1
|
Han Y, Hacker D, Donders BC, Parperis C, Thuenauer R, Leterrier C, Grünewald K, Mikhaylova M. Unveiling the cell biology of hippocampal neurons with dendritic axon origin. J Cell Biol 2025; 224:e202403141. [PMID: 39495320 PMCID: PMC11536041 DOI: 10.1083/jcb.202403141] [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: 03/26/2024] [Revised: 08/01/2024] [Accepted: 09/23/2024] [Indexed: 11/05/2024] Open
Abstract
In mammalian axon-carrying-dendrite (AcD) neurons, the axon emanates from a basal dendrite, instead of the soma, to create a privileged route for action potential generation at the axon initial segment (AIS). However, it is unclear how such unusual morphology is established and whether the structure and function of the AIS in AcD neurons are preserved. By using dissociated hippocampal cultures as a model, we show that the development of AcD morphology can occur prior to synaptogenesis and independently of the in vivo environment. A single precursor neurite first gives rise to the axon and then to the AcD. The AIS possesses a similar cytoskeletal architecture as the soma-derived AIS and similarly functions as a trafficking barrier to retain axon-specific molecular composition. However, it does not undergo homeostatic plasticity, contains lesser cisternal organelles, and receives fewer inhibitory inputs. Our findings reveal insights into AcD neuron biology and underscore AIS structural differences based on axon onset.
Collapse
Affiliation(s)
- Yuhao Han
- AG Optobiology, Institute of Biology, Humboldt Universität zu Berlin, Berlin, Germany
- AG “Neuronal Protein Transport”, Centre for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Centre for Structural Systems Biology, Hamburg, Germany
- Structural Cell Biology of Viruses, Leibniz Institute of Virology (LIV), Hamburg, Germany
| | - Daniela Hacker
- AG Optobiology, Institute of Biology, Humboldt Universität zu Berlin, Berlin, Germany
| | | | | | - Roland Thuenauer
- Advanced Light and Fluorescence Microscopy (ALFM) Facility, Centre for Structural Systems Biology, Hamburg, Germany
- Technology Platform Light Microscopy, University of Hamburg, Hamburg, Germany
- Technology Platform Microscopy and Image Analysis (TP MIA), Leibniz Institute of Virology (LIV), Hamburg, Germany
| | | | - Kay Grünewald
- Centre for Structural Systems Biology, Hamburg, Germany
- Structural Cell Biology of Viruses, Leibniz Institute of Virology (LIV), Hamburg, Germany
- Department of Chemistry, University of Hamburg, Hamburg, Germany
| | - Marina Mikhaylova
- AG Optobiology, Institute of Biology, Humboldt Universität zu Berlin, Berlin, Germany
- AG “Neuronal Protein Transport”, Centre for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
2
|
Shukla S, Comerci CJ, Süel GM, Jahed Z. Bioelectronic tools for understanding the universal language of electrical signaling across species and kingdoms. Biosens Bioelectron 2025; 267:116843. [PMID: 39426280 DOI: 10.1016/j.bios.2024.116843] [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: 07/24/2023] [Revised: 09/10/2024] [Accepted: 10/06/2024] [Indexed: 10/21/2024]
Abstract
Modern bioelectronic tools are rapidly advancing to detect electric potentials within networks of electrogenic cells, such as cardiomyocytes, neurons, and pancreatic beta cells. However, it is becoming evident that electrical signaling is not limited to the animal kingdom but may be a universal form of cell-cell communication. In this review, we discuss the existing evidence of, and tools used to collect, subcellular, single-cell and network-level electrical signals across kingdoms, including bacteria, plants, fungi, and even viruses. We discuss how cellular networks employ altered electrical "circuitry" and intercellular mechanisms across kingdoms, and we assess the functionality and scalability of cutting-edge nanobioelectronics to collect electrical signatures regardless of cell size, shape, or function. Researchers today aim to design micro- and nano-topographic structures which harness mechanosensitive membrane and cytoskeletal pathways that enable tight electrical coupling to subcellular compartments within high-throughput recording systems. Finally, we identify gaps in current knowledge of inter-species and inter-kingdom electrical signaling and propose critical milestones needed to create a central theory of electrical signaling across kingdoms. Our discussion demonstrates the need for high resolution, high throughput tools which can probe multiple, diverse cell types at once in their native or experimentally-modeled environments. These advancements will not only reveal the underlying biophysical laws governing the universal language of electrical communication, but can enable bidirectional electrical communication and manipulation of biological systems.
Collapse
Affiliation(s)
- Shivani Shukla
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, CA, United States; Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, University of California San Diego, La Jolla, CA, United States
| | - Colin J Comerci
- Department of Molecular Biology, University of California San Diego, La Jolla, CA, United States
| | - Gürol M Süel
- Department of Molecular Biology, University of California San Diego, La Jolla, CA, United States
| | - Zeinab Jahed
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, CA, United States; Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, University of California San Diego, La Jolla, CA, United States.
| |
Collapse
|
3
|
Chen M, Liu B. KCl enhances the cryoablation-induced antitumor immune response: A hepatocellular carcinoma murine model research. Cryobiology 2024; 117:105164. [PMID: 39536961 DOI: 10.1016/j.cryobiol.2024.105164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 11/07/2024] [Accepted: 11/09/2024] [Indexed: 11/16/2024]
Abstract
Cryoablation is a valuable treatment for liver cancer. To investigate the effect of KCl solution on the immunological response post cryoablation, we created a tumor-bearing mice model by subcutaneously implanting Hepal-6 cells in adult Balb/c mice. Subsequently, the mice were randomly assigned to three groups: group A (sham cryoablation), group B (cryoablation), and group C (cryoablation plus KCl solution). Mice were sacrificed on days 0, 7, and 14 post-treatment. Immune cell populations were assessed using flow cytometry. Blood samples were analyzed for serum IL-4, HSP70, and TGF-β1 levels with ELISA assays. Ablated tissues stained with immunohistochemistry were utilized to evaluate Ki67 expression at the margins of the ablation site. Our findings revealed higher HSP70 expression levels in groups B and C compared to group A. Cryoablation triggered an immune response, which was enhanced by KCl. On days 0, 7, and 14, the percentages of CD4+ T cells, CD8+ T cells, and NK cells in the spleen of group C were significantly increased compared with groups A and B. Additionally, the Th1/Th2 ratio was significantly increased in group C. Serum TGF-β1 expression was elevated after cryoablation, but KCl solution reduced the high TGF-β1 expression after cryoablation and decreased the invasiveness of cancer cells. Finally, the proliferative activity of untreated tumor tissue was significantly reduced in group C compared to groups A and B. In summary, Cryoablation triggered a systemic immune response in tumor-bearing mice, which was further boosted by combining cryoablation with a KCl solution.
Collapse
Affiliation(s)
- Mu Chen
- Institute of Biothermal Science and Technology, University of Shanghai for Science and Technology, Shanghai, China; School of Biomedical Engineering, Guangzhou Xinhua University, Guangzhou, China
| | - Baolin Liu
- Institute of Biothermal Science and Technology, University of Shanghai for Science and Technology, Shanghai, China; Shanghai Technical Service Platform for Cryopreservation of Biological Resources, Shanghai, China; Shanghai Collaborative Innovation Center for Tumor Treatment with Energy, Shanghai, China.
| |
Collapse
|
4
|
Follmer ML, Isner TJ, Ozekin YH, Levitt CH, Burek CL, Benninger RKP, Bates EA. Depolarization induces calcium-dependent BMP4 release from mouse embryonic palate mesenchymal cells. Nat Commun 2024; 15:9806. [PMID: 39532850 PMCID: PMC11558011 DOI: 10.1038/s41467-024-53642-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 10/17/2024] [Indexed: 11/16/2024] Open
Abstract
Bone Morphogenetic Protein (BMP) signaling is essential for craniofacial development, though little is known about the mechanisms that govern BMP secretion. We show that depolarization induces calcium-dependent BMP4 release from mouse embryonic palate mesenchyme. We show endogenous transient changes in intracellular calcium occur in cranial neural crest cells, the cells from which embryonic palate mesenchyme derives. Waves of transient changes in intracellular calcium suggest that these cells are electrically coupled and may temporally coordinate BMP release. These transient changes in intracellular calcium persist in palate mesenchyme cells from embryonic day 9.5 to 13.5 mice. Disruption of a potassium channel called Kcnj2 significantly decreases the amplitude of calcium transients and the ability of cells to secrete BMP. Kcnj2 knockout mice have cleft palate and reduced BMP signaling. Our data suggest that temporal control of developmental cues is regulated by ion channels, depolarization, and intracellular calcium for mammalian craniofacial morphogenesis.
Collapse
Affiliation(s)
- Mikaela L Follmer
- Department of Pediatrics, Section of Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Trevor J Isner
- Department of Pediatrics, Section of Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Yunus H Ozekin
- Department of Pediatrics, Section of Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Claire H Levitt
- Department of Bioengineering, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA
| | - Carolyn L Burek
- Department of Pediatrics, Section of Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Richard K P Benninger
- Department of Bioengineering, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA
| | - Emily Anne Bates
- Department of Pediatrics, Section of Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
5
|
Borgarelli C, Klingl YE, Escamilla-Ayala A, Scarponi C, La Rovere RML, Stoklund Dittlau K, Bultynck G, Sampaolesi M, Schoenberger M, Munck S, Van Den Bosch L, De Borggraeve WM, Ismalaj E. Novel Far-Red Fluorescent 1,4-Dihydropyridines for L-Type Calcium Channel Imaging. J Med Chem 2024; 67:18038-18052. [PMID: 39388369 DOI: 10.1021/acs.jmedchem.4c00565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Upregulation of L-type calcium channels (LTCCs) is implicated in a range of cardiovascular and neurological disorders. Therefore, the development of toolboxes that unlock fast imaging protocols in live cells is coveted. Herein, we report a library of first-in-class far-red small-molecule-based fluorescent ligands (FluoDiPines), able to target LTCCs. All fluorescent ligands were evaluated in whole-cell patch-clamp and live-cell Ca2+ imaging whereby FluoDiPine 6 was found to be the best candidate for live-cell fluorescence imaging. Low concentration of FluoDiPine 6 (50 nM) and a quick labeling protocol (5 min) are successfully applied to fixed and live cells to image LTCCs with good specificity.
Collapse
Affiliation(s)
- Carlotta Borgarelli
- Sustainable Chemistry for Metals and Molecules (SCM2), Department of Chemistry, KU Leuven, Celestijnenlaan 200F, Box 2404, 3001 Leuven, Belgium
| | - Yvonne E Klingl
- Department of Neurosciences, Experimental Neurology, and Leuven Brain Institute (LBI), KU Leuven-University of Leuven, 3000 Leuven, Belgium
- VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, 3000 Leuven, Belgium
| | - Abril Escamilla-Ayala
- Center for Brain & Disease Research, & VIB BioImaging Core, VIB-KU Leuven Campus Gasthuisberg O&N5, box 602 Herestraat 49, 3000 Leuven, Belgium
- Department of Neurosciences, Leuven Brain Institute, KU Leuven, Campus Gasthuisberg O&N5-box 602 Herestraat 49, 3000 Leuven, Belgium
| | - Carlotta Scarponi
- CIC biomaGUNE, Basque Research and Technology Alliance (BRTA) Paseo Miramon, 20014 San Sebastian, Guipuzcoa, Spain
| | - Rita M L La Rovere
- Dep. Cellular & Molecular Medicine, KU Leuven, Lab. Molecular & Cellular Signaling, Campus Gasthuisberg O/N-1 bus 802, Herestraat 49, 3000 Leuven, Belgium
| | - Katarina Stoklund Dittlau
- Department of Neurosciences, Experimental Neurology, and Leuven Brain Institute (LBI), KU Leuven-University of Leuven, 3000 Leuven, Belgium
- VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, 3000 Leuven, Belgium
| | - Geert Bultynck
- Dep. Cellular & Molecular Medicine, KU Leuven, Lab. Molecular & Cellular Signaling, Campus Gasthuisberg O/N-1 bus 802, Herestraat 49, 3000 Leuven, Belgium
| | - Maurilio Sampaolesi
- Translational Cardiomyology Laboratory, Stem Cell Biology and Embryology, Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
| | - Matthias Schoenberger
- Department for Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium
| | - Sebastian Munck
- Center for Brain & Disease Research, & VIB BioImaging Core, VIB-KU Leuven Campus Gasthuisberg O&N5, box 602 Herestraat 49, 3000 Leuven, Belgium
- Department of Neurosciences, Leuven Brain Institute, KU Leuven, Campus Gasthuisberg O&N5-box 602 Herestraat 49, 3000 Leuven, Belgium
| | - Ludo Van Den Bosch
- Department of Neurosciences, Experimental Neurology, and Leuven Brain Institute (LBI), KU Leuven-University of Leuven, 3000 Leuven, Belgium
- VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, 3000 Leuven, Belgium
| | - Wim M De Borggraeve
- Sustainable Chemistry for Metals and Molecules (SCM2), Department of Chemistry, KU Leuven, Celestijnenlaan 200F, Box 2404, 3001 Leuven, Belgium
| | - Ermal Ismalaj
- Sustainable Chemistry for Metals and Molecules (SCM2), Department of Chemistry, KU Leuven, Celestijnenlaan 200F, Box 2404, 3001 Leuven, Belgium
- CIC biomaGUNE, Basque Research and Technology Alliance (BRTA) Paseo Miramon, 20014 San Sebastian, Guipuzcoa, Spain
| |
Collapse
|
6
|
Martinelli E, Akouissi O, Liebi L, Furfaro I, Maulà D, Savoia N, Remy A, Nikles L, Roux A, Stoppini L, Lacour SP. The e-Flower: A hydrogel-actuated 3D MEA for brain spheroid electrophysiology. SCIENCE ADVANCES 2024; 10:eadp8054. [PMID: 39413178 PMCID: PMC11482305 DOI: 10.1126/sciadv.adp8054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 09/12/2024] [Indexed: 10/18/2024]
Abstract
Traditional microelectrode arrays (MEAs) are limited to measuring electrophysiological activity in two dimensions, failing to capture the complexity of three-dimensional (3D) tissues such as neural organoids and spheroids. Here, we introduce a flower-shaped MEA (e-Flower) that can envelop submillimeter brain spheroids following actuation by the sole addition of the cell culture medium. Inspired by soft microgrippers, its actuation mechanism leverages the swelling properties of a polyacrylic acid hydrogel grafted to a polyimide substrate hosting the electrical interconnects. Compatible with standard electrophysiology recording systems, the e-Flower does not require additional equipment or solvents and is ready to use with preformed 3D tissues. We designed an e-Flower achieving a curvature as low as 300 micrometers within minutes, a value tunable by the choice of reswelling media and hydrogel cross-linker concentration. Furthermore, we demonstrate the ability of the e-Flower to detect spontaneous neural activity across the spheroid surface, demonstrating its potential for comprehensive neural signal recording.
