1
|
Nagaeva E, Turconi G, Mätlik K, Segerstråle M, Olfat S, Iivanainen V, Taira T, Andressoo JO. Motor learning is regulated by postnatal GDNF levels in Purkinje cells. Neuroscience 2025; 576:27-41. [PMID: 40254124 DOI: 10.1016/j.neuroscience.2025.04.030] [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: 12/27/2024] [Revised: 04/08/2025] [Accepted: 04/17/2025] [Indexed: 04/22/2025]
Abstract
Purkinje cells (PCs), the sole output neurons of the cerebellar cortex, are crucial for cerebellum-dependent motor learning. In cerebellar ataxia, reduction in motor function and learning associates with decreased spontaneous activity of PCs. Thus, understanding what molecules regulate PCs activity is important. Previously, we demonstrated that a ubiquitous 2-fold increase of endogenous glial cell line-derived neurotrophic factor (GDNF) improves motor function in adult mice and motor learning and coordination in aged mice. However, since GDNF impacts many organ systems the underlying mechanism remained elusive. Here we utilize GDNF Hypermorphic, conditional GDNF Hypermorphic and conditional knock-out mouse models to reveal that up to a 2-fold increase in endogenous GDNF, specifically in PCs postnatally, is sufficient to enhance motor learning. We find that improved motor learning associates with increased glutamatergic input to PCs and with elevated spontaneous firing rate of PCs, opposite to cerebellar ataxia where reduction in motor function and learning associates with decreased spontaneous activity of PCs. Analysis of the human cerebellum revealed that normal interindividual variation in GDNF expression levels falls in the same variation range as studied in the mouse models, suggesting that interindividual variation in PC GDNF levels may contribute to interindividual variation in PC function. Collectively, our findings reveal how a relatively small change in postnatal GDNF expression level within the physiological range in one cell type, the PCs, affects motor learning. Thus, drugs enhancing postnatal GDNF expression in PCs or cerebellar GDNF signaling may have potential in treating cerebellar ataxias, making an interesting topic for future studies.
Collapse
Affiliation(s)
- Elina Nagaeva
- Department of Pharmacology, Faculty of Medicine & Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Giorgio Turconi
- Department of Pharmacology, Faculty of Medicine & Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Kärt Mätlik
- Department of Pharmacology, Faculty of Medicine & Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Mikael Segerstråle
- Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Soophie Olfat
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, Stockholm, Sweden
| | - Vilma Iivanainen
- Department of Pharmacology, Faculty of Medicine & Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Tomi Taira
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Finland
| | - Jaan-Olle Andressoo
- Department of Pharmacology, Faculty of Medicine & Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland; Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
2
|
Mi X, Chen ABY, Duarte D, Carey E, Taylor CR, Braaker PN, Bright M, Almeida RG, Lim JX, Ruetten VMS, Wang Y, Wang M, Zhang W, Zheng W, Reitman ME, Huang Y, Wang X, Li L, Deng H, Shi SH, Poskanzer KE, Lyons DA, Nimmerjahn A, Ahrens MB, Yu G. Fast, accurate, and versatile data analysis platform for the quantification of molecular spatiotemporal signals. Cell 2025; 188:2794-2809.e21. [PMID: 40203826 DOI: 10.1016/j.cell.2025.03.012] [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: 05/31/2024] [Revised: 01/13/2025] [Accepted: 03/06/2025] [Indexed: 04/11/2025]
Abstract
Optical recording of intricate molecular dynamics is becoming an indispensable technique for biological studies, accelerated by the development of new or improved biosensors and microscopy technology. This creates major computational challenges to extract and quantify biologically meaningful spatiotemporal patterns embedded within complex and rich data sources, many of which cannot be captured with existing methods. Here, we introduce activity quantification and analysis (AQuA2), a fast, accurate, and versatile data analysis platform built upon advanced machine-learning techniques. It decomposes complex live-imaging-based datasets into elementary signaling events, allowing accurate and unbiased quantification of molecular activities and identification of consensus functional units. We demonstrate applications across a wide range of biosensors, cell types, organs, animal models, microscopy techniques, and imaging approaches. As exemplar findings, we show how AQuA2 identified drug-dependent interactions between neurons and astroglia, as well as distinct sensorimotor signal propagation patterns in the mouse spinal cord.
Collapse
Affiliation(s)
- Xuelong Mi
- Bradley Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA 22203, USA
| | - Alex Bo-Yuan Chen
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA; Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA; Graduate Program in Neuroscience, Harvard Medical School, Boston, MA 02115, USA
| | - Daniela Duarte
- Waitt Advanced Biophotonics Center, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Erin Carey
- Waitt Advanced Biophotonics Center, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Charlotte R Taylor
- Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA, USA; Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Philipp N Braaker
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh BioQuarter, Edinburgh EH16 4SB, UK
| | - Mark Bright
- Bradley Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA 22203, USA
| | - Rafael G Almeida
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh BioQuarter, Edinburgh EH16 4SB, UK
| | - Jing-Xuan Lim
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Virginia M S Ruetten
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA; Gatsby Computational Neuroscience Unit, UCL, London W1T 4JG, UK
| | - Yizhi Wang
- Bradley Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA 22203, USA
| | - Mengfan Wang
- Bradley Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA 22203, USA
| | - Weizhan Zhang
- Department of Automation, Tsinghua University, Beijing 100084, China; IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China
| | - Wei Zheng
- Bradley Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA 22203, USA
| | - Michael E Reitman
- Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA, USA; Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Yongkang Huang
- IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China; New Cornerstone Science Laboratory, Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Xiaoyu Wang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Lei Li
- IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China; New Cornerstone Science Laboratory, Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - HanFei Deng
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Song-Hai Shi
- IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China; New Cornerstone Science Laboratory, Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Kira E Poskanzer
- Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA, USA; Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA; Kavli Institute for Fundamental Neuroscience, San Francisco, CA, USA
| | - David A Lyons
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh BioQuarter, Edinburgh EH16 4SB, UK
| | - Axel Nimmerjahn
- Waitt Advanced Biophotonics Center, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA.
| | - Misha B Ahrens
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA.
| | - Guoqiang Yu
- Department of Automation, Tsinghua University, Beijing 100084, China; IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China; Beijing National Research Center for Information Science and Technology, Beijing 100084, China.
| |
Collapse
|
3
|
Bareghamyan A, Deng C, Daoudi S, Yadav SC, Lu X, Zhang W, Campbell RE, Kramer RH, Chenoweth DM, Arnold DB. A toolbox for ablating excitatory and inhibitory synapses. eLife 2025; 13:RP103757. [PMID: 40298577 PMCID: PMC12040315 DOI: 10.7554/elife.103757] [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: 04/30/2025] Open
Abstract
Recombinant optogenetic and chemogenetic proteins are potent tools for manipulating neuronal activity and controlling neural circuit function. However, there are few analogous tools for manipulating the structure of neural circuits. Here, we introduce three rationally designed genetically encoded tools that use E3 ligase-dependent mechanisms to trigger the degradation of synaptic scaffolding proteins, leading to functional ablation of synapses. First, we developed a constitutive excitatory synapse ablator, PFE3, analogous to the inhibitory synapse ablator GFE3. PFE3 targets the RING domain of the E3 ligase Mdm2 and the proteasome-interacting region of Protocadherin 10 to the scaffolding protein PSD-95, leading to efficient ablation of excitatory synapses. In addition, we developed a light-inducible version of GFE3, paGFE3, using a novel photoactivatable complex based on the photocleavable protein PhoCl2c. paGFE3 degrades Gephyrin and ablates inhibitory synapses in response to 400 nm light. Finally, we developed a chemically inducible version of GFE3, chGFE3, which degrades inhibitory synapses when combined with the bio-orthogonal dimerizer HaloTag ligand-trimethoprim. Each tool is specific, reversible, and capable of breaking neural circuits at precise locations.
Collapse
Affiliation(s)
- Aida Bareghamyan
- Department of Biology, Division of Molecular and Computational Biology, University of Southern CaliforniaLos AngelesUnited States
- Neuroscience Graduate Program, University of Southern CaliforniaLos AngelesUnited States
| | - Changfeng Deng
- Department of Chemistry, School of Arts and Sciences, University of PennsylvaniaPhiladelphiaUnited States
| | - Sarah Daoudi
- Department of Biology, Division of Molecular and Computational Biology, University of Southern CaliforniaLos AngelesUnited States
| | - Shubhash C Yadav
- Department of Molecular and Cell Biology, University of CaliforniaBerkeleyUnited States
| | - Xiaocen Lu
- Department of Chemistry, Faculty of Science, University of AlbertaEdmontonUnited States
| | - Wei Zhang
- Department of Chemistry, Faculty of Science, University of AlbertaEdmontonUnited States
| | - Robert E Campbell
- Department of Chemistry, Faculty of Science, University of AlbertaEdmontonUnited States
- Department of Chemistry, Graduate School of Science, The University of Tokyo, Bunkyo-kuTokyoJapan
| | - Richard H Kramer
- Department of Molecular and Cell Biology, University of CaliforniaBerkeleyUnited States
| | - David M Chenoweth
- Department of Chemistry, School of Arts and Sciences, University of PennsylvaniaPhiladelphiaUnited States
| | - Don B Arnold
- Department of Biology, Division of Molecular and Computational Biology, University of Southern CaliforniaLos AngelesUnited States
- Neuroscience Graduate Program, University of Southern CaliforniaLos AngelesUnited States
- Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern CaliforniaLos AngelesUnited States
| |
Collapse
|
4
|
Petrušková A, Guhathakurta D, Akdaş EY, Perelló‐Amorós B, Frischknecht R, Weiss E, Páleníček T, Fejtová A. Serotonergic Psychedelics Rapidly Modulate Evoked Glutamate Release in Cultured Cortical Neurons. J Neurochem 2025; 169:e70020. [PMID: 40022486 PMCID: PMC11871419 DOI: 10.1111/jnc.70020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 01/10/2025] [Accepted: 02/05/2025] [Indexed: 03/03/2025]
Abstract
The serotonergic psychedelics psilocybin, LSD and DMT hold great promise for the development of new treatments for psychiatric conditions such as major depressive disorder, addiction and end-of-life anxiety. Previous studies in both animals and humans have confirmed the effects of these drugs on neuronal activity and plasticity. However, the understanding of the mechanisms of action of these substances is limited. Here we show rapid effects of psychedelics on presynaptic properties, using live cell imaging at the level of single synapses in primary rat cortical neurons. Using the genetically encoded reporter of synaptic vesicle fusion synaptopHluorin, we detected a reduced fraction of synaptic vesicles that fused in response to mild or strong electrical stimulation 3-30 min after application of serotonergic psychedelics. These effects were transient and no longer present 24 h after treatment. While DMT only reduced the total recycling pool, LSD and psilocin also reduced the size of the readily releasable vesicle pool. Imaging with the sensors for glutamate, iGluSnFR, and presynaptic calcium, synGCaMP6, showed that while psilocin and DMT increased evoked glutamate release, LSD and psilocin reduced evoked presynaptic calcium levels. Interestingly, psilocin also affected short-term plasticity leading to a depression of responses to paired stimuli. The rapid and drug-specific modulation of glutamatergic neurotransmission described in this study may contribute to distinct anxiolytic and antidepressant properties of serotonergic psychedelics.
