1
|
Ganglberger M, Koschak A. Exploring the potential for gene therapy in Cav1.4-related retinal channelopathies. Channels (Austin) 2025; 19:2480089. [PMID: 40129245 PMCID: PMC11938310 DOI: 10.1080/19336950.2025.2480089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/07/2025] [Accepted: 03/11/2025] [Indexed: 03/26/2025] Open
Abstract
The visual process begins with photon detection in photoreceptor outer segments within the retina, which processes light signals before transmission to the thalamus and visual cortex. Cav1.4 L-type calcium channels play a crucial role in this process, and dysfunction of these channels due to pathogenic variants in corresponding genes leads to specific manifestations in visual impairments. This review explores the journey from basic research on Cav1.4 L-type calcium channel complexes in retinal physiology and pathophysiology to their potential as gene therapy targets. Moreover, we provide a concise overview of key findings from studies using different animal models to investigate retinal diseases. It will critically examine the constraints these models present when attempting to elucidate retinal channelopathies. Additionally, the paper will explore potential strategies for addressing Cav1.4 channel dysfunction and discuss the current challenges facing gene therapy approaches in this area of research.
Collapse
Affiliation(s)
- Matthias Ganglberger
- Pharmacology and Toxicology, Institute of Pharmacy, University of Innsbruck, Innsbruck, Austria
| | - Alexandra Koschak
- Pharmacology and Toxicology, Institute of Pharmacy, University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
2
|
Zhao K, Zhang L, Lei M, Jin Z, Du T, Zhang H, Sheng Y, Hu Z, Wang S, Ma C. A specific negatively charged sequence confers intramolecular regulation on Munc13-1 function in synaptic exocytosis. Proc Natl Acad Sci U S A 2025; 122:e2508915122. [PMID: 40489622 DOI: 10.1073/pnas.2508915122] [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: 04/15/2025] [Accepted: 04/30/2025] [Indexed: 06/11/2025] Open
Abstract
Munc13 family proteins are crucial for the secretion of neurotransmitters and hormones necessary for cell communication. They share a conserved C-terminal region that includes C2 and the MUN domains, which facilitate membrane interactions and the assembly of soluble N-ethylmaleimide sensitive factor attachment protein receptor (SNARE) complexes. Neuronal isoforms of Munc13 possess a variable N-terminal region that is essential for neurotransmitter release and short-term plasticity, although the precise functions of this region remain not fully understood. Here, we identified a negatively charged sequence within the N terminus of Munc13-1, termed polyE, which is specific to Munc13-1 among all Munc13 isoforms and potentially derived from a common ancestor of homeotherms. We found that polyE binds significantly to the MUN domain through charge-charge interactions, inhibiting MUN activity in promoting SNARE complex assembly. Disrupting the polyE-MUN interaction by introducing pseudophosphorylated mutations in the MUN domain alleviates this inhibition, thereby enhancing neurotransmitter release. Strikingly, Ca2+ ions exhibit significant binding to polyE. We found that 40 μM of Ca2+ adequately competes with the polyE-MUN interaction to reduce polyE inhibition. This concentration is comparable to presynaptic local [Ca2+]i triggered by a single action potential. Taken together, these results indicate an autoinhibition conformation of Munc13-1 mediated by the polyE-MUN interaction. In addition, the relief of this autoinhibition conformation of Munc13-1 by presynaptic Ca2+ influx and/or posttranslational modifications in the MUN domain may underlie Munc13-1 function in neurotransmitter release and short-term plasticity.
Collapse
Affiliation(s)
- Kexu Zhao
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Li Zhang
- Key Clinical Laboratory of Henan Province, Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Mengshi Lei
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Ziqi Jin
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Tianxin Du
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Hong Zhang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Yin Sheng
- School of Artificial Intelligence and Automation, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Zhitao Hu
- Department of Neuroscience, City University of Hong Kong, Kowloon 999077, China
| | - Shen Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Hubei Bioinformatics and Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Cong Ma
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Hubei Key Laboratory of Brain-inspired Intelligent Systems, School of Artificial Intelligence and Automation, Huazhong University of Science and Technology, Wuhan 430074, China
- Key Laboratory of Neurogenetics and Channelopathies of the Ministry of Education, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, China
| |
Collapse
|
3
|
Zhvania MG, Sharikadze I, Japaridze N, Tizabi Y, Rzayev F, Gasimov E, Lobzhanidze G. Status epilepticus alters hippocampal ultrastructure in kainic acid rat model. Tissue Cell 2025; 94:102789. [PMID: 39954563 DOI: 10.1016/j.tice.2025.102789] [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: 09/04/2024] [Revised: 01/21/2025] [Accepted: 02/06/2025] [Indexed: 02/17/2025]
Abstract
Kainic acid (KA) model of epilepsy is a reliable tool to study temporal lobe epilepsy (TLE), the most common type of partial epilepsy in adults. Substantial body of data suggest that the KA-induced status epilepticus (SE) leads to several molecular and structural changes in the hippocampus, including sclerosis, sprouting of mossy fiber, reorganization of inter-neuronal networks, alterations in neuropeptide signaling, gliosis, and synaptic transmission dysregulation. However, no details on the ultrastructural changes, especially in relationship to synapses are available. This information is important in providing a comprehensive understanding of subtle changes that occur in this debilitating disease. Thus, in this study, applying electron-microscopic morphometric analysis, we evaluated the ultrastructural effects of KA on the CA1 region of the hippocampus, an area intimately involved in SE. The total number of synaptic vesicles (SVs), the number of docking SVs, the length of synapse active zone (AZ) and the number and area of presynaptic and postsynaptic mitochondria in axo-dendritic (excitatory) synapses were measured at 24 h, and 8 and 21 days after KA administration. Results indicate a decrease in the total number and docking of SVs, an increase in the length of AZ and the number and area of presynaptic and postsynaptic mitochondria, which were more prominent at 8 days after KA injection. The findings suggest a time-dependent ultrastructural changes in CA1 region of the hippocampus in an animal model of focal epilepsy.
Collapse
Affiliation(s)
- Mzia G Zhvania
- School of Natural Sciences and Medicine, Ilia State University, Tbilisi, Georgia; Department of Brain Ultrastructure and Nanoarchitecture, Ivane Beritashvili Center of Experimental Biomedicine. Tbilisi, Georgia.