Collapse
Affiliation(s)
- Eleonora Martinelli
- Laboratory for Soft Bioelectronic Interfaces, Neuro-X Institute, École Polytechnique Fédérale de Lausanne (EPFL), Geneva, Switzerland
| | - Outman Akouissi
- Laboratory for Soft Bioelectronic Interfaces, Neuro-X Institute, École Polytechnique Fédérale de Lausanne (EPFL), Geneva, Switzerland
- Bertarelli Foundation Chair in Translational NeuroEngineering, Neuro-X Institute, École Polytechnique Fédérale de Lausanne (EPFL), Geneva, Switzerland
| | - Luca Liebi
- Laboratory for Soft Bioelectronic Interfaces, Neuro-X Institute, École Polytechnique Fédérale de Lausanne (EPFL), Geneva, Switzerland
| | - Ivan Furfaro
- Laboratory for Soft Bioelectronic Interfaces, Neuro-X Institute, École Polytechnique Fédérale de Lausanne (EPFL), Geneva, Switzerland
| | - Desirée Maulà
- Laboratory for Soft Bioelectronic Interfaces, Neuro-X Institute, École Polytechnique Fédérale de Lausanne (EPFL), Geneva, Switzerland
| | - Nathan Savoia
- Laboratory for Soft Bioelectronic Interfaces, Neuro-X Institute, École Polytechnique Fédérale de Lausanne (EPFL), Geneva, Switzerland
| | - Antoine Remy
- Laboratory for Soft Bioelectronic Interfaces, Neuro-X Institute, École Polytechnique Fédérale de Lausanne (EPFL), Geneva, Switzerland
| | - Laetitia Nikles
- Tissue Engineering Laboratory, HEPIA HES-SO University of Applied Sciences and Arts Western Switzerland, Geneva, Switzerland
| | - Adrien Roux
- Tissue Engineering Laboratory, HEPIA HES-SO University of Applied Sciences and Arts Western Switzerland, Geneva, Switzerland
| | - Luc Stoppini
- Tissue Engineering Laboratory, HEPIA HES-SO University of Applied Sciences and Arts Western Switzerland, Geneva, Switzerland
| | - Stéphanie P. Lacour
- Laboratory for Soft Bioelectronic Interfaces, Neuro-X Institute, École Polytechnique Fédérale de Lausanne (EPFL), Geneva, Switzerland
| |
Collapse
|
7
|
Ogawa Y, Tsugita S, Torii Y, Iwamoto H, Sato T, Kasahara J, Takeuchi M, Kuwabara T, Iiyama M, Takayanagi T, Mizuguchi H. Microdialysis-integrated HPLC system with dual-electrode detection using track-etched membrane electrodes for in vivo monitoring of dopamine dynamics. J Chromatogr B Analyt Technol Biomed Life Sci 2024; 1247:124318. [PMID: 39299150 DOI: 10.1016/j.jchromb.2024.124318] [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: 07/14/2024] [Revised: 09/08/2024] [Accepted: 09/10/2024] [Indexed: 09/22/2024]
Abstract
A capillary high-performance liquid chromatography (HPLC) system equipped with a dual-electrode detector utilizing track-etched membrane electrodes (TEMEs) was combined with a microdialysis sampling setup. The electrochemical detector benefits from the high electrolysis efficiency of TEMEs, allowing for calibration-free coulometric detection and simplifying data analysis to determine the dopamine recovery through a dialysis probe. Additionally, this system was used for in vivo monitoring of dopamine in the right striatum of a mouse brain. Temporal changes in dopamine levels, including an exponential decay immediately after the dialysis probe insertion and an excess release of dopamine induced by a high concentration of potassium ions, confirmed the system's proper operation. Furthermore, subsequent measurements following the intraperitoneal injection of mirtazapine showed no increase in dopamine levels in the right dorsal striatum. The dual-electrode system displayed characteristic dopamine detection behavior, with anodic and cathodic peak pairs indicative of reversible electrochemical reactions. This capability facilitated the identification of the dopamine peak within the complex chromatogram of the mouse brain dialysate. The consistency between dopamine collection efficiency from standard solutions and dialysate indicated the absence of interfering electroactive substances overlapping with the dopamine peak in the chromatogram. This integrated analysis system successfully tracked temporal fluctuations in dopamine concentration within the mouse brain.
Collapse
Affiliation(s)
- Yukuto Ogawa
- Department of Applied Chemistry, Graduate School of Science and Technology, Tokushima University, Tokushima 770-8506, Japan
| | - Sohei Tsugita
- Department of Applied Chemistry, Graduate School of Science and Technology, Tokushima University, Tokushima 770-8506, Japan
| | - Yuka Torii
- Department of Applied Chemistry, Graduate School of Science and Technology, Tokushima University, Tokushima 770-8506, Japan
| | - Hiten Iwamoto
- Faculty of Pharmaceutical Sciences, Tokushima University, 1-78-1 Shomachi, Tokushima 770-8505, Japan
| | - Tsukasa Sato
- Faculty of Pharmaceutical Sciences, Tokushima University, 1-78-1 Shomachi, Tokushima 770-8505, Japan
| | - Jiro Kasahara
- Graduate School of Biomedical Sciences, Tokushima University, 1-78-1 Shomachi, Tokushima 770-8505, Japan
| | - Masaki Takeuchi
- Graduate School of Biomedical Sciences, Tokushima University, 1-78-1 Shomachi, Tokushima 770-8505, Japan
| | - Tomohiko Kuwabara
- Department of Applied Chemistry, Graduate School of Science and Technology, Tokushima University, Tokushima 770-8506, Japan
| | - Masamitsu Iiyama
- Nomura Micro Science Co., Ltd., 2-9-10 Okada, Atsugi, Kanagawa 243-0021, Japan
| | - Toshio Takayanagi
- Department of Applied Chemistry, Graduate School of Science and Technology, Tokushima University, Tokushima 770-8506, Japan
| | - Hitoshi Mizuguchi
- Department of Applied Chemistry, Graduate School of Science and Technology, Tokushima University, Tokushima 770-8506, Japan.
| |
Collapse
|
8
|
Jiang J, Liu S, Xu Z, Yu S, Wang L, Long S, Ye S, Yan Y, Xu H, Zhang J, Wei W, Zhao Q, Li X. Transcriptome-Wide Profiling of Nascent RNA in Neurons with Enriched H3K27ac Signal Elevates eRNA Identification Efficiency. ACS Chem Neurosci 2024; 15. [PMID: 39377285 PMCID: PMC11487572 DOI: 10.1021/acschemneuro.4c00047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 08/17/2024] [Accepted: 08/19/2024] [Indexed: 10/09/2024] Open
Abstract
Growing evidence suggests that activity-dependent gene expression is crucial for neuronal plasticity and behavioral experience. Enhancer RNAs (eRNAs), a class of long noncoding RNAs, play a key role in these processes. However, eRNAs are highly dynamic and are often present at lower levels than their corresponding mRNAs, making them difficult to detect using total RNA-seq techniques. Nascent RNA sequencing, which separates nascent RNAs from the steady-state RNA population, has been shown to increase the ability to detect activity-induced eRNAs with a higher signal-to-noise ratio. However, there is a lack of bioinformatic tools or pipelines for detecting eRNAs utilizing nascent RNA-seq and other multiomics data sets. In this study, we addressed this gap by developing a novel bioinformatic framework, e-finder, for finding eRNAs and have made it available to the scientific community. Additionally, we reanalyzed our previous nascent RNA sequencing data and compared them with total RNA-seq data to identify activity-regulated RNAs in neuronal cell populations. Using H3K27 acetylome data, we characterized activity-dependent eRNAs that drive the transcriptional activity of the target genes. Our analysis identified a subset of eRNAs involved in mediating synapse organization, which showed increased activity-dependent transcription after the potassium chloride stimulation. Notably, our data suggest that nascent RNA-seq with an enriched H3K27ac signal exhibits high resolution to identify potential eRNAs in response to membrane depolarization. Our findings uncover the role of the eRNA-mediated gene activation network in neuronal systems, providing new insights into the molecular processes characterizing neurological diseases.
Collapse
Affiliation(s)
- Jiazhi Jiang
- Brain
Research Center, Zhongnan Hosptial of Wuhan
University, Wuhan 430071, China
- Department
of Neurosurgery, Zhongnan Hospital of Wuhan
University, Wuhan 430071, China
| | - Sha Liu
- Brain
Research Center, Zhongnan Hosptial of Wuhan
University, Wuhan 430071, China
- Department
of General Practice, Zhongnan Hospital of
Wuhan University, Wuhan 430071, China
| | - Ziyue Xu
- Brain
Research Center, Zhongnan Hosptial of Wuhan
University, Wuhan 430071, China
| | - Shuangqi Yu
- Brain
Research Center, Zhongnan Hosptial of Wuhan
University, Wuhan 430071, China
| | - Lesheng Wang
- Brain
Research Center, Zhongnan Hosptial of Wuhan
University, Wuhan 430071, China
| | - Shengrong Long
- Brain
Research Center, Zhongnan Hosptial of Wuhan
University, Wuhan 430071, China
| | - Shengda Ye
- Brain
Research Center, Zhongnan Hosptial of Wuhan
University, Wuhan 430071, China
- Department
of Neurosurgery, Zhongnan Hospital of Wuhan
University, Wuhan 430071, China
| | - Yu Yan
- Department
of Neurosurgery, Zhongnan Hospital of Wuhan
University, Wuhan 430071, China
| | - Hongyu Xu
- Brain
Research Center, Zhongnan Hosptial of Wuhan
University, Wuhan 430071, China
| | - Jianjian Zhang
- Department
of Neurosurgery, Zhongnan Hospital of Wuhan
University, Wuhan 430071, China
| | - Wei Wei
- Brain
Research Center, Zhongnan Hosptial of Wuhan
University, Wuhan 430071, China
- Department
of Neurosurgery, Zhongnan Hospital of Wuhan
University, Wuhan 430071, China
| | - Qiongyi Zhao
- Cognitive
Neuroepigenetics Laboratory, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Xiang Li
- Brain
Research Center, Zhongnan Hosptial of Wuhan
University, Wuhan 430071, China
- Department
of Neurosurgery, Zhongnan Hospital of Wuhan
University, Wuhan 430071, China
- Frontier
Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430071, China
- Medical
Research
Institute, Wuhan University, Wuhan 430071, China
- Sino-Italian
Ascula Brain Science Joint Laboratory, Zhongnan
Hosptial of Wuhan University, Wuhan 430071, China
| |
Collapse
|
9
|
Oliva MK, Bourke J, Kornienko D, Mattei C, Mao M, Kuanyshbek A, Ovchinnikov D, Bryson A, Karle TJ, Maljevic S, Petrou S. Standardizing a method for functional assessment of neural networks in brain organoids. J Neurosci Methods 2024; 409:110178. [PMID: 38825241 DOI: 10.1016/j.jneumeth.2024.110178] [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: 02/15/2024] [Revised: 04/05/2024] [Accepted: 05/22/2024] [Indexed: 06/04/2024]
Abstract
During the last decade brain organoids have emerged as an attractive model system, allowing stem cells to be differentiated into complex 3D models, recapitulating many aspects of human brain development. Whilst many studies have analysed anatomical and cytoarchitectural characteristics of organoids, their functional characterisation has been limited, and highly variable between studies. Standardised, consistent methods for recording functional activity are critical to providing a functional understanding of neuronal networks at the synaptic and network level that can yield useful information about functional network phenotypes in disease and healthy states. In this study we outline a detailed methodology for calcium imaging and Multi-Electrode Array (MEA) recordings in brain organoids. To illustrate the utility of these functional interrogation techniques in uncovering induced differences in neural network activity we applied various stimulating media protocols. We demonstrate overlapping information from the two modalities, with comparable numbers of active cells in the four treatment groups and an increase in synchronous behaviour in BrainPhys treated groups. Further development of analysis pipelines to reveal network level changes in brain organoids will enrich our understanding of network formation and perturbation in these structures, and aid in the future development of drugs that target neurological disorders at the network level.
