley, Berkeley, CA, USA
| | - Vladislav V Verkhusha
- Department of Anatomy and Structural Biology and Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA. .,Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
44
|
Chenouard N, Xuan F, Tsien RW. Synaptic vesicle traffic is supported by transient actin filaments and regulated by PKA and NO. Nat Commun 2020; 11:5318. [PMID: 33087709 PMCID: PMC7578807 DOI: 10.1038/s41467-020-19120-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 09/25/2020] [Indexed: 11/10/2022] Open
Abstract
Synaptic vesicles (SVs) can be pooled across multiple synapses, prompting questions about their dynamic allocation for neurotransmission and plasticity. We find that the axonal traffic of recycling vesicles is not supported by ubiquitous microtubule-based motility but relies on actin instead. Vesicles freed from synaptic clusters undergo ~1 µm bouts of active transport, initiated by nearby elongation of actin filaments. Long distance translocation arises when successive bouts of active transport were linked by periods of free diffusion. The availability of SVs for active transport can be promptly increased by protein kinase A, a key player in neuromodulation. Vesicle motion is in turn impeded by shutting off axonal actin polymerization, mediated by nitric oxide-cyclic GMP signaling leading to inhibition of RhoA. These findings provide a potential framework for coordinating post-and pre-synaptic strength, using retrograde regulation of axonal actin dynamics to mobilize and recruit presynaptic SV resources.
Collapse
Affiliation(s)
- Nicolas Chenouard
- NYU Neuroscience Institute and Department of Neuroscience and Physiology, NYU Langone Medical Center, New York, NY, 10016, USA.,Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, F-33000, Bordeaux, France
| | - Feng Xuan
- NYU Neuroscience Institute and Department of Neuroscience and Physiology, NYU Langone Medical Center, New York, NY, 10016, USA.,Interdepartmental Neuroscience Program, Northwestern University, Evanston, IL, 60208, USA
| | - Richard W Tsien
- NYU Neuroscience Institute and Department of Neuroscience and Physiology, NYU Langone Medical Center, New York, NY, 10016, USA. .,Center for Neural Science, New York University, New York, NY, 10003, USA.
| |
Collapse
|
45
|
Um JW, Han KA, Choi SY, Ko J. Protocol for Quantitative Analysis of Synaptic Vesicle Clustering in Axons of Cultured Neurons. STAR Protoc 2020; 1:100095. [PMID: 33111124 PMCID: PMC7580093 DOI: 10.1016/j.xpro.2020.100095] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Clustering of synaptic vesicles along the neuronal axons is a critical mechanism underpinning proper synaptic transmission. Here, we provide a detailed protocol for analyzing the distribution of synaptic vesicles in presynaptic boutons of cultured neurons. The protocol covers preparation of cultured neurons, expression of synaptic vesicle-enriched proteins, and quantification procedures. Utilizing neurons from postnatal transgenic mice, this method can be applied to investigate the roles of synaptic genes in regulating vesicle dynamics at synaptic sites. For complete details on the use and execution of this protocol, please refer to Han et al. (2020a).