Collapse
Affiliation(s)
- Aneta Petrušková
- Department of Psychiatry and PsychotherapyUniversitätsklinikum Erlangen, Friedrich‐Alexander‐Universität Erlangen‐NürnbergErlangenGermany
- National Institute of Mental HealthKlecanyCzech Republic
- Third Faculty of MedicineCharles UniversityPragueCzech Republic
| | - Debarpan Guhathakurta
- Department of Psychiatry and PsychotherapyUniversitätsklinikum Erlangen, Friedrich‐Alexander‐Universität Erlangen‐NürnbergErlangenGermany
| | - Enes Yağız Akdaş
- Department of Psychiatry and PsychotherapyUniversitätsklinikum Erlangen, Friedrich‐Alexander‐Universität Erlangen‐NürnbergErlangenGermany
| | - Bartomeu Perelló‐Amorós
- Department of Biology, Animal PhysiologyFriedrich‐Alexander‐Universität Erlangen‐NürnbergErlangenGermany
| | - Renato Frischknecht
- Department of Biology, Animal PhysiologyFriedrich‐Alexander‐Universität Erlangen‐NürnbergErlangenGermany
| | - Eva‐Maria Weiss
- Department of Psychiatry and PsychotherapyUniversitätsklinikum Erlangen, Friedrich‐Alexander‐Universität Erlangen‐NürnbergErlangenGermany
| | - Tomáš Páleníček
- National Institute of Mental HealthKlecanyCzech Republic
- Third Faculty of MedicineCharles UniversityPragueCzech Republic
| | - Anna Fejtová
- Department of Psychiatry and PsychotherapyUniversitätsklinikum Erlangen, Friedrich‐Alexander‐Universität Erlangen‐NürnbergErlangenGermany
| |
Collapse
|
5
|
Pollitt SL, Levy AD, Anderson MC, Blanpied TA. Large Donor CRISPR for Whole-Coding Sequence Replacement of Cell Adhesion Molecule LRRTM2. J Neurosci 2025; 45:e1461242024. [PMID: 39824639 PMCID: PMC11823385 DOI: 10.1523/jneurosci.1461-24.2024] [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: 08/01/2024] [Revised: 11/01/2024] [Accepted: 12/02/2024] [Indexed: 01/20/2025] Open
Abstract
The cell adhesion molecule leucine-rich repeat transmembrane neuronal protein 2 (LRRTM2) is crucial for synapse development and function. However, our understanding of its endogenous trafficking has been limited due to difficulties in manipulating its coding sequence (CDS) using standard genome editing techniques. Instead, we replaced the entire LRRTM2 CDS by adapting a two-guide CRISPR knock-in method, enabling complete control of LRRTM2. In primary rat hippocampal cultures dissociated from embryos of both sexes, N-terminally tagged, endogenous LRRTM2 was found in 80% of synapses, and synaptic LRRTM2 content correlated with PSD-95 and AMPAR levels. LRRTM2 was also enriched with AMPARs outside synapses, demonstrating the sensitivity of this method to detect relevant new biology. Finally, we leveraged total genomic control to increase the synaptic levels of LRRTM2 via simultaneous mutation of its C-terminal domain, which did not correspondingly increase AMPAR enrichment. The coding region of thousands of genes span lengths suitable for whole-CDS replacement, suggesting this simple approach will enable straightforward structure-function analysis in neurons.
Collapse
Affiliation(s)
- Stephanie L Pollitt
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland 21201-1509
- University of Maryland-Medicine Institute for Neuroscience Discovery (UM-MIND), University of Maryland School of Medicine, Baltimore, Maryland 21201-1509
| | - Aaron D Levy
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland 21201-1509
- University of Maryland-Medicine Institute for Neuroscience Discovery (UM-MIND), University of Maryland School of Medicine, Baltimore, Maryland 21201-1509
| | - Michael C Anderson
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland 21201-1509
- University of Maryland-Medicine Institute for Neuroscience Discovery (UM-MIND), University of Maryland School of Medicine, Baltimore, Maryland 21201-1509
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland 21201-1509
| | - Thomas A Blanpied
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland 21201-1509
- University of Maryland-Medicine Institute for Neuroscience Discovery (UM-MIND), University of Maryland School of Medicine, Baltimore, Maryland 21201-1509
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland 21201-1509
| |
Collapse
|
6
|
Bareghamyan A, Deng C, Daoudi S, Yadav SC, Lu X, Zhang W, Campbell RE, Kramer RH, Chenoweth DM, Arnold DB. A toolbox for ablating excitatory and inhibitory synapses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.09.23.614589. [PMID: 39974889 PMCID: PMC11838203 DOI: 10.1101/2024.09.23.614589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Recombinant optogenetic and chemogenetic proteins are potent tools for manipulating neuronal activity and controlling neural circuit function. However, there are few analogous tools for manipulating the structure of neural circuits. Here, we introduce three rationally designed genetically encoded tools that use E3 ligase-dependent mechanisms to trigger the degradation of synaptic scaffolding proteins, leading to functional ablation of synapses. First, we developed a constitutive excitatory synapse ablator, PFE3, analogous to the inhibitory synapse ablator GFE3. PFE3 targets the RING domain of the E3 ligase Mdm2 and the proteasome-interacting region of Protocadherin 10 to the scaffolding protein PSD-95, leading to efficient ablation of excitatory synapses. In addition, we developed a light-inducible version of GFE3, paGFE3, using a novel photoactivatable complex based on the photocleavable protein PhoCl2c. paGFE3 degrades Gephyrin and ablates inhibitory synapses in response to 400 nm light. Finally, we developed a chemically inducible version of GFE3, chGFE3, which degrades inhibitory synapses when combined with the bio-orthogonal dimerizer HaloTag ligand-trimethoprim. Each tool is specific, reversible, and capable of breaking neural circuits at precise locations.
Collapse
Affiliation(s)
- Aida Bareghamyan
- Department of Biology, Division of Molecular and Computational Biology, University of Southern California, Los Angeles, CA 90089, USA
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA 90089, USA
| | - Changfeng Deng
- Department of Chemistry, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sarah Daoudi
- Department of Biology, Division of Molecular and Computational Biology, University of Southern California, Los Angeles, CA 90089, USA
| | - Shubash C Yadav
- Department of Molecular and Cell Biology, University of California, Berkeley, California, 94720, USA
| | - Xiaocen Lu
- Department of Chemistry, Faculty of Science, University of Alberta, Edmonton, Alberta, T6G 2G2, Canada
| | - Wei Zhang
- Department of Chemistry, Faculty of Science, University of Alberta, Edmonton, Alberta, T6G 2G2, Canada
| | - Robert E Campbell
- Department of Chemistry, Faculty of Science, University of Alberta, Edmonton, Alberta, T6G 2G2, Canada
- Department of Chemistry, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Richard H Kramer
- Department of Molecular and Cell Biology, University of California, Berkeley, California, 94720, USA
| | - David M Chenoweth
- Department of Chemistry, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Don B Arnold
- Department of Biology, Division of Molecular and Computational Biology, University of Southern California, Los Angeles, CA 90089, USA
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA 90089, USA
- Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA
| |
Collapse
|
7
|
Barlow ST, Levy AD, Contreras M, Anderson MC, Blanpied TA. Dissecting the functional heterogeneity of glutamatergic synapses with high-throughput optical physiology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.23.629904. [PMID: 39763981 PMCID: PMC11703220 DOI: 10.1101/2024.12.23.629904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
Fluorescent reporters for glutamate release and postsynaptic Ca2+ signaling are essential tools for quantifying synapse functional heterogeneity across neurons and circuits. However, leveraging these probes for neuroscience requires scalable experimental frameworks. Here, we devised a high-throughput approach to efficiently collect and analyze hundreds of optical recordings of glutamate release activity at presynaptic boutons in cultured rat hippocampal neurons. Boutons exhibited remarkable functional heterogeneity and could be separated into multiple functional classes based on their iGluSnFR3 responses to single action potentials, paired stimuli, and synaptic parameters derived from mean-variance analysis. Finally, we developed a novel all-optical assay of pre- and postsynaptic glutamatergic synapse function. We deployed iGluSnFR3 with a red-shifted, postsynaptically-targeted Ca2+ sensor, enabling direct imaging and analysis of NMDA receptor-mediated synaptic transmission at large numbers of dendritic spines. This work enables direct observation of the flow of information at single synapses and should speed detailed investigations of synaptic functional heterogeneity.
Collapse
Affiliation(s)
- Samuel T Barlow
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- UM-MIND, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Aaron D Levy
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- UM-MIND, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Minerva Contreras
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- UM-MIND, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Michael C Anderson
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Thomas A Blanpied
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- UM-MIND, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
8
|
Terpstra HM, Gómez-Sánchez R, Veldsink AC, Otto TA, Veenhoff LM, Heinemann M. PunctaFinder: An algorithm for automated spot detection in fluorescence microscopy images. Mol Biol Cell 2024; 35:mr9. [PMID: 39535892 PMCID: PMC11656481 DOI: 10.1091/mbc.e24-06-0254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 10/24/2024] [Accepted: 11/30/2024] [Indexed: 11/16/2024] Open
Abstract
Fluorescence microscopy has revolutionized biological research by enabling the visualization of subcellular structures at high resolution. With the increasing complexity and volume of microscopy data, there is a growing need for automated image analysis to ensure efficient and consistent interpretation. In this study, we introduce PunctaFinder, a novel Python-based algorithm designed to detect puncta, small bright spots, in raw fluorescence microscopy images without image denoising or signal enhancement steps. Furthermore, unlike other available spot detectors, PunctaFinder not only detects puncta, but also defines the cytoplasmic region, making it a valuable tool to quantify target molecule localization in cellular contexts. PunctaFinder is a widely applicable punctum detector and size estimator, as evidenced by its successful detection of Atg9-positive vesicles, lipid droplets, aggregates of a destabilized luciferase mutant, and the nuclear pore complex. Notably, PunctaFinder excels in detecting puncta in images with a relatively low resolution and signal-to-noise ratio, demonstrating its capability to identify dim puncta and puncta of dynamic target molecules. PunctaFinder reliably detects puncta in fluorescence microscopy images where automated analysis was not possible before, providing researchers with an efficient and robust method for punctum quantification in fluorescence microscopy images.