Collapse
Affiliation(s)
- M K Oliva
- Ion Channels and Diseases Group, The Florey, The University of Melbourne, Parkville, VIC 3052, Australia.
| | - J Bourke
- Ion Channels and Diseases Group, The Florey, The University of Melbourne, Parkville, VIC 3052, Australia
| | - D Kornienko
- Ion Channels and Diseases Group, The Florey, The University of Melbourne, Parkville, VIC 3052, Australia
| | - C Mattei
- Ion Channels and Diseases Group, The Florey, The University of Melbourne, Parkville, VIC 3052, Australia
| | - M Mao
- Ion Channels and Diseases Group, The Florey, The University of Melbourne, Parkville, VIC 3052, Australia
| | - A Kuanyshbek
- Ion Channels and Diseases Group, The Florey, The University of Melbourne, Parkville, VIC 3052, Australia
| | - D Ovchinnikov
- Ion Channels and Diseases Group, The Florey, The University of Melbourne, Parkville, VIC 3052, Australia
| | - A Bryson
- Ion Channels and Diseases Group, The Florey, The University of Melbourne, Parkville, VIC 3052, Australia
| | - T J Karle
- Ion Channels and Diseases Group, The Florey, The University of Melbourne, Parkville, VIC 3052, Australia
| | - S Maljevic
- Ion Channels and Diseases Group, The Florey, The University of Melbourne, Parkville, VIC 3052, Australia
| | - S Petrou
- Ion Channels and Diseases Group, The Florey, The University of Melbourne, Parkville, VIC 3052, Australia; Praxis Precision Medicines, Inc., Cambridge, MA 02142, USA
| |
Collapse
|
10
|
Cerda-Jara CA, Kim SJ, Thomas G, Farsi Z, Zolotarov G, Dube G, Deter A, Bahry E, Georgii E, Woehler A, Piwecka M, Rajewsky N. miR-7 controls glutamatergic transmission and neuronal connectivity in a Cdr1as-dependent manner. EMBO Rep 2024; 25:3008-3039. [PMID: 38831125 PMCID: PMC11239925 DOI: 10.1038/s44319-024-00168-9] [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: 01/04/2024] [Revised: 04/12/2024] [Accepted: 05/14/2024] [Indexed: 06/05/2024] Open
Abstract
The circular RNA (circRNA) Cdr1as is conserved across mammals and highly expressed in neurons, where it directly interacts with microRNA miR-7. However, the biological function of this interaction is unknown. Here, using primary cortical murine neurons, we demonstrate that stimulating neurons by sustained depolarization rapidly induces two-fold transcriptional upregulation of Cdr1as and strong post-transcriptional stabilization of miR-7. Cdr1as loss causes doubling of glutamate release from stimulated synapses and increased frequency and duration of local neuronal bursts. Moreover, the periodicity of neuronal networks increases, and synchronicity is impaired. Strikingly, these effects are reverted by sustained expression of miR-7, which also clears Cdr1as molecules from neuronal projections. Consistently, without Cdr1as, transcriptomic changes caused by miR-7 overexpression are stronger (including miR-7-targets downregulation) and enriched in secretion/synaptic plasticity pathways. Altogether, our results suggest that in cortical neurons Cdr1as buffers miR-7 activity to control glutamatergic excitatory transmission and neuronal connectivity important for long-lasting synaptic adaptations.
Collapse
Affiliation(s)
- Cledi A Cerda-Jara
- Laboratory for Systems Biology of Gene Regulatory Elements, Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, Hannoversche Str. 28, 10115, Berlin, Germany
| | - Seung Joon Kim
- Laboratory for Systems Biology of Gene Regulatory Elements, Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, Hannoversche Str. 28, 10115, Berlin, Germany
| | - Gwendolin Thomas
- Laboratory for Systems Biology of Gene Regulatory Elements, Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, Hannoversche Str. 28, 10115, Berlin, Germany
| | - Zohreh Farsi
- Light Microscopy Platform, Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, Hannoversche Str. 28, 10115, Berlin, Germany
| | - Grygoriy Zolotarov
- Laboratory for Systems Biology of Gene Regulatory Elements, Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, Hannoversche Str. 28, 10115, Berlin, Germany
| | - Giuliana Dube
- Laboratory for Systems Biology of Gene Regulatory Elements, Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, Hannoversche Str. 28, 10115, Berlin, Germany
| | - Aylina Deter
- Laboratory for Systems Biology of Gene Regulatory Elements, Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, Hannoversche Str. 28, 10115, Berlin, Germany
| | - Ella Bahry
- Helmholtz Imaging, Max-Delbrück-Center for Molecular Medicine, Berlin, Germany Hannoversche Str. 28, 10115, Berlin, Germany
| | - Elisabeth Georgii
- Helmholtz AI, Helmholtz Zentrum München, Ingolstädter Landstraße 1, D-85764, Neuherberg, Germany
| | - Andrew Woehler
- Light Microscopy Platform, Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, Hannoversche Str. 28, 10115, Berlin, Germany
| | - Monika Piwecka
- Department of Non-Coding RNAs, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14, 61-704, Poznan, Poland
| | - Nikolaus Rajewsky
- Laboratory for Systems Biology of Gene Regulatory Elements, Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, Hannoversche Str. 28, 10115, Berlin, Germany.
| |
Collapse
|
11
|
Kelly AR, Glover DJ. Information Transmission through Biotic-Abiotic Interfaces to Restore or Enhance Human Function. ACS APPLIED BIO MATERIALS 2024; 7:3605-3628. [PMID: 38729914 DOI: 10.1021/acsabm.4c00435] [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: 05/12/2024]
Abstract
Advancements in reliable information transfer across biotic-abiotic interfaces have enabled the restoration of lost human function. For example, communication between neuronal cells and electrical devices restores the ability to walk to a tetraplegic patient and vision to patients blinded by retinal disease. These impactful medical achievements are aided by tailored biotic-abiotic interfaces that maximize information transfer fidelity by considering the physical properties of the underlying biological and synthetic components. This Review develops a modular framework to define and describe the engineering of biotic and abiotic components as well as the design of interfaces to facilitate biotic-abiotic information transfer using light or electricity. Delineating the properties of the biotic, interface, and abiotic components that enable communication can serve as a guide for future research in this highly interdisciplinary field. Application of synthetic biology to engineer light-sensitive proteins has facilitated the control of neural signaling and the restoration of rudimentary vision after retinal blindness. Electrophysiological methodologies that use brain-computer interfaces and stimulating implants to bypass spinal column injuries have led to the rehabilitation of limb movement and walking ability. Cellular interfacing methodologies and on-chip learning capability have been made possible by organic transistors that mimic the information processing capacity of neurons. The collaboration of molecular biologists, material scientists, and electrical engineers in the emerging field of biotic-abiotic interfacing will lead to the development of prosthetics capable of responding to thought and experiencing touch sensation via direct integration into the human nervous system. Further interdisciplinary research will improve electrical and optical interfacing technologies for the restoration of vision, offering greater visual acuity and potentially color vision in the near future.
Collapse
Affiliation(s)
- Alexander R Kelly
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Dominic J Glover
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| |
Collapse
|
12
|
Benevento M, Alpár A, Gundacker A, Afjehi L, Balueva K, Hevesi Z, Hanics J, Rehman S, Pollak DD, Lubec G, Wulff P, Prevot V, Horvath TL, Harkany T. A brainstem-hypothalamus neuronal circuit reduces feeding upon heat exposure. Nature 2024; 628:826-834. [PMID: 38538787 PMCID: PMC11041654 DOI: 10.1038/s41586-024-07232-3] [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: 08/30/2022] [Accepted: 02/22/2024] [Indexed: 04/06/2024]
Abstract
Empirical evidence suggests that heat exposure reduces food intake. However, the neurocircuit architecture and the signalling mechanisms that form an associative interface between sensory and metabolic modalities remain unknown, despite primary thermoceptive neurons in the pontine parabrachial nucleus becoming well characterized1. Tanycytes are a specialized cell type along the wall of the third ventricle2 that bidirectionally transport hormones and signalling molecules between the brain's parenchyma and ventricular system3-8. Here we show that tanycytes are activated upon acute thermal challenge and are necessary to reduce food intake afterwards. Virus-mediated gene manipulation and circuit mapping showed that thermosensing glutamatergic neurons of the parabrachial nucleus innervate tanycytes either directly or through second-order hypothalamic neurons. Heat-dependent Fos expression in tanycytes suggested their ability to produce signalling molecules, including vascular endothelial growth factor A (VEGFA). Instead of discharging VEGFA into the cerebrospinal fluid for a systemic effect, VEGFA was released along the parenchymal processes of tanycytes in the arcuate nucleus. VEGFA then increased the spike threshold of Flt1-expressing dopamine and agouti-related peptide (Agrp)-containing neurons, thus priming net anorexigenic output. Indeed, both acute heat and the chemogenetic activation of glutamatergic parabrachial neurons at thermoneutrality reduced food intake for hours, in a manner that is sensitive to both Vegfa loss-of-function and blockage of vesicle-associated membrane protein 2 (VAMP2)-dependent exocytosis from tanycytes. Overall, we define a multimodal neurocircuit in which tanycytes link parabrachial sensory relay to the long-term enforcement of a metabolic code.
Collapse
Affiliation(s)
- Marco Benevento
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Alán Alpár
- Department of Anatomy, Histology, and Embryology, Semmelweis University, Budapest, Hungary
- SE NAP Research Group of Experimental Neuroanatomy and Developmental Biology, Semmelweis University, Budapest, Hungary
| | - Anna Gundacker
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Leila Afjehi
- Programme Proteomics, Paracelsus Medizinische Privatuniversität, Salzburg, Austria
| | - Kira Balueva
- Institute of Physiology, Christian Albrechts University, Kiel, Germany
| | - Zsofia Hevesi
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - János Hanics
- Department of Anatomy, Histology, and Embryology, Semmelweis University, Budapest, Hungary
- SE NAP Research Group of Experimental Neuroanatomy and Developmental Biology, Semmelweis University, Budapest, Hungary
| | - Sabah Rehman
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Daniela D Pollak
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Gert Lubec
- Programme Proteomics, Paracelsus Medizinische Privatuniversität, Salzburg, Austria
| | - Peer Wulff
- Institute of Physiology, Christian Albrechts University, Kiel, Germany
| | - Vincent Prevot
- University of Lille, INSERM, CHU Lille, Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience and Cognition, UMR S1172, EGID, Lille, France
| | - Tamas L Horvath
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Tibor Harkany
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria.