| | - Irina Sharikadze
- School of Natural Sciences and Medicine, Ilia State University, Tbilisi, Georgia
| | - Nadezhda Japaridze
- Department of Brain Ultrastructure and Nanoarchitecture, Ivane Beritashvili Center of Experimental Biomedicine. Tbilisi, Georgia; Carl Zeiss Scientific and Education Center, New Vision University, Tbilisi, Georgia
| | - Yousef Tizabi
- Department of Pharmacology, Howard University College of Medicine, Washington DC, USA
| | - Fuad Rzayev
- Department of Histology, Embryology and Cytology, Azerbaijan Medical University, Baku, Azerbaijan
| | - Eldar Gasimov
- Department of Histology, Embryology and Cytology, Azerbaijan Medical University, Baku, Azerbaijan
| | - Giorgi Lobzhanidze
- Department of Brain Ultrastructure and Nanoarchitecture, Ivane Beritashvili Center of Experimental Biomedicine. Tbilisi, Georgia
| |
Collapse
|
4
|
Fye M, Sangowdar P, Jayathilake A, Regan P, Gu G, Kaverina I. Directed insulin secretion from beta cells occurs at cortical sites devoid of microtubules at the edges of ELKS/LL5β patches. Mol Biol Cell 2025; 36:ar68. [PMID: 40366873 DOI: 10.1091/mbc.e24-10-0487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2025] Open
Abstract
To maintain normal blood glucose levels, pancreatic beta cells secrete insulin into the bloodstream at specialized regions at the cell periphery, often called secretion hot spots. While many secretory machinery components are located all over the cell membrane, directed secretion relies on distinct cortical patches of the scaffolding protein ELKS and the microtubule (MT)-anchoring protein LL5β. However, using total internal reflection fluorescence microscopy of intact mouse islets to precisely localize secretion events within ELKS/LL5β patches, we now show that secretion is restricted to only 5% of ELKS/LL5β patch area. Moreover, the majority of secretion occurs at the margins of ELKS patches. This suggests that additional factor(s) must be responsible for hot spot definition. Because the MT cytoskeleton plays a regulatory role in the insulin secretion process via both delivery and removal of secretory granules from the secretion sites, we test whether local MT organization defines secretory activity at hot spots. We find that the majority of secretion events occur at regions devoid of MTs. Based on our findings, we present a model in which local MT disassembly and optimal ELKS content are strong predictors of directed insulin secretion.
Collapse
Affiliation(s)
- Margret Fye
- Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37212
| | - Pranoy Sangowdar
- Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37212
| | - Anissa Jayathilake
- Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37212
| | - Pi'ilani Regan
- Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37212
| | - Guoqiang Gu
- Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37212
| | - Irina Kaverina
- Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37212
| |
Collapse
|
5
|
Zhou BB, Dong HJ, Sun H, Xie XM, Xie HH, Zhu WJ, Li YN, Xu C, Cao JP, Zhao GH, Yin K. Effects of latent infection of Toxoplasma gondii strains with different genotypes on mouse behavior and brain transcripts. Parasit Vectors 2025; 18:190. [PMID: 40420177 DOI: 10.1186/s13071-025-06819-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Accepted: 04/28/2025] [Indexed: 05/28/2025] Open
Abstract
BACKGROUND Toxoplasma gondii can cause severe damage to immunodeficient hosts, and also compromise brain structure and function in immunocompetent hosts during latent infection. In China, the two different isolates, Chinese I (ToxoDB#9) and Chinese III are dominant epidemic strains widely spreading in humans and domestic animals and can lead to latent infection in host brain tissues, but the comparison of their manipulation patterns and mechanisms remains unclear. METHODS Tachyzoites of the TgWh6 (Wh6) strain and the TgCtLHG (LHG) strain were used for establishing in vitro infection models within mouse microglia BV2 cells, and the differences in their invasion and proliferation patterns were observed. C57BL/6 J mice were used to establish in vivo latent infection models. After behavioral tests, the differential expressed transcripts (DETs) of the infected and control animals' cerebral cortex were sequenced by Nanopore RNA-seq. Functional differences of DETs were analyzed by Gene Ontology enrichment analysis (GO), Kyoto Encyclopedia of Genes and Genomes enrichment analysis (KEGG), and protein-protein interaction (PPI) and cluster analysis. Expression of the key candidates were verified by quantitative polymerase chain reaction (qPCR). RESULTS In our infection models, we found that Wh6 had more vigorous invasion and proliferation abilities in vitro, while LHG had a greater ability to form cysts in vivo. In the latent infection phase, behavioral changes, including spatial working memory, cognitive and motor abilities, and anxiety, were observed in both Wh6 and LHG infected mice; however, the LHG group showed more serious anxiety. Among DETs, genes related to major histocompatibility complex (MHC) class II molecules were significantly upregulated in the infected mice, while genes related to synaptic transmission and neurodegenerative diseases were downregulated in the infected groups. The downregulated DETs of Sept4, Kcng4, Unc13c, and Prkcg in the WH6 group, which are related to synaptic transmission, and Ndrg2 and Arc in the LHG group, which are related to neurodegenerative diseases, were selected to be the key candidates in the latent infection phase. CONCLUSIONS Compared with WH6, although LHG has a milder invasion ability, it can cause increased behavioral disorders in hosts. Genes related to synaptic transmission and neurodegenerative diseases may be the main causes of host mental and behavioral disorders.
Collapse
Affiliation(s)
- Bei-Bei Zhou
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research); Shandong Institute of Parasitic Diseases, Jining, 272033, China
- Key Laboratory of Parasite and Vector Biology, National Health Commission of the People's Republic of China, Shanghai, 200025, China
| | - Hong-Jie Dong
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research); Shandong Institute of Parasitic Diseases, Jining, 272033, China
- Key Laboratory of Parasite and Vector Biology, National Health Commission of the People's Republic of China, Shanghai, 200025, China
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Hang Sun
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research); Shandong Institute of Parasitic Diseases, Jining, 272033, China
| | - Xiao-Man Xie
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research); Shandong Institute of Parasitic Diseases, Jining, 272033, China
| | - Huan-Huan Xie
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research); Shandong Institute of Parasitic Diseases, Jining, 272033, China
| | - Wen-Ju Zhu
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research); Shandong Institute of Parasitic Diseases, Jining, 272033, China
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Ya-Nan Li
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research); Shandong Institute of Parasitic Diseases, Jining, 272033, China
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Chao Xu
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research); Shandong Institute of Parasitic Diseases, Jining, 272033, China
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Jian-Ping Cao
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research); Shandong Institute of Parasitic Diseases, Jining, 272033, China.
- Key Laboratory of Parasite and Vector Biology, National Health Commission of the People's Republic of China, Shanghai, 200025, China.
| | - Gui-Hua Zhao
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research); Shandong Institute of Parasitic Diseases, Jining, 272033, China.
| | - Kun Yin
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research); Shandong Institute of Parasitic Diseases, Jining, 272033, China.
- Key Laboratory of Parasite and Vector Biology, National Health Commission of the People's Republic of China, Shanghai, 200025, China.
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China.
| |
Collapse
|
6
|
Südhof TC. Signaling by latrophilin adhesion-GPCRs in synapse assembly. Neuroscience 2025; 575:150-161. [PMID: 40127755 DOI: 10.1016/j.neuroscience.2025.03.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 03/14/2025] [Accepted: 03/19/2025] [Indexed: 03/26/2025]
Abstract
Latrophilins are evolutionarily conserved adhesion-GPCRs with diverse roles, including a prominent function in synapse organization. In mammals, the primary transcripts of three latrophilin genes (ADGRL1-3) are extensively alternatively spliced, producing hundreds of isoforms with diverse cytoplasmic sequences. Extracellularly, latrophilins feature N-terminal lectin- and olfactomedin-like domains that bind to Teneurin and FLRT adhesion molecules, respectively, and are followed by an autoproteolytic GAIN domain typical for adhesion-GPCRs. Since Teneurins and FLRTs in turn interact with other ligands, latrophilins form a large trans-cellular protein interaction network. Intracellularly, latrophilins bind to G proteins, arrestins, and postsynaptic scaffold proteins. Latrophilins stimulate all Gα proteins tested, with the Gα isoform preference regulated by alternative splicing. In brain, latrophilins act as essential postsynaptic organizers that functionally require extracellular binding to teneurins and FLRTs, intracellular activation of GαS, and recruitment of postsynaptic scaffolds. Thus, latrophilins are signaling platforms that connect trans-cellular interactions to cellular responses in a manner regulated by alternative splicing.