Collapse
Affiliation(s)
- Ji Won Um
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
- Core Protein Resources Center, DGIST, Daegu 42988, Korea
- Corresponding author
| | - Kyung Ah Han
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
| | - Se-Young Choi
- Department of Physiology and Neuroscience, Dental Research Institute, Seoul National University School of Dentistry, Seoul 03080, Korea
| | - Jaewon Ko
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
- Corresponding author
| |
Collapse
|
46
|
Ciappelloni S, Bouchet D, Dubourdieu N, Boué-Grabot E, Kellermayer B, Manso C, Marignier R, Oliet SHR, Tourdias T, Groc L. Aquaporin-4 Surface Trafficking Regulates Astrocytic Process Motility and Synaptic Activity in Health and Autoimmune Disease. Cell Rep 2020; 27:3860-3872.e4. [PMID: 31242419 DOI: 10.1016/j.celrep.2019.05.097] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 03/08/2019] [Accepted: 05/23/2019] [Indexed: 01/21/2023] Open
Abstract
Astrocytes constantly adapt their ramified morphology in order to support brain cell assemblies. Such plasticity is partly mediated by ion and water fluxes, which rely on the water channel aquaporin-4 (AQP4). The mechanism by which this channel locally contributes to process dynamics has remained elusive. Using a combination of single-molecule and calcium imaging approaches, we here investigated in hippocampal astrocytes the dynamic distribution of the AQP4 isoforms M1 and M23. Surface AQP4-M1 formed small aggregates that contrast with the large AQP4-M23 clusters that are enriched near glutamatergic synapses. Strikingly, stabilizing surface AQP4-M23 tuned the motility of astrocyte processes and favors glutamate synapse activity. Furthermore, human autoantibodies directed against AQP4 from neuromyelitis optica (NMO) patients impaired AQP4-M23 dynamic distribution and, consequently, astrocyte process and synaptic activity. Collectively, it emerges that the membrane dynamics of AQP4 isoform regulate brain cell assemblies in health and autoimmune brain disease targeting AQP4.
Collapse
Affiliation(s)
- Silvia Ciappelloni
- Interdisciplinary Institute for NeuroSciences, CNRS UMR 5297, 33077 Bordeaux, France; Université de Bordeaux, 33077 Bordeaux, France; INSERM U1215, Neurocentre Magendie, 33077 Bordeaux, France
| | - Delphine Bouchet
- Interdisciplinary Institute for NeuroSciences, CNRS UMR 5297, 33077 Bordeaux, France; Université de Bordeaux, 33077 Bordeaux, France
| | - Nadège Dubourdieu
- Université de Bordeaux, 33077 Bordeaux, France; INSERM U1215, Neurocentre Magendie, 33077 Bordeaux, France
| | - Eric Boué-Grabot
- Université de Bordeaux, 33077 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France
| | - Blanka Kellermayer
- Interdisciplinary Institute for NeuroSciences, CNRS UMR 5297, 33077 Bordeaux, France; Université de Bordeaux, 33077 Bordeaux, France
| | - Constance Manso
- Interdisciplinary Institute for NeuroSciences, CNRS UMR 5297, 33077 Bordeaux, France; Université de Bordeaux, 33077 Bordeaux, France
| | - Romain Marignier
- INSERM U1028, CNRS UMR 5292, Center for Research in Neuroscience of Lyon, Lyon, France
| | - Stéphane H R Oliet
- Université de Bordeaux, 33077 Bordeaux, France; INSERM U1215, Neurocentre Magendie, 33077 Bordeaux, France
| | - Thomas Tourdias
- Université de Bordeaux, 33077 Bordeaux, France; INSERM U1215, Neurocentre Magendie, 33077 Bordeaux, France
| | - Laurent Groc
- Interdisciplinary Institute for NeuroSciences, CNRS UMR 5297, 33077 Bordeaux, France; Université de Bordeaux, 33077 Bordeaux, France.