Collapse
Affiliation(s)
- Hanna M. Terpstra
- Molecular Systems Biology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, 9747 AG, Groningen, The Netherlands
| | - Rubén Gómez-Sánchez
- Department of Biomedical Sciences, University of Groningen, University Medical Center Groningen, 9713 AV Groningen, The Netherlands
| | - Annemiek C. Veldsink
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Center, 9713 AV Groningen, The Netherlands
| | - Tegan A. Otto
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Center, 9713 AV Groningen, The Netherlands
| | - Liesbeth M. Veenhoff
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Center, 9713 AV Groningen, The Netherlands
| | - Matthias Heinemann
- Molecular Systems Biology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, 9747 AG, Groningen, The Netherlands
| |
Collapse
|
9
|
Guhathakurta D, Selzam F, Petrušková A, Weiss EM, Akdaş EY, Montenegro-Venegas C, Zenker M, Fejtová A. Rasopathy-Associated Mutation Ptpn11 D61Y has Age-Dependent Effect on Synaptic Vesicle Recycling. Cell Mol Neurobiol 2024; 44:77. [PMID: 39570442 PMCID: PMC11582327 DOI: 10.1007/s10571-024-01505-1] [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: 05/12/2024] [Accepted: 10/14/2024] [Indexed: 11/22/2024]
Abstract
Rasopathies are genetic disorders often associated with developmental delay and intellectual disability. Noonan syndrome (NS) is one of the most common Rasopathies, caused by mutations in PTPN11 in more than 50% of cases. In mammalian neurons, PTPN11 controls the trafficking of postsynaptic glutamate receptors. This process is disrupted in neurons expressing PTPN11 variants associated with Rasopathies and is thought to contribute to the cognitive impairments in Noonan syndrome. Recent work revealed presynaptic impairments upon expression of RASopathy-linked PTPN11 variants in Drosophila. However, the presynaptic role of PTPN11 has not yet been addressed in mammals. Here, we investigated membrane trafficking of synaptic vesicles in cultured mouse cortical neurons expressing Rasopathy-associated PTPN11D61Y variant. We observed a significantly smaller readily releasable and total recycling pool of synaptic vesicles. The drop in synaptic vesicle release competence was accompanied by a decreased rate of SV retrieval. Interestingly, the presynaptic phenotype was evident in mature (DIV21) but not in immature (DIV12) neurons. Thus, our data reveal importance of balanced PTPN11 activity for normal trafficking of neurotransmitter-filled synaptic vesicles in the presynaptic ending of mature neurons.
Collapse
Affiliation(s)
- Debarpan Guhathakurta
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Franziska Selzam
- RG Presynaptic Plasticity, Leibniz Institute for Neurobiology, Magdeburg, Germany
- Institute of Human Genetics, Medical Faculty, University Hospital Magdeburg, Otto Von Guericke University, Magdeburg, Germany
| | - Aneta Petrušková
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- National Institute of Mental Health, Klecany, Czech Republic
- Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Eva-Maria Weiss
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Enes Yağız Akdaş
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Carolina Montenegro-Venegas
- Department of Neurochemistry and Molecular Biology, Leibniz Institute for Neurobiology, Magdeburg, Germany
- Institute for Pharmacology and Toxicology, Medical Faculty, Otto von Guericke University, Magdeburg, Germany
| | - Martin Zenker
- Institute of Human Genetics, Medical Faculty, University Hospital Magdeburg, Otto Von Guericke University, Magdeburg, Germany
| | - Anna Fejtová
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
- RG Presynaptic Plasticity, Leibniz Institute for Neurobiology, Magdeburg, Germany.
| |
Collapse
|
10
|
Salazar MPR, Kolanukuduru1 S, Ramirez V, Lyu B, Sejourne G, Sesaki H, Yu G, Eroglu C. Mitochondrial fission controls astrocyte morphogenesis and organization in the cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.22.619706. [PMID: 39484572 PMCID: PMC11527035 DOI: 10.1101/2024.10.22.619706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Dysfunctional mitochondrial dynamics are a hallmark of devastating neurodevelopmental disorders such as childhood refractory epilepsy. However, the role of glial mitochondria in proper brain development is not well understood. We show that astrocyte mitochondria undergo extensive fission while populating astrocyte distal branches during postnatal cortical development. Loss of mitochondrial fission regulator, Dynamin-related protein 1 (Drp1), decreases mitochondrial localization to distal astrocyte processes, and this mitochondrial mislocalization reduces astrocyte morphological complexity. Functionally, astrocyte-specific conditional deletion of Drp1 induces astrocyte reactivity and disrupts astrocyte organization in the cortex. These morphological and organizational deficits are accompanied by loss of astrocytic gap junction protein Connexin 43. These findings uncover a crucial role for mitochondrial fission in coordinating astrocytic morphogenesis and organization, revealing the regulation of astrocytic mitochondria dynamics as a critical step in neurodevelopment.
Collapse
Affiliation(s)
| | - Sprihaa Kolanukuduru1
- The Department of Cell Biology, Duke University Medical Center, Durham, NC, USA
- The Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Valentina Ramirez
- The Department of Cell Biology, Duke University Medical Center, Durham, NC, USA
- The Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Boyu Lyu
- Bradley Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA, USA
| | - Gabrielle Sejourne
- The Department of Cell Biology, Duke University Medical Center, Durham, NC, USA
| | - Hiromi Sesaki
- Department of Cell Biology, John Hopkins University School of Medicine, Baltimore, MD
| | - Guoqiang Yu
- Department of Automation, Tsinghua University, Beijing 100084, China
- IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China
| | - Cagla Eroglu
- The Department of Cell Biology, Duke University Medical Center, Durham, NC, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD
- The Department of Neurobiology, Duke University Medical Center, Durham, NC, USA
- Howard Hughes Medical Institute, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
11
|
Cavalli P, Raffauf A, Passarella S, Helmuth M, Dieterich DC, Landgraf P. Manipulation of DHPS activity affects dendritic morphology and expression of synaptic proteins in primary rat cortical neurons. Front Cell Neurosci 2024; 18:1465011. [PMID: 39469305 PMCID: PMC11513877 DOI: 10.3389/fncel.2024.1465011] [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: 07/15/2024] [Accepted: 09/23/2024] [Indexed: 10/30/2024] Open
Abstract
Deoxyhypusine synthase (DHPS) catalyzes the initial step of hypusine incorporation into the eukaryotic initiation factor 5A (eIF5A), leading to its activation. The activated eIF5A, in turn, plays a key role in regulating the protein translation of selected mRNAs and therefore appears to be a suitable target for therapeutic intervention strategies. In the present study, we analyzed the role of DHPS-mediated hypusination in regulating neuronal homeostasis using lentivirus-based gain and loss of function experiments in primary cortical cultures from rats. This model allows us to examine the impact of DHPS function on the composition of the dendritic and synaptic compartments, which may contribute to a better understanding of cognitive function and neurodevelopment in vivo. Our findings revealed that shRNA-mediated DHPS knockdown diminishes the amount of hypusinated eIF5A (eIF5AHyp), resulting in notable alterations in neuronal dendritic architecture. Furthermore, in neurons, the synaptic composition was also affected, showing both pre- and post-synaptic changes, while the overexpression of DHPS had only a minor impact. Therefore, we hypothesize that interfering with the eIF5A hypusination caused by reduced DHPS activity impairs neuronal and synaptic homeostasis.
Collapse
Affiliation(s)
- Paola Cavalli
- Institute for Pharmacology and Toxicology, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Anna Raffauf
- Institute for Pharmacology and Toxicology, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Sergio Passarella
- Institute for Pharmacology and Toxicology, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Martin Helmuth
- Institute for Pharmacology and Toxicology, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Daniela C. Dieterich
- Institute for Pharmacology and Toxicology, Otto von Guericke University Magdeburg, Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
| | - Peter Landgraf
- Institute for Pharmacology and Toxicology, Otto von Guericke University Magdeburg, Magdeburg, Germany
| |
Collapse
|
12
|
Savage JT, Ramirez JJ, Risher WC, Wang Y, Irala D, Eroglu C. SynBot is an open-source image analysis software for automated quantification of synapses. CELL REPORTS METHODS 2024; 4:100861. [PMID: 39255792 PMCID: PMC11440803 DOI: 10.1016/j.crmeth.2024.100861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 04/25/2024] [Accepted: 08/16/2024] [Indexed: 09/12/2024]
Abstract
The formation of precise numbers of neuronal connections, known as synapses, is crucial for brain function. Therefore, synaptogenesis mechanisms have been one of the main focuses of neuroscience. Immunohistochemistry is a common tool for visualizing synapses. Thus, quantifying the numbers of synapses from light microscopy images enables screening the impacts of experimental manipulations on synapse development. Despite its utility, this approach is paired with low-throughput analysis methods that are challenging to learn, and the results are variable between experimenters, especially when analyzing noisy images of brain tissue. We developed an open-source ImageJ-based software, SynBot, to address these technical bottlenecks by automating the analysis. SynBot incorporates the advanced algorithms ilastik and SynQuant for accurate thresholding for synaptic puncta identification, and the code can easily be modified by users. The use of this software will allow for rapid and reproducible screening of synaptic phenotypes in healthy and diseased nervous systems.