Collapse
Affiliation(s)
- Thomas C Südhof
- Dept. of Molecular and Cellular Physiology & of Neurosurgery, Stanford University School of Medicine & Howard Hughes Medical Institute, Stanford Institute of Medicine I (SIM1)/Lorry Lokey Stem Cell Building, 265 Campus Drive, Room G1021, Stanford, CA 94305-5453, USA.
| |
Collapse
|
7
|
Milanick W, Li J, Thomas CI, Al-Yaari M, Guerrero-Given D, Kamasawa N, Young SM. Presynaptic α 2δs specify synaptic gain, not synaptogenesis, in the mammalian brain. Neuron 2025:S0896-6273(25)00296-X. [PMID: 40367942 DOI: 10.1016/j.neuron.2025.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 03/13/2025] [Accepted: 04/14/2025] [Indexed: 05/16/2025]
Abstract
The α2δs are a family of extracellular synaptic molecules that are auxiliary subunits of voltage-gated Ca2+ channel (CaV) complexes. They are linked to brain disorders and are drug targets. The α2δs are implicated in controlling synapse development and function through distinct CaV-dependent and CaV-independent pathways. However, the mechanisms of action remain enigmatic since synapses contain mixtures of α2δ isoforms in the pre- and postsynaptic compartments. We developed a triple conditional knockout mouse model and demonstrated the combined selective presynaptic ablation of α2δs in vivo in a developing mammalian glutamatergic synapse. We identified presynaptic α2δs as positive regulators of Munc13-1 levels, an essential neurotransmitter release protein. We found that mammalian synapse development, presynaptic CaV2.1 organization, and the transsynaptic alignment of presynaptic release sites and postsynaptic glutamate receptors are independent of presynaptic α2δs. Therefore, our results define presynaptic α2δ regulatory roles and suggest a new α2δ role in controlling synaptic strength and plasticity.
Collapse
Affiliation(s)
- William Milanick
- Gene Therapy Center, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA; Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, IA 52242, USA
| | - Jianing Li
- Gene Therapy Center, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA
| | - Connon I Thomas
- The Imaging Center and Electron Microscopy Core Facility, Max Planck Institute for Neuroscience, Jupiter, FL 33458, USA
| | - Mohammed Al-Yaari
- Gene Therapy Center, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA
| | - Debbie Guerrero-Given
- The Imaging Center and Electron Microscopy Core Facility, Max Planck Institute for Neuroscience, Jupiter, FL 33458, USA
| | - Naomi Kamasawa
- The Imaging Center and Electron Microscopy Core Facility, Max Planck Institute for Neuroscience, Jupiter, FL 33458, USA
| | - Samuel M Young
- Gene Therapy Center, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA; Department of Pediatrics, Department of Pharmacology, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
8
|
Hoagland A, Schultz R, Cai Z, Newman ZL, Isacoff EY. Behavioral resilience via dynamic circuit firing homeostasis. Proc Natl Acad Sci U S A 2025; 122:e2421386122. [PMID: 40299703 PMCID: PMC12067288 DOI: 10.1073/pnas.2421386122] [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: 10/16/2024] [Accepted: 03/22/2025] [Indexed: 05/01/2025] Open
Abstract
Homeostatic regulation ensures stable neural circuit output under changing conditions. We find that in Drosophila larvae, either presynaptic weakening due to perturbation of transmitter release or postsynaptic weakening due to perturbation of glutamate receptors at synapses between motor neuron (MN) and muscle has little impact on locomotion, suggesting a nonsynaptic compensatory mechanism. In vivo imaging shows that five different forms of synaptic weakening increase the duration of activity bouts in type I MNs. Strikingly, this compensation is input selective: occurring only in the tonic type Ib MN, not the phasic type Is MN that innervates the same muscle. Moreover, an inhibitory class of central pre-MNs that innervates the tonic-but not phasic-input decreases in activity. The adjustment in activity occurs remarkably quickly: within minutes of synapse perturbation. We propose that MN firing is dynamically regulated by two coordinated mechanisms: a cell-autonomous adjustment of MN excitability and a circuit adjustment of inhibitory central drive. The input selectivity of this process suggests homeostatic adjustment to maintain tonic drive but hold constant the phasic drive that organizes locomotory wave patterns.
Collapse
Affiliation(s)
- Adam Hoagland
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA94720
| | - Ryan Schultz
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA94720
| | - Zerong Cai
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA94720
| | - Zachary L. Newman
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA94720
| | - Ehud Y. Isacoff
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA94720
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA94720
- Department of Neuroscience, University of California Berkeley, Berkeley, CA94720
- Weill Neurohub, University of California Berkeley, Berkeley, CA94720
- Molecular Biophysics and Integrated BioImaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA94720
| |
Collapse
|
9
|
Blanco-Formoso M, Galluzzi F, Vacca F, Gianiorio T, Piergentili I, Cook AB, Welzen PLW, van Hest JCM, Di Marco S, Tantussi F, Benfenati F, Colombo E, De Angelis F. Spiropyran-based glutamate nanovalve for neuronal stimulation. MATERIALS HORIZONS 2025. [PMID: 40314589 DOI: 10.1039/d5mh00082c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
One of the main challenges in medical applications is achieving precise spatial and temporal control over the release of active molecules, such as neurotransmitters. To address this issue, we engineered a nanovalve that can deliver active molecules on demand by activating or deactivating a light-sensitive chemical barrier. This valve is composed of a polymer containing a spiropyran moiety, which can switch from a hydrophobic to a hydrophilic state upon photo-stimulation. Accordingly, the nanovalve either blocks or allows molecular diffusion through a solid-state nanopore array. Here, we demonstrate that the system blocks up to 96% of the translocation of the neurotransmitter glutamate and that the on-demand release of glutamate upon light stimulation reaches 60 μM h-1, mimicking a physiological synaptic release rate. We proved its cytocompatibility and analyzed its potential for the stimulation of primary neurons and blind retinal explants by patch-clamp experiments. These results represent a milestone for the development of biomimetic neuroprostheses restoring chemical synaptic transmission lost by degeneration or delivering drugs in a light-controlled fashion.
Collapse
Affiliation(s)
- M Blanco-Formoso
- Plasmon Nanotechnology, Istituto Italiano di Tecnologia, Genova, Italy.
- CINBIO Universidade de Vigo, Vigo, Spain
| | - F Galluzzi
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy.
- The Open University Affiliated Research Centre at Istituto Italiano di Tecnologia (ARC@IIT), Genova, Italy
| | - F Vacca
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy.
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - T Gianiorio
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy.
- Department of Neuroscience (DINOGMI), University of Genoa, Genova, Italy
| | - I Piergentili
- Bio-Organic Chemistry, Department of Biomedical Engineering, Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, The Netherlands
| | - A B Cook
- Bio-Organic Chemistry, Department of Biomedical Engineering, Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, The Netherlands
| | - P L W Welzen
- Bio-Organic Chemistry, Department of Biomedical Engineering, Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, The Netherlands
| | - J C M van Hest
- Bio-Organic Chemistry, Department of Biomedical Engineering, Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, The Netherlands
| | - S Di Marco
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy.