| |
Collapse
|
47
|
Shemesh OA, Linghu C, Piatkevich KD, Goodwin D, Celiker OT, Gritton HJ, Romano MF, Gao R, Yu CCJ, Tseng HA, Bensussen S, Narayan S, Yang CT, Freifeld L, Siciliano CA, Gupta I, Wang J, Pak N, Yoon YG, Ullmann JFP, Guner-Ataman B, Noamany H, Sheinkopf ZR, Park WM, Asano S, Keating AE, Trimmer JS, Reimer J, Tolias AS, Bear MF, Tye KM, Han X, Ahrens MB, Boyden ES. Precision Calcium Imaging of Dense Neural Populations via a Cell-Body-Targeted Calcium Indicator. Neuron 2020; 107:470-486.e11. [PMID: 32592656 DOI: 10.1016/j.neuron.2020.05.029] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 05/09/2019] [Accepted: 05/20/2020] [Indexed: 01/11/2023]
Abstract
Methods for one-photon fluorescent imaging of calcium dynamics can capture the activity of hundreds of neurons across large fields of view at a low equipment complexity and cost. In contrast to two-photon methods, however, one-photon methods suffer from higher levels of crosstalk from neuropil, resulting in a decreased signal-to-noise ratio and artifactual correlations of neural activity. We address this problem by engineering cell-body-targeted variants of the fluorescent calcium indicators GCaMP6f and GCaMP7f. We screened fusions of GCaMP to natural, as well as artificial, peptides and identified fusions that localized GCaMP to within 50 μm of the cell body of neurons in mice and larval zebrafish. One-photon imaging of soma-targeted GCaMP in dense neural circuits reported fewer artifactual spikes from neuropil, an increased signal-to-noise ratio, and decreased artifactual correlation across neurons. Thus, soma-targeting of fluorescent calcium indicators facilitates usage of simple, powerful, one-photon methods for imaging neural calcium dynamics.
Collapse
Affiliation(s)
- Or A Shemesh
- The MIT Media Laboratory, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA; Department of Biological Engineering, MIT, Cambridge, MA, USA; MIT Center for Neurobiological Engineering, MIT, Cambridge, MA, USA; Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA; MIT McGovern Institute for Brain Research, MIT, Cambridge, MA, USA; Department of Neurobiology and Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Changyang Linghu
- The MIT Media Laboratory, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA; MIT Center for Neurobiological Engineering, MIT, Cambridge, MA, USA; MIT McGovern Institute for Brain Research, MIT, Cambridge, MA, USA; Department of Electrical Engineering and Computer Science, MIT, Cambridge, MA, USA
| | - Kiryl D Piatkevich
- The MIT Media Laboratory, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA; Department of Biological Engineering, MIT, Cambridge, MA, USA; MIT Center for Neurobiological Engineering, MIT, Cambridge, MA, USA; Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA; MIT McGovern Institute for Brain Research, MIT, Cambridge, MA, USA; School of Life Sciences, Westlake University, Hangzhou, Zhejiang Province, China
| | - Daniel Goodwin
- The MIT Media Laboratory, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA; MIT Center for Neurobiological Engineering, MIT, Cambridge, MA, USA; MIT McGovern Institute for Brain Research, MIT, Cambridge, MA, USA
| | - Orhan Tunc Celiker
- MIT McGovern Institute for Brain Research, MIT, Cambridge, MA, USA; Department of Electrical Engineering and Computer Science, MIT, Cambridge, MA, USA
| | - Howard J Gritton
- Boston University, Department of Biomedical Engineering, Boston, MA 02215, USA
| | - Michael F Romano
- Boston University, Department of Biomedical Engineering, Boston, MA 02215, USA
| | - Ruixuan Gao
- The MIT Media Laboratory, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA; MIT McGovern Institute for Brain Research, MIT, Cambridge, MA, USA
| | - Chih-Chieh Jay Yu
- The MIT Media Laboratory, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA; Department of Biological Engineering, MIT, Cambridge, MA, USA; MIT McGovern Institute for Brain Research, MIT, Cambridge, MA, USA
| | - Hua-An Tseng
- Boston University, Department of Biomedical Engineering, Boston, MA 02215, USA
| | - Seth Bensussen
- Boston University, Department of Biomedical Engineering, Boston, MA 02215, USA
| | - Sujatha Narayan
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Chao-Tsung Yang
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Limor Freifeld
- The MIT Media Laboratory, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA; MIT Center for Neurobiological Engineering, MIT, Cambridge, MA, USA; MIT McGovern Institute for Brain Research, MIT, Cambridge, MA, USA; Faculty of Biomedical Engineering, Technion, Haifa, Israel
| | - Cody A Siciliano
- Vanderbilt Center for Addiction Research, Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| | - Ishan Gupta
- The MIT Media Laboratory, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA; Department of Biological Engineering, MIT, Cambridge, MA, USA; MIT Center for Neurobiological Engineering, MIT, Cambridge, MA, USA; MIT McGovern Institute for Brain Research, MIT, Cambridge, MA, USA