Collapse
Affiliation(s)
- Justin T Savage
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Juan J Ramirez
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
| | - W Christopher Risher
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine at Marshall University, Huntington, WV 25755, USA
| | - Yizhi Wang
- Bradley Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA 22203, USA
| | - Dolores Irala
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA.
| | - Cagla Eroglu
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA; Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA; Howard Hughes Medical Institute, Duke University Medical Center, Durham, NC 27710, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA.
| |
Collapse
|
13
|
Wu S, Xie J, Zhao H, Zhao X, Sánchez OF, Rochet JC, Freeman JL, Yuan C. Developmental neurotoxicity of PFOA exposure on hiPSC-derived cortical neurons. ENVIRONMENT INTERNATIONAL 2024; 190:108914. [PMID: 39079332 PMCID: PMC11406754 DOI: 10.1016/j.envint.2024.108914] [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: 04/15/2024] [Revised: 06/27/2024] [Accepted: 07/24/2024] [Indexed: 08/28/2024]
Abstract
PFOA is a legacy Per- and Polyfluorinated Substances (PFAS), a group of chemicals widely used in various industrial applications and consumer products. Although there has been a voluntary phase out of PFOA since 2005, it is still widely detected in various water supplies. A growing body of evidence suggests an association between PFOA exposure, particularly during developmental stages, with increased risks of neurodegenerative diseases (NDs). The neurotoxic mechanism of developmental PFOA exposure, however, remains poorly understood. Utilizing human induced-pluripotent stem cell (hiPSC)-derived cortical neurons, we investigated the effect of PFOA exposure prior to differentiation and assessed changes in neuronal characteristics, transcriptome, and neurodegeneration markers mimicking a Developmental Origin of Health and Disease (DoHAD) paradigm. Exposure to PFOA before neuron differentiation resulted in persistent alterations in nuclear morphology, neuronal network, and calcium activity. RNA sequencing analysis further revealed transcriptomic changes aligning with Alzheimer's Disease (AD) after PFOA exposure. These observations were further corroborated by alterations in tau phosphorylation markers, the presence of fibrillar tau, an increase in liquid droplets, and a decrease in RNA translational efficiency characterized using a battery of biochemical assays. Taken together, our results revealed persistent deficits of key neuronal characteristics induced by pre-differentiation PFOA exposure, suggesting impairments in several AD-related pathways that can together contribute to the elevation of AD risk after pre-differentiation PFOA exposure.
Collapse
Affiliation(s)
- Shichen Wu
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN, 47907, USA
| | - Junkai Xie
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN, 47907, USA
| | - Han Zhao
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN, 47907, USA
| | - Xihui Zhao
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, 47907, USA
| | - Oscar F Sánchez
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN, 47907, USA
| | - Jean-Christophe Rochet
- Department of Medicinal Chemistry and Molecular Pharmacy, Purdue University, West Lafayette, IN, 47907; Purdue Institute of Integrated Neuroscience, Purdue University, West Lafayette, IN, 47907
| | | | - Chongli Yuan
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN, 47907, USA; Purdue Institute of Integrated Neuroscience, Purdue University, West Lafayette, IN, 47907.
| |
Collapse
|
14
|
Cahill MK, Collard M, Tse V, Reitman ME, Etchenique R, Kirst C, Poskanzer KE. Network-level encoding of local neurotransmitters in cortical astrocytes. Nature 2024; 629:146-153. [PMID: 38632406 PMCID: PMC11062919 DOI: 10.1038/s41586-024-07311-5] [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: 04/03/2023] [Accepted: 03/13/2024] [Indexed: 04/19/2024]
Abstract
Astrocytes, the most abundant non-neuronal cell type in the mammalian brain, are crucial circuit components that respond to and modulate neuronal activity through calcium (Ca2+) signalling1-7. Astrocyte Ca2+ activity is highly heterogeneous and occurs across multiple spatiotemporal scales-from fast, subcellular activity3,4 to slow, synchronized activity across connected astrocyte networks8-10-to influence many processes5,7,11. However, the inputs that drive astrocyte network dynamics remain unclear. Here we used ex vivo and in vivo two-photon astrocyte imaging while mimicking neuronal neurotransmitter inputs at multiple spatiotemporal scales. We find that brief, subcellular inputs of GABA and glutamate lead to widespread, long-lasting astrocyte Ca2+ responses beyond an individual stimulated cell. Further, we find that a key subset of Ca2+ activity-propagative activity-differentiates astrocyte network responses to these two main neurotransmitters, and may influence responses to future inputs. Together, our results demonstrate that local, transient neurotransmitter inputs are encoded by broad cortical astrocyte networks over a minutes-long time course, contributing to accumulating evidence that substantial astrocyte-neuron communication occurs across slow, network-level spatiotemporal scales12-14. These findings will enable future studies to investigate the link between specific astrocyte Ca2+ activity and specific functional outputs, which could build a consistent framework for astrocytic modulation of neuronal activity.
Collapse
Affiliation(s)
- Michelle K Cahill
- Department of Biochemistry & Biophysics, University of California, San Francisco, CA, USA
- Neuroscience Graduate Program, University of California, San Francisco, CA, USA
| | - Max Collard
- Department of Biochemistry & Biophysics, University of California, San Francisco, CA, USA
- Neuroscience Graduate Program, University of California, San Francisco, CA, USA
| | - Vincent Tse
- Department of Biochemistry & Biophysics, University of California, San Francisco, CA, USA
| | - Michael E Reitman
- Department of Biochemistry & Biophysics, University of California, San Francisco, CA, USA
- Neuroscience Graduate Program, University of California, San Francisco, CA, USA
| | - Roberto Etchenique
- Departamento de Química Inorgánica, Analítica y Química Física, INQUIMAE, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, CONICET, Buenos Aires, Argentina
| | - Christoph Kirst
- Neuroscience Graduate Program, University of California, San Francisco, CA, USA
- Department of Anatomy, University of California, San Francisco, CA, USA
- Kavli Institute for Fundamental Neuroscience, San Francisco, CA, USA
- Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Kira E Poskanzer
- Department of Biochemistry & Biophysics, University of California, San Francisco, CA, USA.
- Neuroscience Graduate Program, University of California, San Francisco, CA, USA.
- Kavli Institute for Fundamental Neuroscience, San Francisco, CA, USA.
| |
Collapse
|
15
|
Dharmasri PA, Levy AD, Blanpied TA. Differential nanoscale organization of excitatory synapses onto excitatory vs. inhibitory neurons. Proc Natl Acad Sci U S A 2024; 121:e2315379121. [PMID: 38625946 PMCID: PMC11047112 DOI: 10.1073/pnas.2315379121] [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: 09/18/2023] [Accepted: 03/14/2024] [Indexed: 04/18/2024] Open
Abstract
A key feature of excitatory synapses is the existence of subsynaptic protein nanoclusters (NCs) whose precise alignment across the cleft in a transsynaptic nanocolumn influences the strength of synaptic transmission. However, whether nanocolumn properties vary between excitatory synapses functioning in different cellular contexts is unknown. We used a combination of confocal and DNA-PAINT super-resolution microscopy to directly compare the organization of shared scaffold proteins at two important excitatory synapses-those forming onto excitatory principal neurons (Ex→Ex synapses) and those forming onto parvalbumin-expressing interneurons (Ex→PV synapses). As in Ex→Ex synapses, we find that in Ex→PV synapses, presynaptic Munc13-1 and postsynaptic PSD-95 both form NCs that demonstrate alignment, underscoring synaptic nanostructure and the transsynaptic nanocolumn as conserved organizational principles of excitatory synapses. Despite the general conservation of these features, we observed specific differences in the characteristics of pre- and postsynaptic Ex→PV nanostructure. Ex→PV synapses contained larger PSDs with fewer PSD-95 NCs when accounting for size than Ex→Ex synapses. Furthermore, the PSD-95 NCs were larger and denser. The identity of the postsynaptic cell was also represented in Munc13-1 organization, as Ex→PV synapses hosted larger Munc13-1 puncta that contained less dense but larger and more numerous Munc13-1 NCs. Moreover, we measured the spatial variability of transsynaptic alignment in these synapse types, revealing protein alignment in Ex→PV synapses over a distinct range of distances compared to Ex→Ex synapses. We conclude that while general principles of nanostructure and alignment are shared, cell-specific elements of nanodomain organization likely contribute to functional diversity of excitatory synapses.
Collapse
Affiliation(s)
- Poorna A. Dharmasri
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD21201
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD21201
- University of Maryland-Medicine Institute of Neuroscience Discovery, University of Maryland School of Medicine, Baltimore, MD21201
| | - Aaron D. Levy
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD21201
- University of Maryland-Medicine Institute of Neuroscience Discovery, University of Maryland School of Medicine, Baltimore, MD21201
| | - Thomas A. Blanpied
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD21201
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD21201
- University of Maryland-Medicine Institute of Neuroscience Discovery, University of Maryland School of Medicine, Baltimore, MD21201
| |
Collapse
|
16
|
Kellner V, Parker P, Mi X, Yu G, Saher G, Bergles DE. Conservation of neuron-astrocyte coordinated activity among sensory processing centers of the developing brain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.15.589519. [PMID: 38659917 PMCID: PMC11042386 DOI: 10.1101/2024.04.15.589519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Afferent neurons in developing sensory organs exhibit a prolonged period of burst firing prior to the onset of sensory experience. This intrinsically generated activity propagates from the periphery through central processing centers to promote the survival and physiological maturation of neurons and refine their synaptic connectivity. Recent studies in the auditory system indicate that these bursts of action potentials also trigger metabotropic glutamate receptor-mediated calcium increases within astrocytes that are spatially and temporally correlated with neuronal events; however, it is not known if this phenomenon occurs in other sensory modalities. Here we show using in vivo simultaneous imaging of neuronal and astrocyte calcium activity in awake mouse pups that waves of retinal ganglion cell activity induce spatially and temporally correlated waves of astrocyte activity in the superior colliculus that depend on metabotropic glutamate receptors mGluR5 and mGluR3. Astrocyte calcium transients reliably occurred with each neuronal wave, but peaked more than one second after neuronal events. Despite differences in the temporal features of spontaneous activity in auditory and visual processing regions, individual astrocytes exhibited similar overall calcium activity patterns, providing a conserved mechanism to synchronize neuronal and astrocyte maturation within discrete sensory domains.