- Department of Neuroscience, Karolinska Institutet, Solna, Sweden.
| |
Collapse
|
13
|
Waltz TB, Chao D, Prodoehl EK, Enders JD, Ehlers VL, Dharanikota BS, Dahms NM, Isaeva E, Hogan QH, Pan B, Stucky CL. Fabry disease Schwann cells release p11 to induce sensory neuron hyperactivity. JCI Insight 2024; 9:e172869. [PMID: 38646936 PMCID: PMC11141882 DOI: 10.1172/jci.insight.172869] [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: 06/06/2023] [Accepted: 03/05/2024] [Indexed: 04/25/2024] Open
Abstract
Patients with Fabry disease suffer from chronic debilitating pain and peripheral sensory neuropathy with minimal treatment options, but the cellular drivers of this pain are unknown. Here, we propose a mechanism we believe to be novel in which altered signaling between Schwann cells and sensory neurons underlies the peripheral sensory nerve dysfunction we observed in a genetic rat model of Fabry disease. Using in vivo and in vitro electrophysiological recordings, we demonstrated that Fabry rat sensory neurons exhibited pronounced hyperexcitability. Schwann cells probably contributed to this finding because application of mediators released from cultured Fabry Schwann cells induced spontaneous activity and hyperexcitability in naive sensory neurons. We examined putative algogenic mediators using proteomic analysis and found that Fabry Schwann cells released elevated levels of the protein p11 (S100A10), which induced sensory neuron hyperexcitability. Removal of p11 from Fabry Schwann cell media caused hyperpolarization of neuronal resting membrane potentials, indicating that p11 may contribute to the excessive neuronal excitability caused by Fabry Schwann cells. These findings demonstrate that sensory neurons from rats with Fabry disease exhibit hyperactivity caused in part by Schwann cell release of the protein p11.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Nancy M. Dahms
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Elena Isaeva
- Department of Cell Biology, Neurobiology & Anatomy
| | | | - Bin Pan
- Department of Anesthesiology; and
| | | |
Collapse
|
14
|
Ma J, Eglauf J, Grad S, Alini M, Serra T. Engineering Sensory Ganglion Multicellular System to Model Tissue Nerve Ingrowth. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308478. [PMID: 38113315 PMCID: PMC10953573 DOI: 10.1002/advs.202308478] [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: 11/07/2023] [Revised: 12/04/2023] [Indexed: 12/21/2023]
Abstract
Discogenic pain is associated with deep nerve ingrowth in annulus fibrosus tissue (AF) of intervertebral disc (IVD). To model AF nerve ingrowth, primary bovine dorsal root ganglion (DRG) micro-scale tissue units are spatially organised around an AF explant by mild hydrodynamic forces within a collagen matrix. This results in a densely packed multicellular system mimicking the native DRG tissue morphology and a controlled AF-neuron distance. Such a multicellular organisation is essential to evolve populational-level cellular functions and in vivo-like morphologies. Pro-inflammatory cytokine-primed AF demonstrates its neurotrophic and neurotropic effects on nociceptor axons. Both effects are dependent on the AF-neuron distance underpinning the role of recapitulating inter-tissue/organ anatomical proximity when investigating their crosstalk. This is the first in vitro model studying AF nerve ingrowth by engineering mature and large animal tissues in a morphologically and physiologically relevant environment. The new approach can be used to biofabricate multi-tissue/organ models for untangling pathophysiological conditions and develop novel therapies.
Collapse
Affiliation(s)
- Junxuan Ma
- AO Research InstituteClavadelerstrasse 8Davos7270Switzerland
| | - Janick Eglauf
- AO Research InstituteClavadelerstrasse 8Davos7270Switzerland
- ETH ZürichRämistrasse 101Zürich8092Switzerland
| | - Sibylle Grad
- AO Research InstituteClavadelerstrasse 8Davos7270Switzerland
| | - Mauro Alini
- AO Research InstituteClavadelerstrasse 8Davos7270Switzerland
| | - Tiziano Serra
- AO Research InstituteClavadelerstrasse 8Davos7270Switzerland
- Complex Tissue Regeneration DepartmentMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229ETNetherlands
| |
Collapse
|
15
|
Beletskiy A, Zolotar A, Fortygina P, Chesnokova E, Uroshlev L, Balaban P, Kolosov P. Downregulation of Ribosomal Protein Genes Is Revealed in a Model of Rat Hippocampal Neuronal Culture Activation with GABA(A)R/GlyRa2 Antagonist Picrotoxin. Cells 2024; 13:383. [PMID: 38474347 DOI: 10.3390/cells13050383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 02/13/2024] [Accepted: 02/20/2024] [Indexed: 03/14/2024] Open
Abstract
Long-read transcriptome sequencing provides us with a convenient tool for the thorough study of biological processes such as neuronal plasticity. Here, we aimed to perform transcriptional profiling of rat hippocampal primary neuron cultures after stimulation with picrotoxin (PTX) to further understand molecular mechanisms of neuronal activation. To overcome the limitations of short-read RNA-Seq approaches, we performed an Oxford Nanopore Technologies MinION-based long-read sequencing and transcriptome assembly of rat primary hippocampal culture mRNA at three time points after the PTX activation. We used a specific approach to exclude uncapped mRNAs during sample preparation. Overall, we found 23,652 novel transcripts in comparison to reference annotations, out of which ~6000 were entirely novel and mostly transposon-derived loci. Analysis of differentially expressed genes (DEG) showed that 3046 genes were differentially expressed, of which 2037 were upregulated and 1009 were downregulated at 30 min after the PTX application, with only 446 and 13 genes differentially expressed at 1 h and 5 h time points, respectively. Most notably, multiple genes encoding ribosomal proteins, with a high basal expression level, were downregulated after 30 min incubation with PTX; we suggest that this indicates redistribution of transcriptional resources towards activity-induced genes. Novel loci and isoforms observed in this study may help us further understand the functional mRNA repertoire in neuronal plasticity processes. Together with other NGS techniques, differential gene expression analysis of sequencing data obtained using MinION platform might provide a simple method to optimize further study of neuronal plasticity.
Collapse
Affiliation(s)
- Alexander Beletskiy
- Institute of Higher Nervous Activity and Neurophysiology, The Russian Academy of Sciences, 117485 Moscow, Russia
| | - Anastasia Zolotar
- Institute of Higher Nervous Activity and Neurophysiology, The Russian Academy of Sciences, 117485 Moscow, Russia
| | - Polina Fortygina
- Institute of Higher Nervous Activity and Neurophysiology, The Russian Academy of Sciences, 117485 Moscow, Russia
| | - Ekaterina Chesnokova
- Institute of Higher Nervous Activity and Neurophysiology, The Russian Academy of Sciences, 117485 Moscow, Russia
| | - Leonid Uroshlev
- Institute of Higher Nervous Activity and Neurophysiology, The Russian Academy of Sciences, 117485 Moscow, Russia
| | - Pavel Balaban
- Institute of Higher Nervous Activity and Neurophysiology, The Russian Academy of Sciences, 117485 Moscow, Russia
| | - Peter Kolosov
- Institute of Higher Nervous Activity and Neurophysiology, The Russian Academy of Sciences, 117485 Moscow, Russia
- Engelhardt Institute of Molecular Biology, The Russian Academy of Sciences, 119991 Moscow, Russia
| |
Collapse
|
16
|
Bahl E, Chatterjee S, Mukherjee U, Elsadany M, Vanrobaeys Y, Lin LC, McDonough M, Resch J, Giese KP, Abel T, Michaelson JJ. Using deep learning to quantify neuronal activation from single-cell and spatial transcriptomic data. Nat Commun 2024; 15:779. [PMID: 38278804 PMCID: PMC10817898 DOI: 10.1038/s41467-023-44503-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 12/15/2023] [Indexed: 01/28/2024] Open
Abstract
Neuronal activity-dependent transcription directs molecular processes that regulate synaptic plasticity, brain circuit development, behavioral adaptation, and long-term memory. Single cell RNA-sequencing technologies (scRNAseq) are rapidly developing and allow for the interrogation of activity-dependent transcription at cellular resolution. Here, we present NEUROeSTIMator, a deep learning model that integrates transcriptomic signals to estimate neuronal activation in a way that we demonstrate is associated with Patch-seq electrophysiological features and that is robust against differences in species, cell type, and brain region. We demonstrate this method's ability to accurately detect neuronal activity in previously published studies of single cell activity-induced gene expression. Further, we applied our model in a spatial transcriptomic study to identify unique patterns of learning-induced activity across different brain regions in male mice. Altogether, our findings establish NEUROeSTIMator as a powerful and broadly applicable tool for measuring neuronal activation, whether as a critical covariate or a primary readout of interest.
Collapse
Affiliation(s)
- Ethan Bahl
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
- Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, IA, USA
| | - Snehajyoti Chatterjee
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
- Department of Neuroscience & Pharmacology, University of Iowa, Iowa City, IA, USA
| | - Utsav Mukherjee
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
- Department of Neuroscience & Pharmacology, University of Iowa, Iowa City, IA, USA
- Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, IA, USA
| | - Muhammad Elsadany
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
- Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, IA, USA
| | - Yann Vanrobaeys
- Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, IA, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
| | - Li-Chun Lin
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
- Department of Neuroscience & Pharmacology, University of Iowa, Iowa City, IA, USA
| | - Miriam McDonough
- Department of Neuroscience & Pharmacology, University of Iowa, Iowa City, IA, USA
- Interdisciplinary Graduate Program in Molecular Medicine, University of Iowa, Iowa City, IA, USA
| | - Jon Resch
- Department of Neuroscience & Pharmacology, University of Iowa, Iowa City, IA, USA
| | - K Peter Giese
- Department of Basic and Clinical Neuroscience, King's College London, London, UK
| | - Ted Abel
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
- Department of Neuroscience & Pharmacology, University of Iowa, Iowa City, IA, USA
| | - Jacob J Michaelson
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA.
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA.
- Department of Biomedical Engineering, University of Iowa, Iowa City, IA, USA.
- Department of Communication Sciences & Disorders, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
17
|
Dinamarca MC, Colombo L, Brykczynska U, Grimm A, Fruh I, Hossain I, Gabriel D, Eckert A, Müller M, Pecho-Vrieseling E. Transmission-selective muscle pathology induced by the active propagation of mutant huntingtin across the human neuromuscular synapse. Front Mol Neurosci 2024; 16:1287510. [PMID: 38235149 PMCID: PMC10791992 DOI: 10.3389/fnmol.2023.1287510] [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: 09/01/2023] [Accepted: 11/27/2023] [Indexed: 01/19/2024] Open
Abstract
Neuron-to-neuron transmission of aggregation-prone, misfolded proteins may potentially explain the spatiotemporal accumulation of pathological lesions in the brains of patients with neurodegenerative protein-misfolding diseases (PMDs). However, little is known about protein transmission from the central nervous system to the periphery, or how this propagation contributes to PMD pathology. To deepen our understanding of these processes, we established two functional neuromuscular systems derived from human iPSCs. One was suitable for long-term high-throughput live-cell imaging and the other was adapted to a microfluidic system assuring that connectivity between motor neurons and muscle cells was restricted to the neuromuscular junction. We show that the Huntington's disease (HD)-associated mutant HTT exon 1 protein (mHTTEx1) is transmitted from neurons to muscle cells across the human neuromuscular junction. We found that transmission is an active and dynamic process that starts before aggregate formation and is regulated by synaptic activity. We further found that transmitted mHTTEx1 causes HD-relevant pathology at both molecular and functional levels in human muscle cells, even in the presence of the ubiquitous expression of mHTTEx1. In conclusion, we have uncovered a causal link between mHTTEx1 synaptic transmission and HD pathology, highlighting the therapeutic potential of blocking toxic protein transmission in PMDs.
Collapse
Affiliation(s)
- Margarita C. Dinamarca
- Neuronal Development and Degeneration Laboratory, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Laura Colombo
- Neuronal Development and Degeneration Laboratory, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Urszula Brykczynska
- Neuronal Development and Degeneration Laboratory, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Amandine Grimm
- Neurobiology Laboratory for Brain Aging and Mental Health, Transfaculty Research Platform, Molecular and Cognitive Neuroscience, University of Basel, Basel, Switzerland
| | - Isabelle Fruh
- Biomedical Research, Novartis Pharma AG, Novartis Campus, Basel, Switzerland
| | - Imtiaz Hossain
- Biomedical Research, Novartis Pharma AG, Novartis Campus, Basel, Switzerland
| | - Daniela Gabriel
- Biomedical Research, Novartis Pharma AG, Novartis Campus, Basel, Switzerland
| | - Anne Eckert
- Neurobiology Laboratory for Brain Aging and Mental Health, Transfaculty Research Platform, Molecular and Cognitive Neuroscience, University of Basel, Basel, Switzerland
| | - Matthias Müller
- Biomedical Research, Novartis Pharma AG, Novartis Campus, Basel, Switzerland
| | - Eline Pecho-Vrieseling
- Neuronal Development and Degeneration Laboratory, Department of Biomedicine, University of Basel, Basel, Switzerland
| |
Collapse
|
18
|
Rajarathinam T, Thirumalai D, Jayaraman S, Yang S, Ishigami A, Yoon JH, Paik HJ, Lee J, Chang SC. Glutamate oxidase sheets-Prussian blue grafted amperometric biosensor for the real time monitoring of glutamate release from primary cortical neurons. Int J Biol Macromol 2024; 254:127903. [PMID: 37939751 DOI: 10.1016/j.ijbiomac.2023.127903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 10/25/2023] [Accepted: 11/03/2023] [Indexed: 11/10/2023]
Abstract
Glutamate (GLU) is a primary excitatory neurotransmitter, and its dysregulation is associated with several neurodegenerative disorders. A major challenge in GLU estimation is the existence of other biomolecules in the brain that could directly get oxidized at the electrode. Hence, highly selective electroenzymatic biosensors that enable rapid estimation of GLU are needed. Initially, a copolymer, poly(2-dimethylaminoethyl methacrylate- styrene) was synthesized through reversible addition-fragmentation chain transfer polymerization to noncovalently functionalize reduced graphene oxide (rGO), named DS-rGO. Glutamate oxidase macromolecule immobilized DS-rGO formed enzyme nanosheets, which was drop-coated over Prussian blue electrodeposited disposable electrodes to fabricate the GLU biosensor. The interconnectivity between the enzyme nanosheets and the Prussian blue endows the biosensor with enhanced conductivity and electrochemical activity. The biosensor exhibited a linearity: 3.25-250 μM; sensitivity: 3.96 μA mM-1 cm-2, and a limit of detection: 0.96 μM for GLU in the Neurobasal Medium. The biosensor was applied to an in vitro primary rat cortical model to discriminate GLU levels in Neurobasal Medium, before and after KCl mediated depolarization, which provides new insights for elucidating neuronal functioning in the brain.