| | - Joyce Wang
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, MIT, Cambridge, MA 02139, USA
| | - Nikita Pak
- The MIT Media Laboratory, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA; MIT Center for Neurobiological Engineering, MIT, Cambridge, MA, USA; MIT McGovern Institute for Brain Research, MIT, Cambridge, MA, USA; Department of Mechanical Engineering, MIT, Cambridge, MA, USA
| | - Young-Gyu Yoon
- The MIT Media Laboratory, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA; MIT Center for Neurobiological Engineering, MIT, Cambridge, MA, USA; MIT McGovern Institute for Brain Research, MIT, Cambridge, MA, USA; Department of Electrical Engineering and Computer Science, MIT, Cambridge, MA, USA; School of Electrical Engineering, KAIST Institute for Health Science and Technology, Daejeon, Republic of Korea
| | - Jeremy F P Ullmann
- Epilepsy Genetics Program, Department of Neurology, Boston Children's Hospital & Harvard Medical School, Boston, MA, USA
| | - Burcu Guner-Ataman
- The MIT Media Laboratory, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
| | - Habiba Noamany
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, MIT, Cambridge, MA 02139, USA
| | - Zoe R Sheinkopf
- The MIT Media Laboratory, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA; Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
| | | | - Shoh Asano
- Internal Medicine Research Unit, Pfizer, Cambridge, MA, USA
| | - Amy E Keating
- Department of Biological Engineering, MIT, Cambridge, MA, USA; Department of Biology, MIT, Cambridge, MA, USA; Koch Institute, MIT, Cambridge, MA 02139, USA
| | - James S Trimmer
- Department of Physiology and Membrane Biology, University of California, Davis School of Medicine, Davis, CA, USA
| | - Jacob Reimer
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA; Center for Neuroscience and AI, Baylor College of Medicine, Houston, TX, USA
| | - Andreas S Tolias
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA; Center for Neuroscience and AI, Baylor College of Medicine, Houston, TX, USA; Department of Electrical and Computer Engineering, Rice University, Houston, TX, USA
| | - Mark F Bear
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, MIT, Cambridge, MA 02139, USA
| | - Kay M Tye
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, MIT, Cambridge, MA 02139, USA; Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Xue Han
- Boston University, Department of Biomedical Engineering, Boston, MA 02215, USA
| | - Misha B Ahrens
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Edward S Boyden
- The MIT Media Laboratory, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA; Department of Biological Engineering, MIT, Cambridge, MA, USA; MIT Center for Neurobiological Engineering, MIT, Cambridge, MA, USA; Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA; MIT McGovern Institute for Brain Research, MIT, Cambridge, MA, USA; Koch Institute, MIT, Cambridge, MA 02139, USA.
| |
Collapse
|
48
|
Neuregulins 1, 2, and 3 Promote Early Neurite Outgrowth in ErbB4-Expressing Cortical GABAergic Interneurons. Mol Neurobiol 2020; 57:3568-3588. [PMID: 32542595 DOI: 10.1007/s12035-020-01966-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 05/29/2020] [Indexed: 12/27/2022]
Abstract
The neuregulins (Nrgs 1-4) are a family of signaling molecules that play diverse roles in the nervous system. Nrg1 has been implicated in the formation of synapses and in synaptic plasticity. Previous studies have shown Nrg1 can affect neurite outgrowth in several neuronal populations, while the role of Nrg2 and Nrg3 in this process has remained understudied. The Nrgs can bind and activate the ErbB4 receptor tyrosine kinase which is preferentially expressed in GABAergic interneurons in the rodent hippocampus and cerebral cortex. In the present study, we evaluated the effects of Nrgs 1, 2, and 3 on neurite outgrowth of dissociated rat cortical ErbB4-positive (+)/GABA+ interneurons in vitro. All three Nrgs were able to promote neurite outgrowth during the first 2 days in vitro, with increases detected for both the axon (116-120%) and other neurites (100-120%). Increases in the average number of primary and secondary neurites were also observed. Treatment with the Nrgs for an additional 3 days promoted an increase in axonal length (86-96%), with only minimal effects on the remaining neurites (8-13%). ErbB4 expression persisted throughout the dendritic arbor and cell soma at all stages examined, while its expression in the axon was transient and declined with cell maturation. ErbB4 overexpression in GABAergic neurons promoted neurite outgrowth, an effect that was potentiated by Nrg treatment. These results show that Nrgs 1, 2, and 3 are each capable of influencing dendritic and axonal growth at early developmental stages in GABAergic neurons grown in vitro.