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - F Tantussi
- Plasmon Nanotechnology, Istituto Italiano di Tecnologia, Genova, Italy.
| | - F Benfenati
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy.
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - E Colombo
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy.
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - F De Angelis
- Plasmon Nanotechnology, Istituto Italiano di Tecnologia, Genova, Italy.
| |
Collapse
|
10
|
Amaral L, Martins M, Côrte-Real M, Outeiro TF, Chaves SR, Rego A. The neurotoxicity of pesticides: Implications for Parkinson's disease. CHEMOSPHERE 2025; 377:144348. [PMID: 40203643 DOI: 10.1016/j.chemosphere.2025.144348] [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: 09/02/2024] [Revised: 03/04/2025] [Accepted: 03/19/2025] [Indexed: 04/11/2025]
Abstract
Parkinson's disease (PD) is the fastest-growing neurodegenerative disorder worldwide, and no effective cure is currently available. Neuropathologically, PD is characterized by the selective degeneration of dopaminergic neurons in the substantia nigra and by the accumulation of alpha-synuclein (aSyn)-rich proteinaceous inclusions within surviving neurons. As a multifactorial disorder, approximately 85 % of PD cases are sporadic with unknown etiology. Among the many risk factors implicated in PD, exposure to neurotoxic pesticides stands out as a significant contributor. While the effects of many are still uncharacterized, it has already been shown that rotenone, paraquat, maneb, and dieldrin affect critical cellular pathways, including mitochondrial and proteasomal dysfunction, aSyn aggregation, autophagy dysregulation, and disruption of dopamine metabolism. With the constant rise in pesticide usage to meet the demands of a growing human population, the risk of environmental contamination and subsequent PD development is also increasing. This review explores the molecular mechanisms by which pesticide exposure influences PD development, shedding light on their role in the pathogenesis of PD and highlighting the need for preventative measures and regulatory oversight to mitigate these risks.
Collapse
Affiliation(s)
- Leslie Amaral
- CBMA - Centre of Molecular and Environmental Biology / ARNET - Aquatic Research Network, Department of Biology, School of Sciences, University of Minho, 4710-057, Braga, Portugal; University Medical Center Göttingen, Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, Göttingen, Germany
| | - Márcia Martins
- CBMA - Centre of Molecular and Environmental Biology / ARNET - Aquatic Research Network, Department of Biology, School of Sciences, University of Minho, 4710-057, Braga, Portugal
| | - Manuela Côrte-Real
- CBMA - Centre of Molecular and Environmental Biology / ARNET - Aquatic Research Network, Department of Biology, School of Sciences, University of Minho, 4710-057, Braga, Portugal
| | - Tiago F Outeiro
- University Medical Center Göttingen, Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, Göttingen, Germany; Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK; Max Planck Institute for Multidisciplinary Sciences, 37075, Göttingen, Germany; Scientific Employee with an Honorary Contract at Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Göttingen, Germany
| | - Susana R Chaves
- CBMA - Centre of Molecular and Environmental Biology / ARNET - Aquatic Research Network, Department of Biology, School of Sciences, University of Minho, 4710-057, Braga, Portugal.
| | - António Rego
- Department of Biology, School of Sciences, University of Minho, 4710-057, Braga, Portugal; Solfarcos, Pharmaceutical and Cosmetic Solutions, Braga, Portugal.
| |
Collapse
|
11
|
Delvendahl I, Daswani R, Winterer J, Germain PL, Uhr NM, Schratt G, Müller M. MicroRNA-138-5p suppresses excitatory synaptic strength at the cerebellar input layer. J Physiol 2025; 603:3161-3179. [PMID: 40349307 DOI: 10.1113/jp288019] [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: 10/31/2024] [Accepted: 04/14/2025] [Indexed: 05/14/2025] Open
Abstract
MicroRNAs are small, highly conserved non-coding RNAs that negatively regulate mRNA translation and stability. In the brain, miRNAs contribute to neuronal development, synaptogenesis, and synaptic plasticity. MicroRNA 138-5p (miR-138-5p) controls inhibitory synaptic transmission in the hippocampus and is highly expressed in cerebellar excitatory neurons. However, its specific role in cerebellar synaptic transmission remains unknown. Here, we investigated excitatory transmission in the cerebellum of mice expressing a sponge construct that sequesters endogenous miR-138-5p. Mossy fibre stimulation-evoked EPSCs in granule cells were ∼40% larger in miR-138-5p sponge mice compared to controls. Furthermore, we observed larger miniature EPSC amplitudes, suggesting an increased number of functional postsynaptic AMPA receptors. High-frequency train stimulation revealed enhanced short-term depression following miR-138-5p downregulation. Together with computational modelling, this suggests a negative regulation of presynaptic release probability. Overall, our results demonstrate that miR-138-5p suppresses synaptic strength through pre- and postsynaptic mechanisms, providing a potentially powerful mechanism for tuning excitatory synaptic input into the cerebellum. KEY POINTS: MicroRNAs are powerful regulators of mRNA translation and control key cell biological processes including synaptic transmission, but their role in regulating synaptic function in the cerebellum has remained elusive. In this study, we investigated how microRNA-138-5p (miR-138-5p) modulates excitatory transmission at adult murine cerebellar mossy fibre to granule cell synapses. Downregulation of miR-138-5p enhances excitatory synaptic strength at the cerebellar input layer and increases short-term depression. miR-138-5p exerts its regulatory function through both pre- and postsynaptic mechanisms by negatively regulating release probability at mossy fibre boutons, as well as functional AMPA receptor numbers in granule cells. These findings provide insights into the role of miR-138-5p in the cerebellum and expand our understanding of microRNA-dependent control of excitatory synaptic transmission and short-term plasticity.
Collapse
Affiliation(s)
- Igor Delvendahl
- Department of Molecular Life Sciences, University of Zurich (UZH), Zurich, Switzerland
- Neuroscience Center Zurich, Zurich, Switzerland
- Institute of Physiology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Reetu Daswani
- Lab of Systems Neuroscience, Institute for Neuroscience, Department of Health Science and Technology, Swiss Federal Institute of Technology ETH, Zurich, Switzerland
- Present address: Sixfold Bioscience Ltd, Translation and Innovation Hub, London, UK
| | - Jochen Winterer
- Lab of Systems Neuroscience, Institute for Neuroscience, Department of Health Science and Technology, Swiss Federal Institute of Technology ETH, Zurich, Switzerland
| | - Pierre-Luc Germain
- Lab of Systems Neuroscience, Institute for Neuroscience, Department of Health Science and Technology, Swiss Federal Institute of Technology ETH, Zurich, Switzerland
| | - Nora Maria Uhr
- Lab of Systems Neuroscience, Institute for Neuroscience, Department of Health Science and Technology, Swiss Federal Institute of Technology ETH, Zurich, Switzerland
| | - Gerhard Schratt
- Neuroscience Center Zurich, Zurich, Switzerland
- Lab of Systems Neuroscience, Institute for Neuroscience, Department of Health Science and Technology, Swiss Federal Institute of Technology ETH, Zurich, Switzerland
| | - Martin Müller
- Department of Molecular Life Sciences, University of Zurich (UZH), Zurich, Switzerland
- Neuroscience Center Zurich, Zurich, Switzerland
- University Research Priority Program (URPP), Adaptive Brain Circuits in Development and Learning (AdaBD), University of Zurich, Zurich, Switzerland
| |
Collapse
|
12
|
Wilson P, Kim N, Cotter R, Parkes M, Cmelak L, Reed MN, Gramlich MW. Presynaptic recycling pool density regulates spontaneous synaptic vesicle exocytosis rate and is upregulated in the presence of β-amyloid. Cell Rep 2025; 44:115410. [PMID: 40146773 DOI: 10.1016/j.celrep.2025.115410] [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: 09/17/2024] [Revised: 01/17/2025] [Accepted: 02/17/2025] [Indexed: 03/29/2025] Open
Abstract
Synapses represent a fundamental unit of information transfer during cognition via presynaptic vesicle exocytosis. It has been established that evoked release is probabilistic, but the mechanisms behind spontaneous release are less clear. Understanding spontaneous release is vital, as it plays a key role in maintaining synaptic connections. We propose a model framework for spontaneous release where the reserve pool geometrically constrains recycling pool vesicles, creating an entropic force that drives spontaneous release rate. We experimentally support this framework using SEM, fluorescence microscopy, computational modeling, and pharmacological approaches. Our model correctly predicts the spontaneous release rate as a function of presynapse size. Finally, we use our approach to show how β-amyloid mutations linked to Alzheimer's disease lead to increased spontaneous release rates. These results indicate that synapses regulate the density of the recycling pool to control the spontaneous release rate and may serve as an early indicator of Alzheimer's disease.