Collapse
Affiliation(s)
- Thenmozhi Rajarathinam
- Department of Cogno-Mechatronics Engineering, College of Nanoscience and Nanotechnology, Pusan National University, Busan 46241, Republic of Korea
| | - Dinakaran Thirumalai
- BIT Convergence-based Innovative Drug Development Targeting Metainflammation, Pusan National University, Busan 46241, Republic of Korea
| | - Sivaguru Jayaraman
- Department of Cogno-Mechatronics Engineering, College of Nanoscience and Nanotechnology, Pusan National University, Busan 46241, Republic of Korea
| | - Seonguk Yang
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea
| | - Akihito Ishigami
- Molecular Regulation of Aging, Tokyo Metropolitan Institute of Gerontology, Tokyo 173-0015, Japan
| | - Jang-Hee Yoon
- Busan Center, Korea Basic Science Institute, Busan 46241, Republic of Korea
| | - Hyun-Jong Paik
- Department of Polymer Science and Engineering, Pusan National University, Busan 46241, Republic of Korea
| | - Jaewon Lee
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea.
| | - Seung-Cheol Chang
- Department of Cogno-Mechatronics Engineering, College of Nanoscience and Nanotechnology, Pusan National University, Busan 46241, Republic of Korea.
| |
Collapse
|
19
|
Wu D, Huang K, Shi J, Liu S, Wang W, Jiang J, Ren H, Chen T, Ye S, Chen J, Wei W, Li X. Genome-Wide 5-Formylcytosine Redistribution in KCl-Stimulated Mouse Primary Cortical Neurons is Associated with Neuronal Activity. ACS Chem Neurosci 2023; 14:4352-4362. [PMID: 38019771 DOI: 10.1021/acschemneuro.3c00554] [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: 12/01/2023] Open
Abstract
An abundant accumulation of DNA demethylation intermediates has been identified in mammalian neurons. While the roles of 5-methylcytosine (5mC) and 5-hydroxymethylcytosine (5hmC) in neuronal function have been extensively studied, little is known about 5-formylcytosine (5fC) in neurons. Therefore, this study was to investigate the genome-wide distribution and potential functions of 5fC in neurons. In an in vitro culture model of mouse primary cortical neurons, we observed a dynamic increase in the total 5fC level in the neuronal genome after potassium chloride (KCl) stimulation. Subsequently, we employed chemical-labeling-enabled C-to-T conversion sequencing (CLEVER-seq) to examine the 5fC distribution at a single-base resolution. Bioinformatic analysis revealed that 5fC was enriched in promoter regions, and gene ontology (GO) analysis indicated that the differential formylation positions (DFP) were correlated with neuronal activities. Additionally, integration with previously published nascent RNA-seq data revealed a positive correlation between gene formylation and mRNA expression levels. As well, 6 neuro-activity-related genes with a positive correlation were validated. Furthermore, we observed higher chromatin accessibility and RNA pol II binding signals near the 5fC sites through multiomics analysis. Motif analysis identified potential reader proteins for 5fC. In conclusion, our work provides a valuable resource for studying the dynamic changes and functional roles of 5fC in activated mammalian neurons.
Collapse
Affiliation(s)
- Du Wu
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, 430071 Wuhan, China
- Brain Research Center, Zhongnan Hospital of Wuhan University, 430071 Wuhan, China
| | - Kaixin Huang
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, 430071 Wuhan, China
- Brain Research Center, Zhongnan Hospital of Wuhan University, 430071 Wuhan, China
| | - Jichun Shi
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, 430071 Wuhan, China
- Brain Research Center, Zhongnan Hospital of Wuhan University, 430071 Wuhan, China
| | - Sha Liu
- Department of General Practice, Zhongnan Hospital of Wuhan University, 430071 Wuhan, China
| | - Wenjing Wang
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, 430071 Wuhan, China
| | - Jiazhi Jiang
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, 430071 Wuhan, China
| | - Haobin Ren
- Cognitive Neuroepigenetics Laboratory, Queensland Brain Institute, The University of Queensland, Brisbane 4702, Australia
| | - Tongyu Chen
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, 430071 Wuhan, China
| | - Shengda Ye
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, 430071 Wuhan, China
| | - Jincao Chen
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, 430071 Wuhan, China
| | - Wei Wei
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, 430071 Wuhan, China
- Brain Research Center, Zhongnan Hospital of Wuhan University, 430071 Wuhan, China
| | - Xiang Li
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, 430071 Wuhan, China
- Brain Research Center, Zhongnan Hospital of Wuhan University, 430071 Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Wuhan University, 430071 Wuhan, China
- Medical Research Institute, Wuhan University, 430071 Wuhan, China
- Sino-Italian Ascula Brain Science Joint Laboratory, Zhongnan Hospital of Wuhan University, 430071 Wuhan, China
| |
Collapse
|
20
|
Zou Z, Wei J, Chen Y, Kang Y, Shi H, Yang F, Shi Z, Chen S, Zhou Y, Sepich-Poore C, Zhuang X, Zhou X, Jiang H, Wen Z, Jin P, Luo C, He C. FMRP phosphorylation modulates neuronal translation through YTHDF1. Mol Cell 2023; 83:4304-4317.e8. [PMID: 37949069 PMCID: PMC10872974 DOI: 10.1016/j.molcel.2023.10.028] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 09/12/2023] [Accepted: 10/19/2023] [Indexed: 11/12/2023]
Abstract
RNA-binding proteins (RBPs) control messenger RNA fate in neurons. Here, we report a mechanism that the stimuli-induced neuronal translation is mediated by phosphorylation of a YTHDF1-binding protein FMRP. Mechanistically, YTHDF1 can condense with ribosomal proteins to promote the translation of its mRNA targets. FMRP regulates this process by sequestering YTHDF1 away from the ribosome; upon neuronal stimulation, FMRP becomes phosphorylated and releases YTHDF1 for translation upregulation. We show that a new small molecule inhibitor of YTHDF1 can reverse fragile X syndrome (FXS) developmental defects associated with FMRP deficiency in an organoid model. Our study thus reveals that FMRP and its phosphorylation are important regulators of activity-dependent translation during neuronal development and stimulation and identifies YTHDF1 as a potential therapeutic target for FXS in which developmental defects caused by FMRP depletion could be reversed through YTHDF1 inhibition.
Collapse
Affiliation(s)
- Zhongyu Zou
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA; Howard Hughes Medical Institute, The University of Chicago, Chicago, IL 60637, USA
| | - Jiangbo Wei
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA; Howard Hughes Medical Institute, The University of Chicago, Chicago, IL 60637, USA
| | - Yantao Chen
- The Center for Chemical Biology, Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yunhee Kang
- Department of Psychiatry and Behavioral Sciences, Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Hailing Shi
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA; Howard Hughes Medical Institute, The University of Chicago, Chicago, IL 60637, USA
| | - Fan Yang
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA; Howard Hughes Medical Institute, The University of Chicago, Chicago, IL 60637, USA
| | - Zhuoyue Shi
- Department of Human Genetics, The University of Chicago, Chicago, IL 60637, USA
| | - Shijie Chen
- The Center for Chemical Biology, Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310024, China
| | - Ying Zhou
- Department of Psychiatry and Behavioral Sciences, Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Caraline Sepich-Poore
- Howard Hughes Medical Institute, The University of Chicago, Chicago, IL 60637, USA; Medical Scientist Training Program, Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL 60637, USA
| | - Xiaoxi Zhuang
- Department of Neurobiology, The University of Chicago, Chicago, IL 60637, USA
| | - Xiaoming Zhou
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Hualiang Jiang
- The Center for Chemical Biology, Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Zhexing Wen
- Department of Psychiatry and Behavioral Sciences, Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Peng Jin
- Department of Psychiatry and Behavioral Sciences, Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA.
| | - Cheng Luo
- The Center for Chemical Biology, Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310024, China; School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Chuan He
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA; Howard Hughes Medical Institute, The University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
21
|
Gourisankar S, Wenderski W, Paulo JA, Kim SH, Roepke K, Ellis C, Gygi SP, Crabtree GR. Synaptic Activity Causes Minute-scale Changes in BAF Complex Composition and Function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.13.562244. [PMID: 37873481 PMCID: PMC10592824 DOI: 10.1101/2023.10.13.562244] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Genes encoding subunits of the SWI/SNF or BAF ATP-dependent chromatin remodeling complex are among the most enriched for deleterious de novo mutations in intellectual disabilities and autism spectrum disorder, but the causative molecular pathways are not fully known 1,2 . Synaptic activity in neurons is critical for learning and memory and proper neural development 3 . Neural activity prompts calcium influx and transcription within minutes, facilitated in the nucleus by various transcription factors (TFs) and chromatin modifiers 4 . While BAF is required for activity-dependent developmental processes such as dendritic outgrowth 5-7 , the immediate molecular consequences of neural activity on BAF complexes and their functions are unknown. Here we mapped minute-scale biochemical consequences of neural activity, modeled by membrane depolarization of embryonic mouse primary cortical neurons, on BAF complexes. We used acute chemical perturbations of BAF ATPase activity and kinase signaling to define the activity-dependent effects on BAF complexes and activity-dependent BAF functions. Our studies found that BAF complexes change in subunit composition and are selectively phosphorylated within 10 minutes of depolarization. Increased levels of the core PBAF subunit Baf200/ Arid2 , uniquely containing an RFX-like DNA-binding domain, are concurrent with ATPase-dependent opening of chromatin at RFX/X-box motifs. Changes in BAF composition and phosphorylation lead to the regulation of chromatin accessibility for critical neurogenesis TFs. These biochemical effects are a convergent phenomenon downstream of multiple growth factor signaling pathways in mouse neurons and fibroblasts suggesting that BAF integrates signaling information from the membrane. In support of such a membrane-to-nucleus signaling cascade, we also identified a BAF-interacting kinase, Dclk2, whose inhibition attenuates BAF phosphorylation selectively. Our findings support a direct role of BAF complexes in responding to synaptic activity to regulate TF binding and transcription.
Collapse
|
22
|
Zhang Y, Riexinger J, Yang X, Mikhailova E, Jin Y, Zhou L, Bayley H. A microscale soft ionic power source modulates neuronal network activity. Nature 2023; 620:1001-1006. [PMID: 37648756 PMCID: PMC10468398 DOI: 10.1038/s41586-023-06295-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 06/07/2023] [Indexed: 09/01/2023]
Abstract
Bio-integrated devices need power sources to operate1,2. Despite widely used technologies that can provide power to large-scale targets, such as wired energy supplies from batteries or wireless energy transduction3, a need to efficiently stimulate cells and tissues on the microscale is still pressing. The ideal miniaturized power source should be biocompatible, mechanically flexible and able to generate an ionic current for biological stimulation, instead of using electron flow as in conventional electronic devices4-6. One approach is to use soft power sources inspired by the electrical eel7,8; however, power sources that combine the required capabilities have not yet been produced, because it is challenging to obtain miniaturized units that both conserve contained energy before usage and are easily triggered to produce an energy output. Here we develop a miniaturized soft power source by depositing lipid-supported networks of nanolitre hydrogel droplets that use internal ion gradients to generate energy. Compared to the original eel-inspired design7, our approach can shrink the volume of a power unit by more than 105-fold and it can store energy for longer than 24 h, enabling operation on-demand with a 680-fold greater power density of about 1,300 W m-3. Our droplet device can serve as a biocompatible and biological ionic current source to modulate neuronal network activity in three-dimensional neural microtissues and in ex vivo mouse brain slices. Ultimately, our soft microscale ionotronic device might be integrated into living organisms.
Collapse
Affiliation(s)
- Yujia Zhang
- Department of Chemistry, University of Oxford, Oxford, UK.
| | | | - Xingyun Yang
- Department of Chemistry, University of Oxford, Oxford, UK
| | | | - Yongcheng Jin
- Department of Chemistry, University of Oxford, Oxford, UK
| | - Linna Zhou
- Department of Chemistry, University of Oxford, Oxford, UK.