Collapse
|
49
|
Li B, Suutari BS, Sun SD, Luo Z, Wei C, Chenouard N, Mandelberg NJ, Zhang G, Wamsley B, Tian G, Sanchez S, You S, Huang L, Neubert TA, Fishell G, Tsien RW. Neuronal Inactivity Co-opts LTP Machinery to Drive Potassium Channel Splicing and Homeostatic Spike Widening. Cell 2020; 181:1547-1565.e15. [PMID: 32492405 DOI: 10.1016/j.cell.2020.05.013] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 01/28/2020] [Accepted: 05/04/2020] [Indexed: 12/21/2022]
Abstract
Homeostasis of neural firing properties is important in stabilizing neuronal circuitry, but how such plasticity might depend on alternative splicing is not known. Here we report that chronic inactivity homeostatically increases action potential duration by changing alternative splicing of BK channels; this requires nuclear export of the splicing factor Nova-2. Inactivity and Nova-2 relocation were connected by a novel synapto-nuclear signaling pathway that surprisingly invoked mechanisms akin to Hebbian plasticity: Ca2+-permeable AMPA receptor upregulation, L-type Ca2+ channel activation, enhanced spine Ca2+ transients, nuclear translocation of a CaM shuttle, and nuclear CaMKIV activation. These findings not only uncover commonalities between homeostatic and Hebbian plasticity but also connect homeostatic regulation of synaptic transmission and neuronal excitability. The signaling cascade provides a full-loop mechanism for a classic autoregulatory feedback loop proposed ∼25 years ago. Each element of the loop has been implicated previously in neuropsychiatric disease.
Collapse
Affiliation(s)
- Boxing Li
- Neuroscience Program, Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine and The Fifth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510810, China; Department of Neuroscience and Physiology, Neuroscience Institute, NYU Grossman Medical Center, New York, NY 10016, USA.