Collapse
|
17
|
Caznok Silveira AC, Antunes ASLM, Athié MCP, da Silva BF, Ribeiro dos Santos JV, Canateli C, Fontoura MA, Pinto A, Pimentel-Silva LR, Avansini SH, de Carvalho M. Between neurons and networks: investigating mesoscale brain connectivity in neurological and psychiatric disorders. Front Neurosci 2024; 18:1340345. [PMID: 38445254 PMCID: PMC10912403 DOI: 10.3389/fnins.2024.1340345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 01/29/2024] [Indexed: 03/07/2024] Open
Abstract
The study of brain connectivity has been a cornerstone in understanding the complexities of neurological and psychiatric disorders. It has provided invaluable insights into the functional architecture of the brain and how it is perturbed in disorders. However, a persistent challenge has been achieving the proper spatial resolution, and developing computational algorithms to address biological questions at the multi-cellular level, a scale often referred to as the mesoscale. Historically, neuroimaging studies of brain connectivity have predominantly focused on the macroscale, providing insights into inter-regional brain connections but often falling short of resolving the intricacies of neural circuitry at the cellular or mesoscale level. This limitation has hindered our ability to fully comprehend the underlying mechanisms of neurological and psychiatric disorders and to develop targeted interventions. In light of this issue, our review manuscript seeks to bridge this critical gap by delving into the domain of mesoscale neuroimaging. We aim to provide a comprehensive overview of conditions affected by aberrant neural connections, image acquisition techniques, feature extraction, and data analysis methods that are specifically tailored to the mesoscale. We further delineate the potential of brain connectivity research to elucidate complex biological questions, with a particular focus on schizophrenia and epilepsy. This review encompasses topics such as dendritic spine quantification, single neuron morphology, and brain region connectivity. We aim to showcase the applicability and significance of mesoscale neuroimaging techniques in the field of neuroscience, highlighting their potential for gaining insights into the complexities of neurological and psychiatric disorders.
Collapse
Affiliation(s)
- Ana Clara Caznok Silveira
- National Laboratory of Biosciences, Brazilian Center for Research in Energy and Materials, Campinas, Brazil
- School of Electrical and Computer Engineering, University of Campinas, Campinas, Brazil
| | | | - Maria Carolina Pedro Athié
- National Laboratory of Biosciences, Brazilian Center for Research in Energy and Materials, Campinas, Brazil
| | - Bárbara Filomena da Silva
- National Laboratory of Biosciences, Brazilian Center for Research in Energy and Materials, Campinas, Brazil
| | | | - Camila Canateli
- National Laboratory of Biosciences, Brazilian Center for Research in Energy and Materials, Campinas, Brazil
| | - Marina Alves Fontoura
- National Laboratory of Biosciences, Brazilian Center for Research in Energy and Materials, Campinas, Brazil
| | - Allan Pinto
- Brazilian Synchrotron Light Laboratory, Brazilian Center for Research in Energy and Materials, Campinas, Brazil
| | | | - Simoni Helena Avansini
- National Laboratory of Biosciences, Brazilian Center for Research in Energy and Materials, Campinas, Brazil
| | - Murilo de Carvalho
- National Laboratory of Biosciences, Brazilian Center for Research in Energy and Materials, Campinas, Brazil
- Brazilian Synchrotron Light Laboratory, Brazilian Center for Research in Energy and Materials, Campinas, Brazil
| |
Collapse
|
18
|
Guhathakurta D, Petrušková A, Akdaş EY, Perelló-Amorós B, Frischknecht R, Anni D, Weiss EM, Walter M, Fejtová A. Hydroxynorketamine, but not ketamine, acts via α7 nicotinic acetylcholine receptor to control presynaptic function and gene expression. Transl Psychiatry 2024; 14:47. [PMID: 38253622 PMCID: PMC10803733 DOI: 10.1038/s41398-024-02744-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 12/21/2023] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
Ketamine is clinically used fast-acting antidepressant. Its metabolite hydroxynorketamine (HNK) shows a robust antidepressant effect in animal studies. It is unclear, how these chemically distinct compounds converge on similar neuronal effects. While KET acts mostly as N-methyl-d-aspartate receptor (NMDAR) antagonist, the molecular target of HNK remains enigmatic. Here, we show that KET and HNK converge on rapid inhibition of glutamate release by reducing the release competence of synaptic vesicles and induce nuclear translocation of pCREB that controls expression of neuroplasticity genes connected to KET- and HNK-mediated antidepressant action. Ro25-6981, a selective antagonist of GluN2B, mimics effect of KET indicating that GluN2B-containing NMDAR might mediate the presynaptic effect of KET. Selective antagonist of α7 nicotinic acetylcholine receptors (α7nAChRs) or genetic deletion of Chrna7, its pore-forming subunit, fully abolishes HNK-induced synaptic and nuclear regulations, but leaves KET-dependent cellular effects unaffected. Thus, KET or HNK-induced modulation of synaptic transmission and nuclear translocation of pCREB can be mediated by selective signaling via NMDAR or α7nAChRs, respectively. Due to the rapid metabolism of KET to HNK, it is conceivable that subsequent modulation of glutamatergic and cholinergic neurotransmission affects circuits in a cell-type-specific manner and contributes to the therapeutic potency of KET. This finding promotes further exploration of new combined medications for mood disorders.
Collapse
Affiliation(s)
- Debarpan Guhathakurta
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Aneta Petrušková
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- National Institute of Mental Health, Klecany, Czech Republic
- Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Enes Yağız Akdaş
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Bartomeu Perelló-Amorós
- Department of Biology, Animal Physiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Renato Frischknecht
- Department of Biology, Animal Physiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Daniela Anni
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Eva-Maria Weiss
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Martin Walter
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Jena, Jena, Germany
| | - Anna Fejtová
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
| |
Collapse
|
19
|
Zhao H, Xie J, Wu S, Zhao X, Sánchez OF, Min S, Rochet JC, Freeman JL, Yuan C. Elevated parkinsonism pathological markers in dopaminergic neurons with developmental exposure to atrazine. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 908:168307. [PMID: 37949145 PMCID: PMC10843769 DOI: 10.1016/j.scitotenv.2023.168307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 10/30/2023] [Accepted: 11/01/2023] [Indexed: 11/12/2023]
Abstract
Atrazine (ATZ) is one of the most used herbicides in the US and a known endocrine disruptor. ATZ is frequently detected in drinking water, especially in Midwestern regions of the United States, exceeding the EPA regulation of maximum contamination level (MCL) of 3 ppb. Epidemiology studies have suggested an association between ATZ exposure and neurodegeneration. Less, however, is known about the neurotoxic mechanism of ATZ, particularly for exposures at a developmental stage. Here, we exposed floor plate progenitors (FPPs) derived from human induced pluripotent stem cells (hiPSCs) to low concentrations of ATZ at 0.3 and 3 ppb for two days followed by differentiation into dopaminergic (DA) neurons in ATZ-free medium. We then examined the morphology, activity, pathological protein aggregation, and transcriptomic changes of differentiated DA neurons. We observed significant decrease in the complexity of neurite network, increase of neuronal activity, and elevated tau- and α-synuclein (aSyn) pathologies after ATZ exposure. The ATZ-induced neuronal changes observed here align with pathological characteristics in Parkinson's disease (PD). Transcriptomic analysis further corroborates our findings; and collectively provides a strong evidence base that low-concentration ATZ exposure during development can elicit increased risk of neurodegeneration.
Collapse
Affiliation(s)
- Han Zhao
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN 47907, United States of America
| | - Junkai Xie
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN 47907, United States of America
| | - Shichen Wu
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN 47907, United States of America
| | - Xihui Zhao
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, United States of America
| | - Oscar F Sánchez
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN 47907, United States of America
| | - Sehong Min
- Department of Medicinal Chemistry and Molecular Pharmacy, Purdue University, West Lafayette, IN 47907, United States of America
| | - Jean-Christophe Rochet
- Department of Medicinal Chemistry and Molecular Pharmacy, Purdue University, West Lafayette, IN 47907, United States of America; Purdue Institute of Integrated Neuroscience, Purdue University, West Lafayette, IN 47907, United States of America
| | - Jennifer L Freeman
- School of Health Sciences, Purdue University, West Lafayette, IN 47907, United States of America; Purdue Institute of Integrated Neuroscience, Purdue University, West Lafayette, IN 47907, United States of America
| | - Chongli Yuan
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN 47907, United States of America; Purdue Institute of Integrated Neuroscience, Purdue University, West Lafayette, IN 47907, United States of America; Purdue Center of Cancer Research, West Lafayette, IN 47907, United States of America.
| |
Collapse
|
20
|
Culibrk RA, Ebbert KA, Yeisley DJ, Chen R, Qureshi FA, Hahn J, Hahn MS. Impact of Suramin on Key Pathological Features of Sporadic Alzheimer's Disease-Derived Forebrain Neurons. J Alzheimers Dis 2024; 98:301-318. [PMID: 38427475 DOI: 10.3233/jad-230600] [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: 03/03/2024]
Abstract
Background Alzheimer's disease (AD) is characterized by disrupted proteostasis and macroautophagy (hereafter "autophagy"). The pharmacological agent suramin has known autophagy modulation properties with potential efficacy in mitigating AD neuronal pathology. Objective In the present work, we investigate the impact of forebrain neuron exposure to suramin on the Akt/mTOR signaling pathway, a major regulator of autophagy, in comparison with rapamycin and chloroquine. We further investigate the effect of suramin on several AD-related biomarkers in sporadic AD (sAD)-derived forebrain neurons. Methods Neurons differentiated from ReNcell neural progenitors were used to assess the impact of suramin on the Akt/mTOR signaling pathway relative to the autophagy inducer rapamycin and autophagy inhibitor chloroquine. Mature forebrain neurons were differentiated from induced pluripotent stem cells (iPSCs) sourced from a late-onset sAD patient and treated with 100μM suramin for 72 h, followed by assessments for amyloid-β, phosphorylated tau, oxidative/nitrosative stress, and synaptic puncta density. Results Suramin treatment of sAD-derived neurons partially ameliorated the increased p-Tau(S199)/Tau ratio, and fully remediated the increased glutathione to oxidized nitric oxide ratio, observed in untreated sAD-derived neurons relative to healthy controls. These positive results may be due in part to the distinct increases in Akt/mTOR pathway mediator p-p70S6K noted with suramin treatment of both ReNcell-derived and iPSC-derived neurons. Longer term neuronal markers, such as synaptic puncta density, were unaffected by suramin treatment. Conclusions These findings provide initial evidence supporting the potential of suramin to reduce the degree of dysregulation in sAD-derived forebrain neurons in part via the modulation of autophagy.