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| | - Hagan Bayley
- Department of Chemistry, University of Oxford, Oxford, UK.
| |
Collapse
|
23
|
Ross MM, Aizenman E. GluA1-Shank3 interaction decreases in response to chronic neuronal depolarization. Neurosci Lett 2023; 809:137305. [PMID: 37210067 PMCID: PMC10330850 DOI: 10.1016/j.neulet.2023.137305] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/13/2023] [Accepted: 05/15/2023] [Indexed: 05/22/2023]
Abstract
Interactions between AMPA receptors and synaptic scaffolding proteins are key regulators of synaptic receptor density and, thereby, synapse strength. Shank3 is one such scaffolding protein with high clinical relevance, as genetic variants and deletions of this protein have been linked to autism spectrum disorder. Shank3 acts as a master regulator of the postsynaptic density of glutamatergic synapses, interacting with ionotropic and metabotropic glutamate receptors and cytoskeletal elements to modulate synaptic structure. Notably, Shank3 has been shown to interact directly with the AMPAR subunit GluA1, and Shank3 knockout animals show deficits in AMPAR-mediated synaptic transmission. In this study, we sought to characterize the stability of GluA1-Shank3 interaction in response to chronic stimuli using a highly sensitive and specific proximity ligation assay. We found that GluA1-Shank3 interactions decrease in response to prolonged neuronal depolarization induced by elevated extracellular potassium, and that this reduced interaction is blocked by NMDA receptor antagonism. These results firmly establish the close interaction of GluA1 and Shank3 in cortical neurons in vitro, and that this select interaction is subject to modulation by depolarization.
Collapse
Affiliation(s)
- Madeline M Ross
- Department of Neurobiology and Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Elias Aizenman
- Department of Neurobiology and Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA.
| |
Collapse
|
24
|
Orlov NA, Kryukova EV, Efremenko AV, Yakimov SA, Toporova VA, Kirpichnikov MP, Nekrasova OV, Feofanov AV. Interactions of the Kv1.1 Channel with Peptide Pore Blockers: A Fluorescent Analysis on Mammalian Cells. MEMBRANES 2023; 13:645. [PMID: 37505011 PMCID: PMC10383195 DOI: 10.3390/membranes13070645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 06/23/2023] [Accepted: 06/27/2023] [Indexed: 07/29/2023]
Abstract
The voltage-gated potassium channel Kv1.1, which is abundant in the CNS and peripheral nervous system, controls neuronal excitability and neuromuscular transmission and mediates a number of physiological functions in non-excitable cells. The development of some diseases is accompanied by changes in the expression level and/or activity of the channels in particular types of cells. To meet the requirements of studies related to the expression and localization of the Kv1.1 channels, we report on the subnanomolar affinity of hongotoxin 1 N-terminally labeled with Atto 488 fluorophore (A-HgTx) for the Kv1.1 channel and its applicability for fluorescent imaging of the channel in living cells. Taking into consideration the pharmacological potential of the Kv1.1 channel, a fluorescence-based analytical system was developed for the study of peptide ligands that block the ion conductivity of Kv1.1 and are potentially able to correct abnormal activity of the channel. The system is based on analysis of the competitive binding of the studied compounds and A-HgTx to the mKate2-tagged human Kv1.1 (S369T) channel, expressed in the plasma membrane of Neuro2a cells. The system was validated by measuring the affinities of the known Kv1.1-channel peptide blockers, such as agitoxin 2, kaliotoxin 1, hongotoxin 1, and margatoxin. Peptide pore blocker Ce1, from the venom of the scorpion Centruroides elegans, was shown to possess a nanomolar affinity for the Kv1.1 channel. It is reported that interactions of the Kv1.1 channel with the studied peptide blockers are not affected by the transition of the channel from the closed to open state. The conclusion is made that the structural rearrangements accompanying the channel transition into the open state do not change the conformation of the P-loop (including the selectivity filter) involved in the formation of the binding site of the peptide pore blockers.
Collapse
Affiliation(s)
- Nikita A Orlov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
- Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Elena V Kryukova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Anastasia V Efremenko
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Sergey A Yakimov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Victoria A Toporova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Mikhail P Kirpichnikov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
- Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Oksana V Nekrasova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Alexey V Feofanov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
- Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
| |
Collapse
|
25
|
Ghaffari LT, Trotti D, Haeusler AR. Differential response of C9orf72 transcripts following neuronal depolarization. iScience 2023; 26:106959. [PMID: 37332610 PMCID: PMC10272498 DOI: 10.1016/j.isci.2023.106959] [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: 01/24/2023] [Revised: 04/19/2023] [Accepted: 05/22/2023] [Indexed: 06/20/2023] Open
Abstract
The (G4C2)n nucleotide repeat expansion (NRE) mutation in C9orf72 is the most common genetic cause of ALS and FTD. The biological functions of C9orf72 are becoming understood, but it is unclear if this gene is regulated in a neural-specific manner. Neuronal activity is a crucial modifier of biological processes in health and neurodegenerative disease contexts. Here, we show that prolonged membrane depolarization in healthy human iPSC-cortical neurons leads to a significant downregulation of a transcript variant 3 (V3) of C9orf72, with a concomitant increase in variant 2 (V2), which leads to total C9orf72 RNA transcript levels remaining unchanged. However, the same response is not observed in cortical neurons derived from patients with the C9-NRE mutation. These findings reveal the impact of depolarization on C9orf72 transcripts, and how this response diverges in C9-NRE-carriers, which may have important implications in the underlying unique clinical associations of C9-NRE transcripts and disease pathogenesis.
Collapse
Affiliation(s)
- Layla T. Ghaffari
- Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Davide Trotti
- Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Aaron R. Haeusler
- Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
26
|
Fernandes AR, Martins JP, Gomes ER, Mendes CS, Teodoro RO. Drosophila motor neuron boutons remodel through membrane blebbing coupled with muscle contraction. Nat Commun 2023; 14:3352. [PMID: 37291089 PMCID: PMC10250368 DOI: 10.1038/s41467-023-38421-9] [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: 03/30/2021] [Accepted: 04/26/2023] [Indexed: 06/10/2023] Open
Abstract
Wired neurons form new presynaptic boutons in response to increased synaptic activity, however the mechanism(s) by which this occurs remains uncertain. Drosophila motor neurons (MNs) have clearly discernible boutons that display robust structural plasticity, being therefore an ideal system in which to study activity-dependent bouton genesis. Here, we show that in response to depolarization and in resting conditions, MNs form new boutons by membrane blebbing, a pressure-driven mechanism that occurs in 3-D cell migration, but to our knowledge not previously described to occur in neurons. Accordingly, F-actin is decreased in boutons during outgrowth, and non-muscle myosin-II is dynamically recruited to newly formed boutons. Furthermore, muscle contraction plays a mechanical role, which we hypothesize promotes bouton addition by increasing MN confinement. Overall, we identified a mechanism by which established circuits form new boutons allowing their structural expansion and plasticity, using trans-synaptic physical forces as the main driving force.
Collapse
Affiliation(s)
- Andreia R Fernandes
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Lisboa, Portugal
| | - João P Martins
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028, Lisboa, Portugal
| | - Edgar R Gomes
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028, Lisboa, Portugal
| | - César S Mendes
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Lisboa, Portugal
| | - Rita O Teodoro
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Lisboa, Portugal.
| |
Collapse
|
27
|
Girardi G, Zumpano D, Goshi N, Raybould H, Seker E. Cultured Vagal Afferent Neurons as Sensors for Intestinal Effector Molecules. BIOSENSORS 2023; 13:601. [PMID: 37366967 DOI: 10.3390/bios13060601] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 05/23/2023] [Accepted: 05/24/2023] [Indexed: 06/28/2023]
Abstract
The gut-brain axis embodies the bi-directional communication between the gastrointestinal tract and the central nervous system (CNS), where vagal afferent neurons (VANs) serve as sensors for a variety of gut-derived signals. The gut is colonized by a large and diverse population of microorganisms that communicate via small (effector) molecules, which also act on the VAN terminals situated in the gut viscera and consequently influence many CNS processes. However, the convoluted in vivo environment makes it difficult to study the causative impact of the effector molecules on VAN activation or desensitization. Here, we report on a VAN culture and its proof-of-principle demonstration as a cell-based sensor to monitor the influence of gastrointestinal effector molecules on neuronal behavior. We initially compared the effect of surface coatings (poly-L-lysine vs. Matrigel) and culture media composition (serum vs. growth factor supplement) on neurite growth as a surrogate of VAN regeneration following tissue harvesting, where the Matrigel coating, but not the media composition, played a significant role in the increased neurite growth. We then used both live-cell calcium imaging and extracellular electrophysiological recordings to show that the VANs responded to classical effector molecules of endogenous and exogenous origin (cholecystokinin serotonin and capsaicin) in a complex fashion. We expect this study to enable platforms for screening various effector molecules and their influence on VAN activity, assessed by their information-rich electrophysiological fingerprints.
Collapse
Affiliation(s)
- Gregory Girardi
- Department of Biomedical Engineering, University of California-Davis, Davis, CA 95616, USA
| | - Danielle Zumpano
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California-Davis, Davis, CA 95616, USA
| | - Noah Goshi
- Department of Biomedical Engineering, University of California-Davis, Davis, CA 95616, USA
| | - Helen Raybould
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California-Davis, Davis, CA 95616, USA
| | - Erkin Seker
- Department of Electrical and Computer Engineering, University of California-Davis, Davis, CA 95616, USA
| |
Collapse
|
28
|
Jeong S, Kang HW, Kim SH, Hong GS, Nam MH, Seong J, Yoon ES, Cho IJ, Chung S, Bang S, Kim HN, Choi N. Integration of reconfigurable microchannels into aligned three-dimensional neural networks for spatially controllable neuromodulation. SCIENCE ADVANCES 2023; 9:eadf0925. [PMID: 36897938 PMCID: PMC10005277 DOI: 10.1126/sciadv.adf0925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 02/08/2023] [Indexed: 06/18/2023]
Abstract
Anisotropically organized neural networks are indispensable routes for functional connectivity in the brain, which remains largely unknown. While prevailing animal models require additional preparation and stimulation-applying devices and have exhibited limited capabilities regarding localized stimulation, no in vitro platform exists that permits spatiotemporal control of chemo-stimulation in anisotropic three-dimensional (3D) neural networks. We present the integration of microchannels seamlessly into a fibril-aligned 3D scaffold by adapting a single fabrication principle. We investigated the underlying physics of elastic microchannels' ridges and interfacial sol-gel transition of collagen under compression to determine a critical window of geometry and strain. We demonstrated the spatiotemporally resolved neuromodulation in an aligned 3D neural network by local deliveries of KCl and Ca2+ signal inhibitors, such as tetrodotoxin, nifedipine, and mibefradil, and also visualized Ca2+ signal propagation with a speed of ~3.7 μm/s. We anticipate that our technology will pave the way to elucidate functional connectivity and neurological diseases associated with transsynaptic propagation.
Collapse
Affiliation(s)
- Sohyeon Jeong
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul 02792, Korea
- MEPSGEN Co. Ltd., Seoul 05836, Korea
| | - Hyun Wook Kang
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
- School of Mechanical Engineering, Korea University, Seoul 02841, Korea
| | - So Hyun Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
- SK Biopharmaceuticals Co. Ltd., Seongnam 13494, Korea
| | - Gyu-Sang Hong
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
| | - Min-Ho Nam
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
| | - Jihye Seong
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul 02792, Korea
- Department of Life Sciences, Korea University, Seoul 02841, Korea
- KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul 02453, Korea
| | - Eui-Sung Yoon
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
- Division of Nano and Information Technology, KIST School, University of Science and Technology (UST), Seoul 02792, Korea
| | - Il-Joo Cho
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Korea
- Department of Anatomy, College of Medicine, Korea University, Seoul 02841, Korea
| | - Seok Chung
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
- School of Mechanical Engineering, Korea University, Seoul 02841, Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Korea
| | - Seokyoung Bang
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
- Department of Medical Biotechnology, Dongguk University, Goyang 10326, Korea
| | - Hong Nam Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul 02792, Korea
- School of Mechanical Engineering, Yonsei University, Seoul 03722, Korea
- Yonsei-KIST Convergence Research Institute, Yonsei University, Seoul 03722, Korea
| | - Nakwon Choi
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Korea
| |
Collapse
|
29
|
McLeod F, Dimtsi A, Marshall AC, Lewis-Smith D, Thomas R, Clowry GJ, Trevelyan AJ. Altered synaptic connectivity in an in vitro human model of STXBP1 encephalopathy. Brain 2023; 146:850-857. [PMID: 36315647 PMCID: PMC9976961 DOI: 10.1093/brain/awac396] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 08/05/2022] [Accepted: 10/01/2022] [Indexed: 01/14/2023] Open
Abstract
Early infantile developmental and epileptic encephalopathies are devastating conditions, generally of genetic origin, but the pathological mechanisms often remain obscure. A major obstacle in this field of research is the difficulty of studying cortical brain development in humans, at the relevant time period in utero. To address this, we established an in vitro assay to study the impact of gene variants on the developing human brain by using living organotypic cultures of the human subplate and neighbouring cortical regions, prepared from ethically sourced, 14-17 post-conception week brain tissue (www.hdbr.org). We were able to maintain cultures for several months, during which time the gross anatomical structures of the cortical plate, subplate and marginal zone persisted, while neurons continued to develop morphologically and form new synaptic networks. This preparation thus permits the study of genetic manipulations and their downstream effects on an intact developing human cortical network. We focused on STXBP1 haploinsufficiency, which is among the most common genetic causes of developmental and epileptic encephalopathy. This was induced using shRNA interference, leading to impaired synaptic function and a reduced density of glutamatergic synapses. We thereby provide a critical proof-of-principle for how to study the impact of any gene of interest on the development of the human cortex.