| | - Benjamin S Suutari
- Department of Neuroscience and Physiology, Neuroscience Institute, NYU Grossman Medical Center, New York, NY 10016, USA; Center for Neural Science, New York University, New York, NY 10003, USA
| | - Simón(e) D. Sun
- Department of Neuroscience and Physiology, Neuroscience Institute, NYU Grossman Medical Center, New York, NY 10016, USA; Center for Neural Science, New York University, New York, NY 10003, USA
| | - Zhengyi Luo
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510120, China
| | - Chuanchuan Wei
- Neuroscience Program, Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine and The Fifth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510810, China
| | - Nicolas Chenouard
- Department of Neuroscience and Physiology, Neuroscience Institute, NYU Grossman Medical Center, New York, NY 10016, USA
| | - Nataniel J Mandelberg
- Department of Neuroscience and Physiology, Neuroscience Institute, NYU Grossman Medical Center, New York, NY 10016, USA
| | - Guoan Zhang
- Department of Biochemistry and Molecular Pharmacology and Skirball Institute, NYU Grossman Medical Center, New York, NY 10016, USA
| | - Brie Wamsley
- Department of Neuroscience and Physiology, Neuroscience Institute, NYU Grossman Medical Center, New York, NY 10016, USA; Stanley Center for Psychiatric Research, The Broad Institute, Cambridge, MA 02142, USA
| | - Guoling Tian
- Department of Neuroscience and Physiology, Neuroscience Institute, NYU Grossman Medical Center, New York, NY 10016, USA
| | - Sandrine Sanchez
- Department of Neuroscience and Physiology, Neuroscience Institute, NYU Grossman Medical Center, New York, NY 10016, USA
| | - Sikun You
- Neuroscience Program, Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine and The Fifth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510810, China
| | - Lianyan Huang
- Neuroscience Program, Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine and The Fifth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510810, China
| | - Thomas A Neubert
- Department of Biochemistry and Molecular Pharmacology and Skirball Institute, NYU Grossman Medical Center, New York, NY 10016, USA
| | - Gordon Fishell
- Department of Neuroscience and Physiology, Neuroscience Institute, NYU Grossman Medical Center, New York, NY 10016, USA; Stanley Center for Psychiatric Research, The Broad Institute, Cambridge, MA 02142, USA
| | - Richard W Tsien
- Department of Neuroscience and Physiology, Neuroscience Institute, NYU Grossman Medical Center, New York, NY 10016, USA; Center for Neural Science, New York University, New York, NY 10003, USA.
| |
Collapse
|
50
|
Ripamonti S, Shomroni O, Rhee JS, Chowdhury K, Jahn O, Hellmann KP, Bonn S, Brose N, Tirard M. SUMOylation controls the neurodevelopmental function of the transcription factor Zbtb20. J Neurochem 2020; 154:647-661. [PMID: 32233089 DOI: 10.1111/jnc.15008] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 02/12/2020] [Accepted: 03/10/2020] [Indexed: 12/15/2022]
Abstract
SUMOylation is a dynamic post-translational protein modification that primarily takes place in cell nuclei, where it plays a key role in multiple DNA-related processes. In neurons, the SUMOylation-dependent control of a subset of neuronal transcription factors is known to regulate various aspects of nerve cell differentiation, development, and function. In an unbiased screen for endogenous SUMOylation targets in the developing mouse brain, based on a His6 -HA-SUMO1 knock-in mouse line, we previously identified the transcription factor Zinc finger and BTB domain-containing 20 (Zbtb20) as a new SUMO1-conjugate. We show here that the three key SUMO paralogues SUMO1, SUMO2, and SUMO3 can all be conjugated to Zbtb20 in vitro in HEK293FT cells, and we confirm the SUMOylation of Zbtb20 in vivo in mouse brain. Using primary hippocampal neurons from wild-type and Zbtb20 knock-out (KO) mice as a model system, we then demonstrate that the expression of Zbtb20 is required for proper nerve cell development and neurite growth and branching. Furthermore, we show that the SUMOylation of Zbtb20 is essential for its function in this context, and provide evidence indicating that SUMOylation affects the Zbtb20-dependent transcriptional profile of neurons. Our data highlight the role of SUMOylation in the regulation of neuronal transcription factors that determine nerve cell development, and they demonstrate that key functions of the transcription factor Zbtb20 in neuronal development and neurite growth are under obligatory SUMOylation control.
Collapse
Affiliation(s)
- Silvia Ripamonti
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Orr Shomroni
- NGS Integrative Genomics Core Unit, Department of Human Genetics, University Medical Center Göttingen, Göttingen, Germany
| | - Jeong Seop Rhee
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Kamal Chowdhury
- Max Planck Institute of Biophysical Chemistry, Göttingen, Germany
| | - Olaf Jahn
- Proteomics Group, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Klaus Peter Hellmann
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Stefan Bonn
- Institute of Medical Systems Biology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,German Center for Neurodegenerative Diseases, Tübingen, Germany
| | - Nils Brose
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Marilyn Tirard
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| |
Collapse
|