Collapse
Affiliation(s)
- Robert A Culibrk
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Katherine A Ebbert
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Daniel J Yeisley
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Rui Chen
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Fatir A Qureshi
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Juergen Hahn
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Mariah S Hahn
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, USA
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA
| |
Collapse
|
21
|
Cahill MK, Collard M, Tse V, Reitman ME, Etchenique R, Kirst C, Poskanzer KE. Network-level encoding of local neurotransmitters in cortical astrocytes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.01.568932. [PMID: 38106119 PMCID: PMC10723263 DOI: 10.1101/2023.12.01.568932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Astrocytes-the most abundant non-neuronal cell type in the mammalian brain-are crucial circuit components that respond to and modulate neuronal activity via calcium (Ca 2+ ) signaling 1-8 . Astrocyte Ca 2+ activity is highly heterogeneous and occurs across multiple spatiotemporal scales: from fast, subcellular activity 3,4 to slow, synchronized activity that travels across connected astrocyte networks 9-11 . Furthermore, astrocyte network activity has been shown to influence a wide range of processes 5,8,12 . While astrocyte network activity has important implications for neuronal circuit function, the inputs that drive astrocyte network dynamics remain unclear. Here we used ex vivo and in vivo two-photon Ca 2+ imaging of astrocytes while mimicking neuronal neurotransmitter inputs at multiple spatiotemporal scales. We find that brief, subcellular inputs of GABA and glutamate lead to widespread, long-lasting astrocyte Ca 2+ responses beyond an individual stimulated cell. Further, we find that a key subset of Ca 2+ activity-propagative events-differentiates astrocyte network responses to these two major neurotransmitters, and gates responses to future inputs. Together, our results demonstrate that local, transient neurotransmitter inputs are encoded by broad cortical astrocyte networks over the course of minutes, contributing to accumulating evidence across multiple model organisms that significant astrocyte-neuron communication occurs across slow, network-level spatiotemporal scales 13-15 . We anticipate that this study will be a starting point for future studies investigating the link between specific astrocyte Ca 2+ activity and specific astrocyte functional outputs, which could build a consistent framework for astrocytic modulation of neuronal activity.
Collapse
|
22
|
Coulon A, Siedlecki-Wullich D, Najdek C, Gelle C, Ayral AM, Demiautte F, Lambert E, Vandeputte A, Brodin P, Mendes T, Lambert JC, Kilinc D, Dumont J, Chapuis J. High-Content Screening of Synaptic Density Modulators in Primary Neuronal Cultures. Curr Protoc 2023; 3:e904. [PMID: 37882787 DOI: 10.1002/cpz1.904] [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: 10/27/2023]
Abstract
The synapse, which represents the structural and functional basis of neuronal communication, is one of the first elements affected in several neurodegenerative diseases. To better understand the potential role of gene expression in synapse loss, we developed an original high-content screening (HCS) model capable of quantitatively assessing the impact of gene silencing on synaptic density. Our approach is based on a model of primary neuronal cultures (PNCs) from the neonatal rat hippocampus, whose mature synapses are visualized by the relative localization of the presynaptic protein Synaptophysin with the postsynaptic protein Homer1. The heterogeneity of PNCs and the small sizes of the synaptic structures pose technical challenges associated with the level of automation necessary for HCS studies. We overcame these technical challenges, automated the processes of image analysis and data analysis, and carried out tests under real-world conditions to demonstrate the robustness of the model developed. In this article, we describe the screening of a custom library of 198 shRNAs in PNCs in the 384-well plate format. © 2023 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Culture of primary hippocampal rat neurons in 384-well plates Basic Protocol 2: Lentiviral shRNA transduction of primary neuronal culture in 384-well plates Basic Protocol 3: Immunostaining of the neuronal network and synaptic markers in 384-well plates Basic Protocol 4: Image acquisition using a high-throughput reader Basic Protocol 5: Image segmentation and analysis Basic Protocol 6: Synaptic density analysis.
Collapse
Affiliation(s)
- Audrey Coulon
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, Lille, France
| | - Dolores Siedlecki-Wullich
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, Lille, France
| | - Chloé Najdek
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, Lille, France
| | - Carla Gelle
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, Lille, France
| | - Anne-Marie Ayral
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, Lille, France
| | - Florie Demiautte
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, Lille, France
| | - Erwan Lambert
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, Lille, France
| | - Alexandre Vandeputte
- Université de Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, Center for Infection and Immunity of Lille (CIIL), Lille, France
| | - Priscille Brodin
- Université de Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, Center for Infection and Immunity of Lille (CIIL), Lille, France
| | - Tiago Mendes
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, Lille, France
| | - Jean-Charles Lambert
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, Lille, France
| | - Devrim Kilinc
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, Lille, France
| | - Julie Dumont
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, Lille, France
| | - Julien Chapuis
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, Lille, France
| |
Collapse
|
23
|
Dharmasri PA, Levy AD, Blanpied TA. Differential nanoscale organization of excitatory synapses onto excitatory vs inhibitory neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.06.556279. [PMID: 37732271 PMCID: PMC10508768 DOI: 10.1101/2023.09.06.556279] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
A key feature of excitatory synapses is the existence of subsynaptic protein nanoclusters whose precise alignment across the cleft in a trans-synaptic nanocolumn influences the strength of synaptic transmission. However, whether nanocolumn properties vary between excitatory synapses functioning in different cellular contexts is unknown. We used a combination of confocal and DNA-PAINT super-resolution microscopy to directly compare the organization of shared scaffold proteins at two important excitatory synapses - those forming onto excitatory principal neurons (Ex→Ex synapses) and those forming onto parvalbumin-expressing interneurons (Ex→PV synapses). As in Ex→Ex synapses, we find that in Ex→PV synapses presynaptic Munc13-1 and postsynaptic PSD-95 both form nanoclusters that demonstrate alignment, underscoring synaptic nanostructure and the trans-synaptic nanocolumn as conserved organizational principles of excitatory synapses. Despite the general conservation of these features, we observed specific differences in the characteristics of pre- and postsynaptic Ex→PV nanostructure. Ex→PV synapses contained larger PSDs with fewer PSD-95 NCs when accounting for size than Ex→Ex synapses. Furthermore, the PSD-95 NCs were larger and denser. The identity of the postsynaptic cell also had a retrograde impact on Munc13-1 organization, as Ex→PV synapses hosted larger Munc13-1 puncta that contained less dense but larger and more numerous Munc13-1 NCs. Moreover, we measured the spatial variability of transsynaptic alignment in these synapse types, revealing protein alignment in Ex→PV synapses over a distinct range of distances compared to Ex→Ex synapses. We conclude that while general principles of nanostructure and alignment are shared, cell-specific elements of nanodomain organization likely contribute to functional diversity of excitatory synapses. Understanding the rules of synapse nanodomain assembly, which themselves are cell-type specific, will be essential for illuminating brain network dynamics.
Collapse
Affiliation(s)
- Poorna A Dharmasri
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland 21201
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland 21201
- University of Maryland Medicine Institute of Neuroscience Discovery, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Aaron D Levy
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland 21201
- University of Maryland Medicine Institute of Neuroscience Discovery, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Thomas A Blanpied
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland 21201
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland 21201
- University of Maryland Medicine Institute of Neuroscience Discovery, University of Maryland School of Medicine, Baltimore, Maryland 21201
| |
Collapse
|
24
|
Szigeti K, Ihnatovych I, Rosas N, Dorn RP, Notari E, Cortes Gomez E, He M, Maly I, Prasad S, Nimmer E, Heo Y, Fuchsova B, Bennett DA, Hofmann WA, Pralle A, Bae Y, Wang J. Neuronal actin cytoskeleton gain of function in the human brain. EBioMedicine 2023; 95:104725. [PMID: 37517100 PMCID: PMC10404607 DOI: 10.1016/j.ebiom.2023.104725] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 06/21/2023] [Accepted: 07/07/2023] [Indexed: 08/01/2023] Open
Abstract
BACKGROUND While advancements in imaging techniques have led to major strides in deciphering the human brain, successful interventions are elusive and represent some of the most persistent translational gaps in medicine. Human restricted CHRFAM7A has been associated with neuropsychiatric disorders. METHODS The physiological role of CHRFAM7A in human brain is explored using multiomics approach on 600 post mortem human brain tissue samples. The emerging pathways and mechanistic hypotheses are tested and validated in an isogenic hiPSC model of CHRFAM7A knock-in medial ganglionic eminence progenitors and neurons. FINDINGS CHRFAM7A is identified as a modulator of intracellular calcium dynamics and an upstream regulator of Rac1. Rac1 activation re-designs the actin cytoskeleton leading to dynamic actin driven remodeling of membrane protrusion and a switch from filopodia to lamellipodia. The reinforced cytoskeleton leads to an advantage to tolerate stiffer mechanical properties of the extracellular environment. INTERPRETATION CHRFAM7A modifies the actin cytoskeleton to a more dynamic and stiffness resistant state in an α7nAChR dependent manner. CHRFAM7A may facilitate neuronal adaptation to changes in the brain environment in physiological and pathological conditions contributing to risk or recovery. Understanding how CHRFAM7A affects human brain requires human studies in the areas of memory formation and erasure, cognitive reserve, and neuronal plasticity. FUNDING This work is supported in part by the Community Foundation for Greater Buffalo (Kinga Szigeti). Also, in part by the International Society for Neurochemistry (ISN) and The Company of Biologists (Nicolas Rosas). ROSMAP is supported by NIA grants P30AG10161, P30AG72975, R01AG15819, R01AG17917. U01AG46152, and U01AG61356.