Collapse
Affiliation(s)
- Paxton Wilson
- Department of Physics, Auburn University, Auburn, AL, USA
| | - Noah Kim
- Department of Physics, Auburn University, Auburn, AL, USA
| | - Rachel Cotter
- Department of Physics, Auburn University, Auburn, AL, USA
| | - Mason Parkes
- Department of Physics, Auburn University, Auburn, AL, USA
| | - Luca Cmelak
- Department of Psychological Sciences, Auburn University, Auburn, AL, USA
| | - Miranda N Reed
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, USA; Center for Neuroscience Initiative, Auburn University, Auburn, AL, USA
| | - Michael W Gramlich
- Department of Physics, Auburn University, Auburn, AL, USA; Center for Neuroscience Initiative, Auburn University, Auburn, AL, USA.
| |
Collapse
|
13
|
Ainatzi S, Kaufmann SV, Silbern I, Georgiev SV, Lorenz S, Rizzoli SO, Urlaub H. Ca 2+-Triggered (de)ubiquitination Events in Synapses. Mol Cell Proteomics 2025; 24:100946. [PMID: 40089065 PMCID: PMC12008530 DOI: 10.1016/j.mcpro.2025.100946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 01/29/2025] [Accepted: 03/11/2025] [Indexed: 03/17/2025] Open
Abstract
Neuronal communication relies on neurotransmitter release from synaptic vesicles (SVs), whose dynamics are controlled by Ca2+-dependent pathways, as many thoroughly studied phosphorylation cascades. However, little is known about other post-translational modifications, such as ubiquitination. To address this, we analyzed resting and stimulated synaptosomes (isolated synapses) by quantitative mass spectrometry. We identified more than 5000 ubiquitination sites on ∼2000 proteins, the majority of which participate in SV recycling processes. Several proteins showed significant changes in ubiquitination in response to Ca2+ influx, with the most pronounced changes in CaMKIIα and the clathrin adaptor protein AP180. To validate this finding, we generated a CaMKIIα mutant lacking the ubiquitination target site (K291) and analyzed it both in neurons and non-neuronal cells. K291 ubiquitination, close to an important site for CaMKIIα autophosphorylation (T286), influences the synaptic function of this kinase. We suggest that ubiquitination in response to synaptic activity is an important regulator of synaptic function.
Collapse
Affiliation(s)
- Sofia Ainatzi
- Bioanalytical Mass Spectrometry, Max Planck Institute for Multidisciplinary Sciences, Goettingen, Germany; Bioanalytics, Institute of Clinical Chemistry, University Medical Center, Goettingen, Germany
| | - Svenja V Kaufmann
- Bioanalytical Mass Spectrometry, Max Planck Institute for Multidisciplinary Sciences, Goettingen, Germany; Bioanalytics, Institute of Clinical Chemistry, University Medical Center, Goettingen, Germany
| | - Ivan Silbern
- Bioanalytical Mass Spectrometry, Max Planck Institute for Multidisciplinary Sciences, Goettingen, Germany; Bioanalytics, Institute of Clinical Chemistry, University Medical Center, Goettingen, Germany
| | - Svilen V Georgiev
- Department of Neuro- and Sensory Physiology, University Medical Center, Goettingen, Germany
| | - Sonja Lorenz
- Ubiquitin Signaling Specificity, Max Planck Institute for Multidisciplinary Sciences, Goettingen, Germany
| | - Silvio O Rizzoli
- Department of Neuro- and Sensory Physiology, University Medical Center, Goettingen, Germany
| | - Henning Urlaub
- Bioanalytical Mass Spectrometry, Max Planck Institute for Multidisciplinary Sciences, Goettingen, Germany; Bioanalytics, Institute of Clinical Chemistry, University Medical Center, Goettingen, Germany; Cluster of Excellence Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells (MBExC), University of Göttingen, Germany; Göttingen Center for Molecular Biosciences, Georg August University Göttingen, Germany.
| |
Collapse
|
14
|
Fye M, Sangowdar P, Jayathilake A, Noguchi P, Gu G, Kaverina I. Directed insulin secretion from beta cells occurs at cortical sites devoid of microtubules at the edges of ELKS/LL5β patches. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.10.31.621333. [PMID: 39553950 PMCID: PMC11565951 DOI: 10.1101/2024.10.31.621333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
To maintain normal blood glucose levels, pancreatic beta cells secrete insulin into the bloodstream at specialized regions at the cell periphery, often called secretion hot spots. While many secretory machinery components are located all over the cell membrane, directed secretion relies on distinct cortical patches of the scaffolding protein ELKS and the microtubule (MT)-anchoring protein LL5β. However, using TIRF microscopy of intact mouse islets to precisely localize secretion events within ELKS/LL5β patches, we now show that secretion is restricted to only 5% of ELKS/LL5β patch area. Moreover, the majority of secretion occurs at the margins of ELKS patches. This suggests that additional factor(s) must be responsible for hot spot definition. Because the MT cytoskeleton plays a regulatory role in the insulin secretion process via both delivery and removal of secretory granules from the secretion sites, we test whether local MT organization defines secretory activity at hot spots. We find that the majority of secretion events occur at regions devoid of MTs. Based on our findings, we present a model in which local MT disassembly and optimal ELKS content are strong predictors of directed insulin secretion.
Collapse
Affiliation(s)
- Margret Fye
- Cell & Developmental Biology, Vanderbilt University School of Medicine
| | - Pranoy Sangowdar
- Cell & Developmental Biology, Vanderbilt University School of Medicine
| | | | - Pi'ilani Noguchi
- Cell & Developmental Biology, Vanderbilt University School of Medicine
| | - Guoqiang Gu
- Cell & Developmental Biology, Vanderbilt University School of Medicine
| | - Irina Kaverina
- Cell & Developmental Biology, Vanderbilt University School of Medicine
| |
Collapse
|
15
|
Torun A, Tuğral H, Banerjee S. Crosstalk Between Phase-Separated Membraneless Condensates and Membrane-Bound Organelles in Cellular Function and Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025. [PMID: 40095243 DOI: 10.1007/5584_2025_852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Compartmentalization in eukaryotic cells allows the spatiotemporal regulation of biochemical processes, in addition to allowing specific sets of proteins to interact in a regulated as well as stochastic manner. Although membrane-bound organelles are thought to be the key players of cellular compartmentalization, membraneless biomolecular condensates such as stress granules, P bodies, and many others have recently emerged as key players that are also thought to bring order to a highly chaotic environment. Here, we have evaluated the latest studies on biomolecular condensates, specifically focusing on how they interact with membrane-bound organelles and modulate each other's functions. We also highlight the importance of this interaction in neurodegenerative and cardiovascular diseases as well as in cancer.