Collapse
Affiliation(s)
- Faye McLeod
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Anna Dimtsi
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Amy C Marshall
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - David Lewis-Smith
- Newcastle University Translational and Clinical Research Institute, Newcastle upon Tyne NE2 4HH, UK
- Department of Clinical Neurosciences, Royal Victoria Infirmary, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne NE1 4LP, UK
| | - Rhys Thomas
- Newcastle University Translational and Clinical Research Institute, Newcastle upon Tyne NE2 4HH, UK
- Department of Clinical Neurosciences, Royal Victoria Infirmary, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne NE1 4LP, UK
| | - Gavin J Clowry
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Andrew J Trevelyan
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| |
Collapse
|
30
|
Nikolaeva-Koleva M, Espinosa A, Vergassola M, Polenzani L, Mangano G, Ragni L, Zucchi S, Ferrer-Montiel A, Devesa I. Benzydamine plays a role in limiting inflammatory pain induced by neuronal sensitization. Mol Pain 2023; 19:17448069231204191. [PMID: 37710969 PMCID: PMC10583526 DOI: 10.1177/17448069231204191] [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: 06/02/2023] [Revised: 08/28/2023] [Accepted: 09/12/2023] [Indexed: 09/16/2023] Open
Abstract
Benzydamine is an active pharmaceutical compound used in the oral care pharmaceutical preparation as NSAID. Beside from its anti-inflammatory action, benzydamine local application effectively reliefs pain showing analgesic and anaesthetic properties. Benzydamine mechanism of action has been characterized on inflammatory cell types and mediators highlighting its capacity to inhibit pro-inflammatory mediators' synthesis and release. On the other hand, the role of benzydamine as neuronal excitability modulator has not yet fully explored. Thus, we studied benzydamine's effect over primary cultured DRG nociceptors excitability and after acute and chronic inflammatory sensitization, as a model to evaluate relative nociceptive response. Benzydamine demonstrated to effectively inhibit neuronal basal excitability reducing its firing frequency and increasing rheobase and afterhyperpolarization amplitude. Its effect was time and dose-dependent. At higher doses, benzydamine induced changes in action potential wavelength, decreasing its height and slightly increasing its duration. Moreover, the compound reduced neuronal acute and chronic inflammatory sensitization. It inhibited neuronal excitability mediated either by an inflammatory cocktail, acidic pH or high external KCl. Notably, higher potency was evidenced under inflammatory sensitized conditions. This effect could be explained either by modulation of inflammatory and/or neuronal sensitizing signalling cascades or by direct modulation of proalgesic and action potential firing initiating ion channels. Apparently, the compound inhibited Nav1.8 channel but had no effect over Kv7.2, Kv7.3, TRPV1 and TRPA1. In conclusion, the obtained results strengthen the analgesic and anti-inflammatory effect of benzydamine, highlighting its mode of action on local pain and inflammatory signalling.
Collapse
Affiliation(s)
| | | | - Matteo Vergassola
- Angelini Pharma S.p.A, Global R&D PLCM Preclinical Development, Ancona, Italy
| | - Lorenzo Polenzani
- Independent Consultant in Life Sciences & Healthcare, Enischio, Grottaferrata, Italy
| | | | - Lorella Ragni
- Angelini Pharma S.p.A, Global R&D PLCM Preclinical Development, Ancona, Italy
| | - Sara Zucchi
- Angelini Pharma S.p.A, Global R&D PLCM Preclinical Development, Ancona, Italy
| | - Antonio Ferrer-Montiel
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universitas Miguel Hernández, Elche, Spain
| | | |
Collapse
|
31
|
Activity-dependent translation dynamically alters the proteome of the perisynaptic astrocyte process. Cell Rep 2022; 41:111474. [PMID: 36261025 PMCID: PMC9624251 DOI: 10.1016/j.celrep.2022.111474] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 06/16/2022] [Accepted: 09/19/2022] [Indexed: 01/07/2023] Open
Abstract
Within eukaryotic cells, translation is regulated independent of transcription, enabling nuanced, localized, and rapid responses to stimuli. Neurons respond transcriptionally and translationally to synaptic activity. Although transcriptional responses are documented in astrocytes, here we test whether astrocytes have programmed translational responses. We show that seizure activity rapidly changes the transcripts on astrocyte ribosomes, some predicted to be downstream of BDNF signaling. In acute slices, we quantify the extent to which cues of neuronal activity activate translation in astrocytes and show that this translational response requires the presence of neurons, indicating that the response is non-cell autonomous. We also show that this induction of new translation extends into the periphery of astrocytes. Finally, synaptic proteomics show that new translation is required for changes that occur in perisynaptic astrocyte protein composition after fear conditioning. Regulation of translation in astrocytes by neuronal activity suggests an additional mechanism by which astrocytes may dynamically modulate nervous system functioning.
Collapse
|
32
|
A Promising Method for the Determination of Cell Viability: The Membrane Potential Cell Viability Assay. Cells 2022; 11:cells11152314. [PMID: 35954159 PMCID: PMC9367465 DOI: 10.3390/cells11152314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 07/19/2022] [Accepted: 07/26/2022] [Indexed: 02/04/2023] Open
Abstract
Determining the viability of cells is fraught with many uncertainties. It is often difficult to determine whether a cell is still alive, approaching the point of no return, or dead. Today, there are many methods for determining cell viability. Most rely on an indirect determination of cell death (metabolism, molecular transport, and leakage, to name a few). In contrast, we have developed a promising novel method for a “direct” determination of cell viability. The potential method assesses cell membrane integrity (which is essential for all viable cells) by measuring the electrical potential of the cell membrane. To test the assay, we chose two different cell types, blood macrophages (TLT) and breast cancer epithelial cells (MCF 7). We exposed them to seven different toxic scenarios (arsenic (V), UV light, hydrogen peroxide, nutrient starvation, Tetrabromobisphenol A, fatty acids, and 5-fluorouracil) to induce different cell death pathways. Under controlled test conditions, the assay showed good accuracy when comparing the toxicity assessment with well-established methods. Moreover, the method showed compatibility with live cell imaging. Although we know that further studies are needed to confirm the performance of the assay in other situations, the results obtained are promising for their wider application in the future.
Collapse
|
33
|
Rienecker KDA, Poston RG, Segales JS, Finholm IW, Sono MH, Munteanu SJ, Ghaninejad-Esfahani M, Rejepova A, Tejeda-Garibay S, Wickman K, Marron Fernandez de Velasco E, Thayer SA, Saha RN. Mild membrane depolarization in neurons induces immediate early gene transcription and acutely subdues responses to successive stimulus. J Biol Chem 2022; 298:102278. [PMID: 35863435 PMCID: PMC9396413 DOI: 10.1016/j.jbc.2022.102278] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 07/12/2022] [Accepted: 07/13/2022] [Indexed: 11/22/2022] Open
Abstract
Immediate early genes (IEGs) are transcribed in response to neuronal activity from sensory stimulation during multiple adaptive processes in the brain. The transcriptional profile of IEGs is indicative of the duration of neuronal activity, but its sensitivity to the strength of depolarization remains unknown. Also unknown is whether activity history of graded potential changes influence future neuronal activity. In this work with dissociated rat cortical neurons, we found that mild depolarization—mediated by elevated extracellular potassium (K+)—induces a wide array of rapid IEGs and transiently depresses transcriptional and signaling responses to a successive stimulus. This latter effect was independent of de novo transcription, translation, and signaling via calcineurin or mitogen-activated protein kinase. Furthermore, as measured by multiple electrode arrays and calcium imaging, mild depolarization acutely subdues subsequent spontaneous and bicuculline-evoked activity via calcium- and N-methyl-d-aspartate receptor–dependent mechanisms. Collectively, this work suggests that a recent history of graded potential changes acutely depress neuronal intrinsic properties and subsequent responses. Such effects may have several potential downstream implications, including reducing signal-to-noise ratio during synaptic plasticity processes.
Collapse
Affiliation(s)
- Kira D A Rienecker
- Molecular and Cell Biology Department, School of Natural Sciences, University of California, Merced, 5200 North Lake Road, Merced, California, USA, 95343
| | - Robert G Poston
- Molecular and Cell Biology Department, School of Natural Sciences, University of California, Merced, 5200 North Lake Road, Merced, California, USA, 95343
| | - Joshua S Segales
- Molecular and Cell Biology Department, School of Natural Sciences, University of California, Merced, 5200 North Lake Road, Merced, California, USA, 95343
| | - Isabelle W Finholm
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN, 55455, USA
| | - Morgan H Sono
- Molecular and Cell Biology Department, School of Natural Sciences, University of California, Merced, 5200 North Lake Road, Merced, California, USA, 95343
| | - Sorina J Munteanu
- Molecular and Cell Biology Department, School of Natural Sciences, University of California, Merced, 5200 North Lake Road, Merced, California, USA, 95343
| | - Mina Ghaninejad-Esfahani
- Molecular and Cell Biology Department, School of Natural Sciences, University of California, Merced, 5200 North Lake Road, Merced, California, USA, 95343
| | - Ayna Rejepova
- Molecular and Cell Biology Department, School of Natural Sciences, University of California, Merced, 5200 North Lake Road, Merced, California, USA, 95343
| | - Susana Tejeda-Garibay
- Molecular and Cell Biology Department, School of Natural Sciences, University of California, Merced, 5200 North Lake Road, Merced, California, USA, 95343
| | - Kevin Wickman
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN, 55455, USA
| | | | - Stanley A Thayer
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN, 55455, USA
| | - Ramendra N Saha
- Molecular and Cell Biology Department, School of Natural Sciences, University of California, Merced, 5200 North Lake Road, Merced, California, USA, 95343.
| |
Collapse
|
34
|
Lim JS, Kim HJ, Park I, Woo S, Kim JH, Park JW. Force Mapping Reveals the Spatial Distribution of Individual Proteins in a Neuron. NANO LETTERS 2022; 22:3865-3871. [PMID: 35549313 DOI: 10.1021/acs.nanolett.1c04395] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Conventional methods for studying the spatial distribution and expression level of proteins within neurons have primarily relied on immunolabeling and/or signal amplification. Here, we present an atomic force microscopy (AFM)-based nanoscale force mapping method, where Anti-LIMK1-tethered AFM probes were used to visualize individual LIMK1 proteins in cultured neurons directly through force measurements. We observed that the number density of LIMK1 decreased in neuronal somas after the cells were depolarized. We also elucidated the spatial distribution of LIMK1 in single spine areas and found that the protein predominantly locates at heads of spines rather than dendritic shafts. The study demonstrates that our method enables unveiling of the abundance and spatial distribution of a protein of interest in neurons without signal amplification or labeling. We expected that this approach should facilitate the studies of protein expression phenomena in depth in a wide range of biological systems.