Collapse
Affiliation(s)
- Kinga Szigeti
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA.
| | - Ivanna Ihnatovych
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Nicolás Rosas
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA; Instituto de Investigaciones Biotecnológicas, Escuela de Bio y Nanotecnologías (EByN), Universidad Nacional de San Martín (UNSAM) - Consejo Nacional de, Investigaciones Científicas y Técnicas (CONICET), San Martín, Buenos Aires, Argentina
| | - Ryu P Dorn
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Emily Notari
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | | | - Muye He
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Ivan Maly
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Shreyas Prasad
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Erik Nimmer
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Yuna Heo
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Beata Fuchsova
- Instituto de Investigaciones Biotecnológicas, Escuela de Bio y Nanotecnologías (EByN), Universidad Nacional de San Martín (UNSAM) - Consejo Nacional de, Investigaciones Científicas y Técnicas (CONICET), San Martín, Buenos Aires, Argentina
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Wilma A Hofmann
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Arnd Pralle
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Yongho Bae
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Jianmin Wang
- Roswell Park Comprehensive Cancer Center, 665 Elm St, Buffalo, NY 14203, USA
| |
Collapse
|
25
|
Xie J, Wu S, Szadowski H, Min S, Yang Y, Bowman AB, Rochet JC, Freeman JL, Yuan C. Developmental Pb exposure increases AD risk via altered intracellular Ca 2+ homeostasis in hiPSC-derived cortical neurons. J Biol Chem 2023; 299:105023. [PMID: 37423307 PMCID: PMC10413359 DOI: 10.1016/j.jbc.2023.105023] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 06/21/2023] [Accepted: 06/22/2023] [Indexed: 07/11/2023] Open
Abstract
Exposure to environmental chemicals such as lead (Pb) during vulnerable developmental periods can result in adverse health outcomes later in life. Human cohort studies have demonstrated associations between developmental Pb exposure and Alzheimer's disease (AD) onset in later life which were further corroborated by findings from animal studies. The molecular pathway linking developmental Pb exposure and increased AD risk, however, remains elusive. In this work, we used human iPSC-derived cortical neurons as a model system to study the effects of Pb exposure on AD-like pathogenesis in human cortical neurons. We exposed neural progenitor cells derived from human iPSC to 0, 15, and 50 ppb Pb for 48 h, removed Pb-containing medium, and further differentiated them into cortical neurons. Immunofluorescence, Western blotting, RNA-sequencing, ELISA, and FRET reporter cell lines were used to determine changes in AD-like pathogenesis in differentiated cortical neurons. Exposing neural progenitor cells to low-dose Pb, mimicking a developmental exposure, can result in altered neurite morphology. Differentiated neurons exhibit altered calcium homeostasis, synaptic plasticity, and epigenetic landscape along with elevated AD-like pathogenesis markers, including phosphorylated tau, tau aggregates, and Aβ42/40. Collectively, our findings provide an evidence base for Ca dysregulation caused by developmental Pb exposure as a plausible molecular mechanism accounting for increased AD risk in populations with developmental Pb exposure.
Collapse
Affiliation(s)
- Junkai Xie
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, Indiana, USA
| | - Shichen Wu
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, Indiana, USA
| | - Hailey Szadowski
- Agriculture and Biological Engineering, Purdue University, West Lafayette, Indiana, USA
| | - Sehong Min
- Department of Medicinal Chemistry and Molecular Pharmacy, Purdue University, West Lafayette, Indiana, USA
| | - Yang Yang
- Department of Medicinal Chemistry and Molecular Pharmacy, Purdue University, West Lafayette, Indiana, USA; Purdue Institute of Integrated Neuroscience, Purdue University, West Lafayette, Indiana, USA
| | - Aaron B Bowman
- Purdue Institute of Integrated Neuroscience, Purdue University, West Lafayette, Indiana, USA; School of Health Sciences, Purdue University, West Lafayette, Indiana, USA
| | - Jean-Christophe Rochet
- Department of Medicinal Chemistry and Molecular Pharmacy, Purdue University, West Lafayette, Indiana, USA; Purdue Institute of Integrated Neuroscience, Purdue University, West Lafayette, Indiana, USA
| | - Jennifer L Freeman
- Purdue Institute of Integrated Neuroscience, Purdue University, West Lafayette, Indiana, USA; School of Health Sciences, Purdue University, West Lafayette, Indiana, USA; Purdue Center of Cancer Research, Purdue University, West Lafayette, Indiana, USA
| | - Chongli Yuan
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, Indiana, USA; Purdue Institute of Integrated Neuroscience, Purdue University, West Lafayette, Indiana, USA; Purdue Center of Cancer Research, Purdue University, West Lafayette, Indiana, USA.
| |
Collapse
|
26
|
Berryer MH, Rizki G, Nathanson A, Klein JA, Trendafilova D, Susco SG, Lam D, Messana A, Holton KM, Karhohs KW, Cimini BA, Pfaff K, Carpenter AE, Rubin LL, Barrett LE. High-content synaptic phenotyping in human cellular models reveals a role for BET proteins in synapse assembly. eLife 2023; 12:80168. [PMID: 37083703 PMCID: PMC10121225 DOI: 10.7554/elife.80168] [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: 05/11/2022] [Accepted: 04/10/2023] [Indexed: 04/22/2023] Open
Abstract
Resolving fundamental molecular and functional processes underlying human synaptic development is crucial for understanding normal brain function as well as dysfunction in disease. Based upon increasing evidence of species-divergent features of brain cell types, coupled with emerging studies of complex human disease genetics, we developed the first automated and quantitative high-content synaptic phenotyping platform using human neurons and astrocytes. To establish the robustness of our platform, we screened the effects of 376 small molecules on presynaptic density, neurite outgrowth, and cell viability, validating six small molecules that specifically enhanced human presynaptic density in vitro. Astrocytes were essential for mediating the effects of all six small molecules, underscoring the relevance of non-cell-autonomous factors in synapse assembly and their importance in synaptic screening applications. Bromodomain and extraterminal (BET) inhibitors emerged as the most prominent hit class and global transcriptional analyses using multiple BET inhibitors confirmed upregulation of synaptic gene expression. Through these analyses, we demonstrate the robustness of our automated screening platform for identifying potent synaptic modulators, which can be further leveraged for scaled analyses of human synaptic mechanisms and drug discovery efforts.
Collapse
Affiliation(s)
- Martin H Berryer
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, United States
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, United States
| | - Gizem Rizki
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, United States
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, United States
| | - Anna Nathanson
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, United States
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, United States
| | - Jenny A Klein
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, United States
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, United States
| | - Darina Trendafilova
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, United States
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, United States
| | - Sara G Susco
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, United States
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, United States
| | - Daisy Lam
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, United States
| | - Angelica Messana
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, United States
| | - Kristina M Holton
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, United States
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, United States
| | - Kyle W Karhohs
- Imaging Platform, Broad Institute of MIT and Harvard, Cambridge, United States
| | - Beth A Cimini
- Imaging Platform, Broad Institute of MIT and Harvard, Cambridge, United States
| | - Kathleen Pfaff
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, United States
| | - Anne E Carpenter
- Imaging Platform, Broad Institute of MIT and Harvard, Cambridge, United States
| | - Lee L Rubin
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, United States
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, United States
| | - Lindy E Barrett
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, United States
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, United States
| |
Collapse
|
27
|
Barnes CC, Yee KT, Vetter DE. Conditional Ablation of Glucocorticoid and Mineralocorticoid Receptors from Cochlear Supporting Cells Reveals Their Differential Roles for Hearing Sensitivity and Dynamics of Recovery from Noise-Induced Hearing Loss. Int J Mol Sci 2023; 24:3320. [PMID: 36834731 PMCID: PMC9961551 DOI: 10.3390/ijms24043320] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 02/01/2023] [Accepted: 02/03/2023] [Indexed: 02/11/2023] Open
Abstract
Endogenous glucocorticoids (GC) are known to modulate basic elements of cochlear physiology. These include both noise-induced injury and circadian rhythms. While GC signaling in the cochlea can directly influence auditory transduction via actions on hair cells and spiral ganglion neurons, evidence also indicates that GC signaling exerts effects via tissue homeostatic processes that can include effects on cochlear immunomodulation. GCs act at both the glucocorticoid receptor (GR) and the mineralocorticoid receptor (MR). Most cell types in the cochlea express both receptors sensitive to GCs. The GR is associated with acquired sensorineural hearing loss (SNHL) through its effects on both gene expression and immunomodulatory programs. The MR has been associated with age-related hearing loss through dysfunction of ionic homeostatic balance. Cochlear supporting cells maintain local homeostatic requirements, are sensitive to perturbation, and participate in inflammatory signaling. Here, we have used conditional gene manipulation techniques to target Nr3c1 (GR) or Nr3c2 (MR) for tamoxifen-induced gene ablation in Sox9-expressing cochlear supporting cells of adult mice to investigate whether either of the receptors sensitive to GCs plays a role in protecting against (or exacerbating) noise-induced cochlear damage. We have selected mild intensity noise exposure to examine the role of these receptors related to more commonly experienced noise levels. Our results reveal distinct roles of these GC receptors for both basal auditory thresholds prior to noise exposure and during recovery from mild noise exposure. Prior to noise exposure, auditory brainstem responses (ABRs) were measured in mice carrying the floxed allele of interest and the Cre recombinase transgene, but not receiving tamoxifen injections (defined as control (no tamoxifen treatment), versus conditional knockout (cKO) mice, defined as mice having received tamoxifen injections. Results revealed hypersensitive thresholds to mid- to low-frequencies after tamoxifen-induced GR ablation from Sox9-expressing cochlear supporting cells compared to control (no tamoxifen) mice. GR ablation from Sox9-expressing cochlear supporting cells resulted in a permanent threshold shift in mid-basal cochlear frequency regions after mild noise exposure that produced only a temporary threshold shift in both control (no tamoxifen) f/fGR:Sox9iCre+ and heterozygous f/+GR:Sox9iCre+ tamoxifen-treated mice. A similar comparison of basal ABRs measured in control (no tamoxifen) and tamoxifen-treated, floxed MR mice prior to noise exposure indicated no difference in baseline thresholds. After mild noise exposure, MR ablation was initially associated with a complete threshold recovery at 22.6 kHz by 3 days post-noise. Threshold continued to shift to higher sensitivity over time such that by 30 days post-noise exposure the 22.6 kHz ABR threshold was 10 dB more sensitive than baseline. Further, MR ablation produced a temporary reduction in peak 1 neural amplitude one day post-noise. While supporting cell GR ablation trended towards reducing numbers of ribbon synapses, MR ablation reduced ribbon synapse counts but did not exacerbate noise-induced damage including synapse loss at the experimental endpoint. GR ablation from the targeted supporting cells increased the basal resting number of Iba1-positive (innate) immune cells (no noise exposure) and decreased the number of Iba1-positive cells seven days following noise exposure. MR ablation did not alter innate immune cell numbers at seven days post-noise exposure. Taken together, these findings support differential roles of cochlear supporting cell MR and GR expression at basal, resting conditions and especially during recovery from noise exposure.
Collapse
Affiliation(s)
- Charles C. Barnes
- Graduate Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Kathleen T. Yee
- Department of Otolaryngology–Head and Neck Surgery, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Douglas E. Vetter
- Graduate Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Department of Otolaryngology–Head and Neck Surgery, University of Mississippi Medical Center, Jackson, MS 39216, USA
| |
Collapse
|
28
|
Tjahjono N, Jin Y, Hsu A, Roukes M, Tian L. Letting the little light of mind shine: Advances and future directions in neurochemical detection. Neurosci Res 2022; 179:65-78. [PMID: 34861294 PMCID: PMC9508992 DOI: 10.1016/j.neures.2021.11.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 11/29/2021] [Indexed: 12/12/2022]
Abstract
Synaptic transmission via neurochemical release is the fundamental process that integrates and relays encoded information in the brain to regulate physiological function, cognition, and emotion. To unravel the biochemical, biophysical, and computational mechanisms of signal processing, one needs to precisely measure the neurochemical release dynamics with molecular and cell-type specificity and high resolution. Here we reviewed the development of analytical, electrochemical, and fluorescence imaging approaches to detect neurotransmitter and neuromodulator release. We discussed the advantages and practicality in implementation of each technology for ease-of-use, flexibility for multimodal studies, and challenges for future optimization. We hope this review will provide a versatile guide for tool engineering and applications for recording neurochemical release.