Collapse
Affiliation(s)
- Aydan Torun
- Department of Biological Sciences, Orta Dogu Teknik Universitesi, Ankara, Türkiye
| | - Hoşnaz Tuğral
- Department of Biological Sciences, Orta Dogu Teknik Universitesi, Ankara, Türkiye
| | - Sreeparna Banerjee
- Department of Biological Sciences, Orta Dogu Teknik Universitesi, Ankara, Türkiye.
| |
Collapse
|
16
|
Kurashina M, Snow AW, Mizumoto K. A modular system to label endogenous presynaptic proteins using split fluorophores in Caenorhabditis elegans. Genetics 2025; 229:iyae214. [PMID: 39708832 PMCID: PMC11912834 DOI: 10.1093/genetics/iyae214] [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: 09/21/2024] [Accepted: 12/15/2024] [Indexed: 12/23/2024] Open
Abstract
Visualizing the subcellular localization of presynaptic proteins with fluorescent proteins is a powerful tool to dissect the genetic and molecular mechanisms underlying synapse formation and patterning in live animals. Here, we utilize split green and red fluorescent proteins to visualize the localization of endogenously expressed presynaptic proteins at a single-neuron resolution in Caenorhabditis elegans. By using CRISPR/Cas9 genome editing, we generated a collection of C. elegans strains in which endogenously expressed presynaptic proteins (RAB-3/Rab3, SNG-1/Synaptogyrin, CLA-1/Piccolo, SYD-2/Liprin-α, UNC-10/RIM, RIMB-1/RIM-BP, and ELKS-1/ELKS) are tagged with tandem repeats of GFP11 and/or wrmScarlet11. We show that the expression of GFP1-10 and wrmScarlet1-10 under neuron-specific promoters can robustly label presynaptic proteins in different neuron types. We believe that the combination of our knock-in strains and GFP1-10 and wrmScarlet1-10 plasmids is a versatile modular system useful for neuroscientists to examine the localization of endogenous presynaptic proteins in any neuron type in C. elegans.
Collapse
Affiliation(s)
- Mizuki Kurashina
- Graduate Program of Cell and Developmental Biology, Life Sciences Institute, The University of British Columbia, Vancouver, Canada V6T 1Z3
- Department of Zoology, The University of British Columbia, Vancouver, Canada V6T 1Z3
| | - Andrew W Snow
- Graduate Program of Cell and Developmental Biology, Life Sciences Institute, The University of British Columbia, Vancouver, Canada V6T 1Z3
- Department of Zoology, The University of British Columbia, Vancouver, Canada V6T 1Z3
| | - Kota Mizumoto
- Graduate Program of Cell and Developmental Biology, Life Sciences Institute, The University of British Columbia, Vancouver, Canada V6T 1Z3
- Department of Zoology, The University of British Columbia, Vancouver, Canada V6T 1Z3
- Djavad Mowafaghian Centre for Brain Health, The University of British Columbia, Vancouver, Canada V6T 1Z3
| |
Collapse
|
17
|
Xu N, Chen SY, Tang AH. Tuning synapse strength by nanocolumn plasticity. Trends Neurosci 2025; 48:200-212. [PMID: 39848836 DOI: 10.1016/j.tins.2024.12.009] [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: 10/01/2024] [Revised: 12/12/2024] [Accepted: 12/31/2024] [Indexed: 01/25/2025]
Abstract
The precise organization of the complex set of synaptic proteins at the nanometer scale is crucial for synaptic transmission. At the heart of this nanoscale architecture lies the nanocolumn. This aligns presynaptic neurotransmitter release with a high local density of postsynaptic receptor channels, thereby optimizing synaptic strength. Although synapses exhibit diverse protein compositions and nanoscale organizations, the role of structural diversity in the notable differences observed in synaptic physiology remains poorly understood. In this review we examine the current literature on the molecular mechanisms underlying the formation and maintenance of nanocolumns, as well as their role in modulating various aspects of synaptic transmission. We also discuss how the reorganization of nanocolumns contributes to functional dynamics in both synaptic plasticity and pathology.
Collapse
Affiliation(s)
- Na Xu
- Hefei National Laboratory for Physical Sciences at the Microscale, Department of Neurology in the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230088, China; School of Medicine, Anhui University of Science and Technology, Huainan 232001, China.
| | - Si-Yu Chen
- Hefei National Laboratory for Physical Sciences at the Microscale, Department of Neurology in the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230088, China
| | - Ai-Hui Tang
- Hefei National Laboratory for Physical Sciences at the Microscale, Department of Neurology in the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230088, China.
| |
Collapse
|
18
|
Cheng Q, Fan Y, Zhang P, Liu H, Han J, Yu Q, Wang X, Wu S, Lu Z. Biomarkers of synaptic degeneration in Alzheimer's disease. Ageing Res Rev 2025; 104:102642. [PMID: 39701184 DOI: 10.1016/j.arr.2024.102642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 12/13/2024] [Accepted: 12/14/2024] [Indexed: 12/21/2024]
Abstract
Synapse has been considered a critical neuronal structure in the procession of Alzheimer's disease (AD), attacked by two pathological molecule aggregates (amyloid-β and phosphorylated tau) in the brain, disturbing synaptic homeostasis before disease manifestation and subsequently causing synaptic degeneration. Recently, evidence has emerged indicating that soluble oligomeric amyloid-β (AβO) and tau exert direct toxicity on synapses, causing synaptic damage. Synaptic degeneration is closely linked to cognitive decline in AD, even in the asymptomatic stages of AD. Therefore, the identification of novel, specific, and sensitive biomarkers involved in synaptic degeneration holds significant promise for early diagnosis of AD, reducing synaptic degeneration and loss, and controlling the progression of AD. Currently, a range of biomarkers in cerebrospinal fluid (CSF), such as synaptosome-associated protein 25 (SNAP-25), synaptotagmin-1, growth-associated protein-43 (GAP-43), and neurogranin (Ng), along with functional brain imaging techniques, can detect variations in synaptic density, offering high sensitivity and specificity for AD diagnosis. However, these methods face challenges, including invasiveness, high cost, and limited accessibility. In contrast, biomarkers found in blood or urine provide a minimally invasive, cost-effective, and more accessible alternative to traditional diagnostic methods. Notably, neuron-derived exosomes in blood, which contain synaptic proteins, show variations in concentration that can serve as indicators of synaptic injury, providing an additional, less invasive approach to AD diagnosis and monitoring.