Collapse
Affiliation(s)
- Ji-Seon Lim
- Department of Chemistry, Pohang University of Science and Technology, 77 Cheongam-Ro, Nam-Gu, Pohang 37673, Republic of Korea
| | - Hyun Jin Kim
- Department of Life Sciences, Pohang University of Science and Technology, 77 Cheongam-Ro, Nam-Gu, Pohang 37673, Republic of Korea
| | - Ikbum Park
- Analysis and Assessment Research Center, Research Institute of Industrial Science and Technology, 67 Cheongam-Ro, Nam-Gu, Pohang 37673, Republic of Korea
| | - Sungwook Woo
- Department of Chemistry, Pohang University of Science and Technology, 77 Cheongam-Ro, Nam-Gu, Pohang 37673, Republic of Korea
| | - Joung-Hun Kim
- Department of Life Sciences, Pohang University of Science and Technology, 77 Cheongam-Ro, Nam-Gu, Pohang 37673, Republic of Korea
- Institute of Convergence Science, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Joon Won Park
- Department of Chemistry, Pohang University of Science and Technology, 77 Cheongam-Ro, Nam-Gu, Pohang 37673, Republic of Korea
- Institute of Convergence Science, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| |
Collapse
|
35
|
Zhang X, Lee H, Zhang Y, Walmsley TS, Li D, Levine E, Xu YQ. Probing Light-Stimulated Activities in the Retina via Transparent Graphene Electrodes. ACS APPLIED BIO MATERIALS 2022; 5:305-312. [PMID: 35034456 PMCID: PMC10505038 DOI: 10.1021/acsabm.1c01091] [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: 11/28/2022]
Abstract
Graphene has triggered tremendous research due to its superior properties. In particular, the intrinsic high light transmission illustrates the unique advantage in neural biosensing. Here, we combine perforated flexible graphene electrodes with microfluidic platforms to explore real-time extracellular electrical activities of retinal ganglion cells (RGCs). Under light stimulation, the transparent graphene electrodes have demonstrated the capability of recording the electrical activities of stimulated RGCs in direct contact. Different types of RGCs have shown three distinct light induced patterns, ON, OFF, and ON-OFF, which are primarily operated by cone photoreceptors. Moreover, the observed spiking waveforms can be divided into two groups: the biphasic waveform usually occurs at contacts with soma, while the triphasic waveform is likely related to the axon. Under high K+ stimulation, the graphene electrodes exhibit higher electrical sensitivity than gold counterparts with an average 2.5-fold enhancement in spiking amplitude. Furthermore, a strong response has been observed with the firing rate first increasing and then ceasing, which could be due to the potassium-induced neural depolarization. These results show that graphene electrodes can be a promising candidate in the electrophysiology studies of retina and offer a route to engineering future two-dimensional materials-based biosensors.
Collapse
Affiliation(s)
- Xiaosi Zhang
- Department of Electrical and Computer Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Hannah Lee
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN 37235, USA
| | - Yuchen Zhang
- Department of Electrical and Computer Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Thayer S. Walmsley
- Department of Physics and Astronomy, Vanderbilt University, Nashville, TN 37235, USA
| | - Deyu Li
- Department of Mechanical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Edward Levine
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN 37235, USA
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37235, USA
| | - Ya-Qiong Xu
- Department of Electrical and Computer Engineering, Vanderbilt University, Nashville, TN 37235, USA
- Department of Physics and Astronomy, Vanderbilt University, Nashville, TN 37235, USA
| |
Collapse
|
36
|
Nouri P, Zimmer A, Brüggemann S, Friedrich R, Kühn R, Prakash N. Generation of a NES-mScarlet Red Fluorescent Reporter Human iPSC Line for Live Cell Imaging and Flow Cytometric Analysis and Sorting Using CRISPR-Cas9-Mediated Gene Editing. Cells 2022; 11:268. [PMID: 35053384 PMCID: PMC8773741 DOI: 10.3390/cells11020268] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/07/2022] [Accepted: 01/11/2022] [Indexed: 12/16/2022] Open
Abstract
Advances in the regenerative stem cell field have propelled the generation of tissue-specific cells in the culture dish for subsequent transplantation, drug screening purposes, or the elucidation of disease mechanisms. One major obstacle is the heterogeneity of these cultures, in which the tissue-specific cells of interest usually represent only a fraction of all generated cells. Direct identification of the cells of interest and the ability to specifically isolate these cells in vitro is, thus, highly desirable for these applications. The type VI intermediate filament protein NESTIN is widely used as a marker for neural stem/progenitor cells (NSCs/NPCs) in the developing and adult central and peripheral nervous systems. Applying CRISPR-Cas9 technology, we have introduced a red fluorescent reporter (mScarlet) into the NESTIN (NES) locus of a human induced pluripotent stem cell (hiPSC) line. We describe the generation and characterization of NES-mScarlet reporter hiPSCs and demonstrate that this line is an accurate reporter of NSCs/NPCs during their directed differentiation into human midbrain dopaminergic (mDA) neurons. Furthermore, NES-mScarlet hiPSCs can be used for direct identification during live cell imaging and for flow cytometric analysis and sorting of red fluorescent NSCs/NPCs in this paradigm.
Collapse
Affiliation(s)
- Parivash Nouri
- Laboratory of Applied Genetics and Stem Cell Biology, Department Hamm 2, Hamm-Lippstadt University of Applied Sciences, 59063 Hamm, Germany; (P.N.); (S.B.); (R.F.)
| | - Anja Zimmer
- Genome Engineering & Disease Models, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany;
| | - Stefanie Brüggemann
- Laboratory of Applied Genetics and Stem Cell Biology, Department Hamm 2, Hamm-Lippstadt University of Applied Sciences, 59063 Hamm, Germany; (P.N.); (S.B.); (R.F.)
| | - Robin Friedrich
- Laboratory of Applied Genetics and Stem Cell Biology, Department Hamm 2, Hamm-Lippstadt University of Applied Sciences, 59063 Hamm, Germany; (P.N.); (S.B.); (R.F.)
| | - Ralf Kühn
- Genome Engineering & Disease Models, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany;
| | - Nilima Prakash
- Laboratory of Applied Genetics and Stem Cell Biology, Department Hamm 2, Hamm-Lippstadt University of Applied Sciences, 59063 Hamm, Germany; (P.N.); (S.B.); (R.F.)
| |
Collapse
|
37
|
Sahu G, Turner RW. The Molecular Basis for the Calcium-Dependent Slow Afterhyperpolarization in CA1 Hippocampal Pyramidal Neurons. Front Physiol 2022; 12:759707. [PMID: 35002757 PMCID: PMC8730529 DOI: 10.3389/fphys.2021.759707] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 11/01/2021] [Indexed: 12/02/2022] Open
Abstract
Neuronal signal transmission depends on the frequency, pattern, and timing of spike output, each of which are shaped by spike afterhyperpolarizations (AHPs). There are classically three post-spike AHPs of increasing duration categorized as fast, medium and slow AHPs that hyperpolarize a cell over a range of 10 ms to 30 s. Intensive early work on CA1 hippocampal pyramidal cells revealed that all three AHPs incorporate activation of calcium-gated potassium channels. The ionic basis for a fAHP was rapidly attributed to the actions of big conductance (BK) and the mAHP to small conductance (SK) or Kv7 potassium channels. In stark contrast, the ionic basis for a prominent slow AHP of up to 30 s duration remained an enigma for over 30 years. Recent advances in pharmacological, molecular, and imaging tools have uncovered the expression of a calcium-gated intermediate conductance potassium channel (IK, KCa3.1) in central neurons that proves to contribute to the slow AHP in CA1 hippocampal pyramidal cells. Together the data show that the sAHP arises in part from a core tripartite complex between Cav1.3 (L-type) calcium channels, ryanodine receptors, and IK channels at endoplasmic reticulum-plasma membrane junctions. Work on the sAHP in CA1 pyramidal neurons has again quickened pace, with identified contributions by both IK channels and the Na-K pump providing answers to several mysteries in the pharmacological properties of the sAHP.
Collapse
Affiliation(s)
- Giriraj Sahu
- National Institute of Pharmaceutical Education and Research Ahmedabad, Ahmedabad, India
| | - Ray W Turner
- Department Cell Biology & Anatomy, Cumming School of Medicine, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
38
|
Pak RW, Kang J, Boctor E, Kang JU. Optimization of Near-Infrared Fluorescence Voltage-Sensitive Dye Imaging for Neuronal Activity Monitoring in the Rodent Brain. Front Neurosci 2021; 15:742405. [PMID: 34776848 PMCID: PMC8582490 DOI: 10.3389/fnins.2021.742405] [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: 07/16/2021] [Accepted: 10/05/2021] [Indexed: 11/13/2022] Open
Abstract
Many currently employed clinical brain functional imaging technologies rely on indirect measures of activity such as hemodynamics resulting in low temporal and spatial resolutions. To improve upon this, optical systems were developed in conjunction with methods to deliver near-IR voltage-sensitive dye (VSD) to provide activity-dependent optical contrast to establish a clinical tool to facilitate direct monitoring of neuron depolarization through the intact skull. Following the previously developed VSD delivery protocol through the blood-brain barrier, IR-780 perchlorate VSD concentrations in the brain were varied and stimulus-evoked responses were observed. In this paper, a range of optimal VSD tissue concentrations was established that maximized fluorescence fractional change for detection of membrane potential responses to external stimuli through a series of phantom, in vitro, ex vivo, and in vivo experiments in mouse models.
Collapse
Affiliation(s)
- Rebecca W Pak
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Jeeun Kang
- Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD, United States
| | - Emad Boctor
- Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD, United States
| | - Jin U Kang
- Department of Electrical and Computer Engineering, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
39
|
DeKorver NW, Chaudoin TR, Zhao G, Wang D, Arikkath J, Bonasera SJ. Complement Component C3 Loss leads to Locomotor Deficits and Altered Cerebellar Internal Granule Cell In Vitro Synaptic Protein Expression in C57BL/6 Mice. Mol Neurobiol 2021; 58:5857-5875. [PMID: 34415487 DOI: 10.1007/s12035-021-02480-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 06/24/2021] [Indexed: 01/14/2023]
Abstract
Complement component 3 (C3) expression is increased in the cerebellum of aging mice that demonstrate locomotor impairments and increased excitatory synapse density. However, C3 regulation of locomotion, as well as C3 roles in excitatory synapse function, remains poorly understood. Here, we demonstrate that constitutive loss of C3 function in mice evokes a locomotor phenotype characterized by decreased speed, increased active state locomotor probability, and gait ataxia. C3 loss does not alter metabolism or body mass composition. No evidence of significant muscle weakness or degenerative arthritis was found in C3 knockout mice to explain decreased gait speeds. In an enriched primary cerebellar granule cell culture model, loss of C3 protein results in increased excitatory synaptic density and increased response to KCl depolarization. Our analysis of excitatory synaptic density in the cerebellar internal granule cell and molecular layers did not demonstrate increased synaptic density in vivo, suggesting the presence of compensatory mechanisms regulating synaptic development. Functional deficits in C3 knockout mice are therefore more likely to result from altered synaptic function and/or connectivity than gross synaptic deficits. Our data demonstrate a novel role for complement proteins in cerebellar regulation of locomotor output and control.
Collapse
Affiliation(s)
- Nicholas W DeKorver
- Department of Internal Medicine, Division of Geriatrics, University of Nebraska Medical Center, 3028 Durham Research Center II, Omaha, NE, 68198-5039, USA
| | - Tammy R Chaudoin
- Department of Internal Medicine, Division of Geriatrics, University of Nebraska Medical Center, 3028 Durham Research Center II, Omaha, NE, 68198-5039, USA
| | - Gang Zhao
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, 986125 Nebraska Medical Center, PDD 3020, Omaha, NE, 68198-6125, USA
| | - Dong Wang
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, 986125 Nebraska Medical Center, PDD 3020, Omaha, NE, 68198-6125, USA
| | - Jyothi Arikkath
- Monroe-Meyer Institute, University of Nebraska Medical Center, 3031 Durham Research Center II, Omaha, NE, 68198-5960, USA
| | - Stephen J Bonasera
- Department of Internal Medicine, Division of Geriatrics, University of Nebraska Medical Center, 3028 Durham Research Center II, Omaha, NE, 68198-5039, USA.
| |
Collapse
|
40
|
Liakath-Ali K, Südhof TC. The Perils of Navigating Activity-Dependent Alternative Splicing of Neurexins. Front Mol Neurosci 2021; 14:659681. [PMID: 33767611 PMCID: PMC7985251 DOI: 10.3389/fnmol.2021.659681] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 02/16/2021] [Indexed: 11/13/2022] Open
Abstract
Neurexins are presynaptic cell-adhesion molecules essential for synaptic function that are expressed in thousands of alternatively spliced isoforms. Recent studies suggested that alternative splicing at splice site 4 (SS4) of Nrxn1 is tightly regulated by an activity-dependent mechanism. Given that Nrxn1 alternative splicing at SS4 controls NMDA-receptor-mediated synaptic responses, activity-dependent SS4 alternative splicing would suggest a new synaptic plasticity mechanism. However, conflicting results confound the assessment of neurexin alternative splicing, prompting us to re-evaluate this issue. We find that in cortical cultures, membrane depolarization by elevated extracellular K+-concentrations produced an apparent shift in Nrxn1-SS4 alternative splicing by inducing neuronal but not astroglial cell death, resulting in persistent astroglial Nrxn1-SS4+ expression and decreased neuronal Nrxn1-SS4- expression. in vivo, systemic kainate-induced activation of neurons in the hippocampus produced no changes in Nrxn1-SS4 alternative splicing. Moreover, focal kainate injections into the mouse cerebellum induced small changes in Nrxn1-SS4 alternative splicing that, however, were associated with large decreases in Nrxn1 expression and widespread DNA damage. Our results suggest that although Nrxn1-SS4 alternative splicing may represent a mechanism of activity-dependent synaptic plasticity, common procedures for testing this hypothesis are prone to artifacts, and more sophisticated approaches will be necessary to test this important question.
Collapse
Affiliation(s)
- Kif Liakath-Ali
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, United States
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, United States
| | - Thomas C. Südhof
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, United States
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, United States
| |
Collapse
|