Collapse
Affiliation(s)
- Nikki Tjahjono
- Biomedical Engineering Graduate Group, University of California, Davis, Davis, CA, 95616, USA
| | - Yihan Jin
- Neuroscience Graduate Group, University of California, Davis, Davis, CA, 95618, USA
| | - Alice Hsu
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Michael Roukes
- Department of Physics, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Lin Tian
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Davis, CA, 95616, USA.
| |
Collapse
|
29
|
Guhathakurta D, Akdaş EY, Fejtová A, Weiss EM. Development and Application of Automatized Routines for Optical Analysis of Synaptic Activity Evoked by Chemical and Electrical Stimulation. FRONTIERS IN BIOINFORMATICS 2022; 2:814081. [PMID: 36304276 PMCID: PMC9580924 DOI: 10.3389/fbinf.2022.814081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 01/24/2022] [Indexed: 11/24/2022] Open
Abstract
The recent development of cellular imaging techniques and the application of genetically encoded sensors of neuronal activity led to significant methodological progress in neurobiological studies. These methods often result in complex and large data sets consisting of image stacks or sets of multichannel fluorescent images. The detection of synapses, visualized by fluorescence labeling, is one major challenge in the analysis of these datasets, due to variations in synapse shape, size, and fluorescence intensity across the images. For their detection, most labs use manual or semi-manual techniques that are time-consuming and error-prone. We developed SynEdgeWs, a MATLAB-based segmentation algorithm that combines the application of an edge filter, morphological operators, and marker-controlled watershed segmentation. SynEdgeWs does not need training data and works with low user intervention. It was superior to methods based on cutoff thresholds and local maximum guided approaches in a realistic set of data. We implemented SynEdgeWs in two automatized routines that allow accurate, direct, and unbiased identification of fluorescently labeled synaptic puncta and their consecutive analysis. SynEval routine enables the analysis of three-channel images, and ImgSegRout routine processes image stacks. We tested the feasibility of ImgSegRout on a realistic live-cell imaging data set from experiments designed to monitor neurotransmitter release using synaptic phluorins. Finally, we applied SynEval to compare synaptic vesicle recycling evoked by electrical field stimulation and chemical depolarization in dissociated cortical cultures. Our data indicate that while the proportion of active synapses does not differ between stimulation modes, significantly more vesicles are mobilized upon chemical depolarization.
Collapse
Affiliation(s)
| | | | - Anna Fejtová
- *Correspondence: Anna Fejtová, ; Eva-Maria Weiss,
| | | |
Collapse
|
30
|
Patel PA, Hegert JV, Cristian I, Kerr A, LaConte LEW, Fox MA, Srivastava S, Mukherjee K. Complete loss of the X-linked gene CASK causes severe cerebellar degeneration. J Med Genet 2022; 59:1044-1057. [PMID: 35149592 DOI: 10.1136/jmedgenet-2021-108115] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 01/13/2022] [Indexed: 01/19/2023]
Abstract
BACKGROUND Heterozygous loss of X-linked genes like CASK and MeCP2 (Rett syndrome) causes developmental delay in girls, while in boys, loss of the only allele of these genes leads to epileptic encephalopathy. The mechanism for these disorders remains unknown. CASK-linked cerebellar hypoplasia is presumed to result from defects in Tbr1-reelin-mediated neuronal migration. METHOD Here we report clinical and histopathological analyses of a deceased 2-month-old boy with a CASK-null mutation. We next generated a mouse line where CASK is completely deleted (hemizygous and homozygous) from postmigratory neurons in the cerebellum. RESULT The CASK-null human brain was smaller in size but exhibited normal lamination without defective neuronal differentiation, migration or axonal guidance. The hypoplastic cerebellum instead displayed astrogliosis and microgliosis, which are markers for neuronal loss. We therefore hypothesise that CASK loss-induced cerebellar hypoplasia is the result of early neurodegeneration. Data from the murine model confirmed that in CASK loss, a small cerebellum results from postdevelopmental degeneration of cerebellar granule neurons. Furthermore, at least in the cerebellum, functional loss from CASK deletion is secondary to degeneration of granule cells and not due to an acute molecular functional loss of CASK. Intriguingly, female mice with heterozygous deletion of CASK in the cerebellum do not display neurodegeneration. CONCLUSION We suggest that X-linked neurodevelopmental disorders like CASK mutation and Rett syndrome are pathologically neurodegenerative; random X-chromosome inactivation in heterozygous mutant girls, however, results in 50% of cells expressing the functional gene, resulting in a non-progressive pathology, whereas complete loss of the only allele in boys leads to unconstrained degeneration and encephalopathy.
Collapse
Affiliation(s)
- Paras A Patel
- Fralin Biomedical Research Institute at VTC, Roanoke, Virginia, USA
| | - Julia V Hegert
- Department of Pathology, Orlando Health, Orlando, Florida, USA
| | | | - Alicia Kerr
- Fralin Biomedical Research Institute at VTC, Roanoke, Virginia, USA
| | | | - Michael A Fox
- Fralin Biomedical Research Institute at VTC, Roanoke, Virginia, USA.,School of Neuroscience, Blacksburg, Virginia, USA
| | - Sarika Srivastava
- Fralin Biomedical Research Institute at VTC, Roanoke, Virginia, USA.,Department of Internal Medicine, Virginia Tech Carilion School of Medicine, Roanoke, Virginia, USA
| | - Konark Mukherjee
- Fralin Biomedical Research Institute at VTC, Roanoke, Virginia, USA .,Department of Psychiatry, Virginia Tech Carilion School of Medicine, Roanoke, Virginia, USA
| |
Collapse
|
31
|
Ali MF, Latimer AJ, Wang Y, Hogenmiller L, Fontenas L, Isabella AJ, Moens CB, Yu G, Kucenas S. Met is required for oligodendrocyte progenitor cell migration in Danio rerio. G3 (BETHESDA, MD.) 2021; 11:jkab265. [PMID: 34568921 PMCID: PMC8473979 DOI: 10.1093/g3journal/jkab265] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 07/22/2021] [Indexed: 11/13/2022]
Abstract
During vertebrate central nervous system development, most oligodendrocyte progenitor cells (OPCs) are specified in the ventral spinal cord and must migrate throughout the neural tube until they become evenly distributed, occupying non-overlapping domains. While this process of developmental OPC migration is well characterized, the nature of the molecular mediators that govern it remain largely unknown. Here, using zebrafish as a model, we demonstrate that Met signaling is required for initial developmental migration of OPCs, and, using cell-specific knock-down of Met signaling, show that Met acts cell-autonomously in OPCs. Taken together, these findings demonstrate in vivo, the role of Met signaling in OPC migration and provide new insight into how OPC migration is regulated during development.
Collapse
Affiliation(s)
- Maria F Ali
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
| | - Andrew J Latimer
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
| | - Yinxue Wang
- Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA 22203, USA
| | - Leah Hogenmiller
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
| | - Laura Fontenas
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
| | - Adam J Isabella
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Cecilia B Moens
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Guoqiang Yu
- Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA 22203, USA
| | - Sarah Kucenas
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
| |
Collapse
|
32
|
Mersman B, Zaidi W, Syed NI, Xu F. Taurine Promotes Neurite Outgrowth and Synapse Development of Both Vertebrate and Invertebrate Central Neurons. Front Synaptic Neurosci 2020; 12:29. [PMID: 32792935 PMCID: PMC7387692 DOI: 10.3389/fnsyn.2020.00029] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 06/24/2020] [Indexed: 12/13/2022] Open
Abstract
Taurine is a sulfur-containing amino acid that is widely expressed throughout the human brain, heart, retina, and muscle tissues. Taurine deficiency is associated with cardiomyopathy, renal dysfunction, abnormalities of the developing nervous system, and epilepsy which suggests a role specific to excitable tissues. Like vertebrates, invertebrates maintain high levels of taurine during embryonic and larval development, which decline during aging, indicating a potential developmental role. Notwithstanding its extensive presence throughout, taurine’s precise role/s during early brain development, function, and repair remains largely unknown in both vertebrate and invertebrate. Here, we investigated whether taurine affects neurite outgrowth, synapse formation, and synaptic transmission between postnatal day 0 rat cortical neurons in vitro, whereas its synaptogenic role was tested more directly using the Lymnaea soma-soma synapse model. We provide direct evidence that when applied at physiological concentrations, taurine exerts a significant neurotrophic effect on neuritic outgrowth and thickness of neurites as well as the expression of synaptic puncta as revealed by immunostaining of presynaptic synaptophysin and postsynaptic PSD95 proteins in rat cortical neurons, indicating direct involvement in synapse development. To demonstrate taurine’s direct effects on neurons in the absence of glia and other confounding factors, we next exploited individually identified pre- and postsynaptic neurons from the mollusk Lymnaea stagnalis. We found that taurine increased both the incidence of synapse formation (percent of cells that form synapses) and the efficacy of synaptic transmission between the paired neurons. This effect was comparable, but not additive, to Lymnaea trophic factor-induced synaptogenesis. This study thus provides direct morphological and functional evidence that taurine plays an important role in neurite outgrowth, synaptogenesis, and synaptic transmission during the early stages of brain development and that this role is conserved across both vertebrate and invertebrate species.
Collapse
Affiliation(s)
- Brittany Mersman
- Department of Biology, College of Arts and Sciences, Saint Louis University, St. Louis, MO, United States.,Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University, St. Louis, MO, United States
| | - Wali Zaidi
- Department of Cell Biology and Anatomy, Hotchkiss Brain Institute and Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Naweed I Syed
- Department of Cell Biology and Anatomy, Hotchkiss Brain Institute and Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Fenglian Xu
- Department of Biology, College of Arts and Sciences, Saint Louis University, St. Louis, MO, United States.,Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University, St. Louis, MO, United States
| |
Collapse
|