Collapse
Affiliation(s)
- Qian Cheng
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Yiou Fan
- Laboratory and Quality Management Department, Centers for Disease Control and Prevention of Shandong, Jinan, Shandong, China
| | - Pengfei Zhang
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Huan Liu
- Department of Clinical Laboratory, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China
| | - Jialin Han
- Department of Clinical Laboratory, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China
| | - Qian Yu
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Xueying Wang
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Shuang Wu
- Department of Clinical Laboratory, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China
| | - Zhiming Lu
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China.
| |
Collapse
|
19
|
Zhu S, Shen Z, Wu X, Zhang M. Phase separation in the multi-compartment organization of synapses. Curr Opin Neurobiol 2025; 90:102975. [PMID: 39893931 DOI: 10.1016/j.conb.2025.102975] [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: 10/15/2024] [Revised: 12/12/2024] [Accepted: 01/13/2025] [Indexed: 02/04/2025]
Abstract
A neuronal synapse is formed by juxtaposition of a transmitter releasing presynaptic bouton of one neuron with a transmitter receiving postsynaptic compartment such as a spine protrusion of another neuron. Each presynaptic bouton and postsynaptic spine, though very small in their volumes already, are further compartmentalized to micro-/nano-domains with distinct molecular organizations and synaptic functions. This review summarizes studies in recent years demonstrating that multivalent protein-protein interaction-induced phase separation underlies formation and coexistence of multiple distinct molecular condensates within tiny synapses. In post-synapses where synaptic compartmentalization via phase separation was first demonstrated, phase separation allows clustering of transmitter receptors into distinct nanodomains and renders postsynaptic densities to be regulated by synaptic stimulation signals for plasticity. In pre-synapses, such phase separation-mediated synaptic condensates formation allows SVs to be stored as distinct pools and directly transported for activity-induced transmitter release.
Collapse
Affiliation(s)
- Shihan Zhu
- School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China; Division of Life Science, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, Kowloon, China
| | - Zeyu Shen
- School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China; Division of Life Science, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, Kowloon, China
| | - Xiandeng Wu
- School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Mingjie Zhang
- School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China.
| |
Collapse
|
20
|
Plaza-Alonso S, Cano-Astorga N, DeFelipe J, Alonso-Nanclares L. Volume electron microscopy reveals unique laminar synaptic characteristics in the human entorhinal cortex. eLife 2025; 14:e96144. [PMID: 39882848 PMCID: PMC11867616 DOI: 10.7554/elife.96144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 01/27/2025] [Indexed: 01/31/2025] Open
Abstract
The entorhinal cortex (EC) plays a pivotal role in memory function and spatial navigation, connecting the hippocampus with the neocortex. The EC integrates a wide range of cortical and subcortical inputs, but its synaptic organization in the human brain is largely unknown. We used volume electron microscopy to perform a 3D analysis of the microanatomical features of synapses in all layers of the medial EC (MEC) from the human brain. Using this technology, 12,974 synapses were fully 3D reconstructed at the ultrastructural level. The MEC presented a distinct set of synaptic features, differentiating this region from other human cortical areas. Furthermore, ultrastructural synaptic characteristics within the MEC was predominantly similar, although layers I and VI exhibited several synaptic characteristics that were distinct from other layers. The present study constitutes an extensive description of the synaptic characteristics of the neuropil of all layers of the EC, a crucial step to better understand the connectivity of this cortical region, in both health and disease.
Collapse
Affiliation(s)
- Sergio Plaza-Alonso
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de MadridMadridSpain
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC)MadridSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIIIMadridSpain
| | - Nicolas Cano-Astorga
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de MadridMadridSpain
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC)MadridSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIIIMadridSpain
| | - Javier DeFelipe
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de MadridMadridSpain
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC)MadridSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIIIMadridSpain
| | - Lidia Alonso-Nanclares
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de MadridMadridSpain
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC)MadridSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIIIMadridSpain
| |
Collapse
|
21
|
Maikap S, Lucaciu A, Chakraborty A, Kestner RI, Vutukuri R, Annamneedi A. Targeting the Neuro-vascular Presynaptic Signalling in STROKE: Evidence and Therapeutic Implications. Ann Neurosci 2025:09727531241310048. [PMID: 39886458 PMCID: PMC11775937 DOI: 10.1177/09727531241310048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 12/09/2024] [Indexed: 02/01/2025] Open
Abstract
Background Stroke is one of the leading causes of death and long-term adult disability worldwide. Stroke causes neurodegeneration and impairs synaptic function. Understanding the role of synaptic proteins and associated signalling pathways in stroke pathology could offer insights into therapeutic approaches as well as improving rehabilitation-related treatment regimes. Purpose The current study aims to analyse synaptic transcriptome changes in acute and long-term post-stroke (1 day, 7 day timepoints), especially focusing on pre- and postsynaptic genes. Methods We performed data mining of the recent mRNA sequence from isolated mouse brain micro-vessels (MBMVs) after transient middle cerebral artery occlusion (tMCAO) stroke model. Using the SynGO (Synaptic Gene Ontologies and annotations) bioinformatics platform we assessed synaptic protein expression and associated pathways, and compared synaptic protein changes at 1 day and 7 day post-stroke. Results Enrichment analysis of the MBMVs identified significant alterations in the expression of genes related to synaptic physiology, synaptic transmission, neuronal structure, and organisation. We identified that the synaptic changes observed at the 7 day timepoint were initiated by the regulation of specific presynaptic candidates 1 day (24h) post-stroke, highlighting the significance of presynaptic regulation in mediating organising of synaptic structures and physiology. Analysis of transcriptomic data from human postmortem stroke brains confirmed similar presynaptic signalling patterns. Conclusion Our findings identify the changes in presynaptic gene regulation in micro-vessels following ischaemic stroke. Targeting presynaptic active zone protein signalling could represent a promising therapeutic target in mitigating ischaemic stroke.
Collapse
Affiliation(s)
- Shimantika Maikap
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Alexandra Lucaciu
- Department of Neurology, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Aheli Chakraborty
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Roxane Isabelle Kestner
- Department of Neurology, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main, Germany
- Institute of General Pharmacology and Toxicology, Pharmazentrum Frankfurt, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Rajkumar Vutukuri
- Institute of General Pharmacology and Toxicology, Pharmazentrum Frankfurt, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Anil Annamneedi
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| |
Collapse
|
22
|
Zhang X, Chen X, Matúš D, Südhof TC. Reconstitution of synaptic junctions orchestrated by teneurin-latrophilin complexes. Science 2025; 387:322-329. [PMID: 39818903 PMCID: PMC11808628 DOI: 10.1126/science.adq3586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 09/05/2024] [Accepted: 11/18/2024] [Indexed: 01/30/2025]
Abstract
Synapses are organized by trans-synaptic adhesion molecules that coordinate assembly of pre- and postsynaptic specializations, which, in turn, are composed of scaffolding proteins forming liquid-liquid phase-separated condensates. Presynaptic teneurins mediate excitatory synapse organization by binding to postsynaptic latrophilins; however, the mechanism of action of teneurins, driven by extracellular domains evolutionarily derived from bacterial toxins, remains unclear. In this work, we show that only the intracellular sequence, a dimerization sequence, and extracellular bacterial toxin-derived latrophilin-binding domains of Teneurin-3 are required for synapse organization, suggesting that teneurin-induced latrophilin clustering mediates synaptogenesis. Intracellular Teneurin-3 sequences capture liquid-liquid phase-separated presynaptic active zone scaffolds, enabling us to reconstitute an entire synaptic junction from purified proteins in which trans-synaptic teneurin-latrophilin complexes recruit phase-separated pre- and postsynaptic specializations.
Collapse
Affiliation(s)
| | | | - Daniel Matúš
- Department of Molecular and Cellular Physiology, Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Thomas C. Südhof
- Department of Molecular and Cellular Physiology, Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
23
|
Lovatt C, O'Sullivan TJ, Luis CODS, Ryan TJ, Frank RAW. Memory engram synapse 3D molecular architecture visualized by cryoCLEM-guided cryoET. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.09.632151. [PMID: 39829918 PMCID: PMC11741270 DOI: 10.1101/2025.01.09.632151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Memory is incorporated into the brain as physicochemical changes to engram cells. These are neuronal populations that form complex neuroanatomical circuits, are modified by experiences to store information, and allow for memory recall. At the molecular level, learning modifies synaptic communication to rewire engram circuits, a mechanism known as synaptic plasticity. However, despite its functional role on memory formation, the 3D molecular architecture of synapses within engram circuits is unknown. Here, we demonstrate the use of engram labelling technology and cryogenic correlated light and electron microscopy (cryoCLEM)-guided cryogenic electron tomography (cryoET) to visualize the in-tissue 3D molecular architecture of engram synapses of a contextual fear memory within the CA1 region of the mouse hippocampus. Engram cells exhibited structural diversity of macromolecular constituents and organelles in both pre- and postsynaptic compartments and within the synaptic cleft, including in clusters of membrane proteins, synaptic vesicle occupancy, and F-actin copy number. This 'engram to tomogram' approach, harnessing in vivo functional neuroscience and structural biology, provides a methodological framework for testing fundamental molecular plasticity mechanisms within engram circuits during memory encoding, storage and recall.
Collapse
Affiliation(s)
- Charlie Lovatt
- Astbury Centre for Structural Biology, School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Thomas J O'Sullivan
- Astbury Centre for Structural Biology, School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Clara Ortega-de San Luis
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
- Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
- Department of Health Sciences, University of Jaén, Jaén, Spain
| | - Tomás J Ryan
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
- Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Melbourne, Victoria, Australia
- Child & Brain Development Program, Canadian Institute for Advanced Research (CIFAR), Toronto, Ontario, Canada
| | - René A W Frank
- Astbury Centre for Structural Biology, School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
24
|
Lützkendorf J, Matkovic-Rachid T, Liu S, Götz T, Gao L, Turrel O, Maglione M, Grieger M, Putignano S, Ramesh N, Ghelani T, Neumann A, Gimber N, Schmoranzer J, Stawrakakis A, Brence B, Baum D, Ludwig K, Heine M, Mielke T, Liu F, Walter AM, Wahl MC, Sigrist SJ. Blobby is a synaptic active zone assembly protein required for memory in Drosophila. Nat Commun 2025; 16:271. [PMID: 39747038 PMCID: PMC11696761 DOI: 10.1038/s41467-024-55382-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 12/10/2024] [Indexed: 01/04/2025] Open
Abstract
At presynaptic active zones (AZs), scaffold proteins are critical for coordinating synaptic vesicle release and forming essential nanoarchitectures. However, regulatory principles steering AZ scaffold assembly, function, and plasticity remain insufficiently understood. We here identify an additional Drosophila AZ protein, "Blobby", essential for proper AZ nano-organization. Blobby biochemically associates with the ELKS family AZ scaffold protein Bruchpilot (BRP) and integrates into newly forming AZs. Loss of Blobby results in fewer AZs forming, ectopic AZ scaffold protein accumulations ("blobs") and disrupts nanoscale architecture of the BRP-AZ scaffold. Functionally, blobby mutants show diminished evoked synaptic currents due to reduced synaptic vesicle release probability and fewer functional release sites. Blobby is also present in adult brain synapses, and post-developmental knockdown of Blobby in the mushroom body impairs olfactory aversive memory consolidation. Thus, our analysis identifies an additional layer of AZ regulation critical for developmental AZ assembly but also for AZ-mediated plasticity controlling behavior.
Collapse
Affiliation(s)
- J Lützkendorf
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
| | - T Matkovic-Rachid
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
| | - S Liu
- Freie Universität Berlin, Institute of Chemistry and Biochemistry/Structural Biochemistry, Berlin, Germany
| | - T Götz
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
| | - L Gao
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
| | - O Turrel
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
| | - M Maglione
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, SupraFAB, Berlin, Germany
| | - M Grieger
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
| | - S Putignano
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
| | - N Ramesh
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
| | - T Ghelani
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Campus Berlin-Buch, Berlin, Germany
| | - A Neumann
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
| | - N Gimber
- Charité- Universitätsmedizin, Advanced Medical Bioimaging Core Facility, Berlin, Germany
| | - J Schmoranzer
- Charité- Universitätsmedizin, Advanced Medical Bioimaging Core Facility, Berlin, Germany
| | - A Stawrakakis
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
| | - B Brence
- Zuse Institute Berlin, Department of Visual and Data-Centric Computing, Berlin, Germany
| | - D Baum
- Zuse Institute Berlin, Department of Visual and Data-Centric Computing, Berlin, Germany
| | - Kai Ludwig
- Freie Universität Berlin, Institut für Chemie and Biochemie, Forschungszentrum für Elektronenmikroskopie, Berlin, Germany
| | - M Heine
- Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University Mainz, Mainz, Germany
| | - T Mielke
- Max Planck Institute for Molecular Genetics, Berlin, Microscopy and Cryo-Electron Microscopy Service Group, Berlin, Germany
| | - F Liu
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Campus Berlin-Buch, Berlin, Germany
| | - A M Walter
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Campus Berlin-Buch, Berlin, Germany
- University of Copenhagen, Department of Neuroscience, Copenhagen, Denmark
| | - M C Wahl
- Freie Universität Berlin, Institute of Chemistry and Biochemistry/Structural Biochemistry, Berlin, Germany
| | - S J Sigrist
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany.
- Charité Universitätsmedizin, NeuroCure Cluster of Excellence, Charitéplatz, Berlin, Germany.
| |
Collapse
|
25
|
Wu Z, Pang L, Ding M. CFI-1 functions unilaterally to restrict gap junction formation in C. elegans. Development 2025; 152:dev202955. [PMID: 39679967 PMCID: PMC11829774 DOI: 10.1242/dev.202955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Accepted: 11/08/2024] [Indexed: 12/17/2024]
Abstract
Electrical coupling is vital to neural communication, facilitating synchronized activity among neurons. Despite its significance, the precise mechanisms governing the establishment of gap junction connections between specific neurons remain elusive. Here, we identified that the PVC interneuron in Caenorhabditis elegans forms gap junction connections with the PVR interneuron. The transcriptional regulator CFI-1 (ARID3) is specifically expressed in the PVC but not PVR interneuron. Reducing cfi-1 expression in the PVC interneuron leads to enhanced gap junction formation in the PVR neuron, while ectopic expression of cfi-1 in the PVR neuron restores the proper level of gap junction connections in the PVC neuron, along with the normal touch response. These findings unveil the pivotal role of CFI-1 in bidirectionally regulating the formation of gap junctions within a specific neuronal pair, shedding light on the intricate molecular mechanisms governing neuronal connectivity in vivo.
Collapse
Affiliation(s)
- Zan Wu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lin Pang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mei Ding
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
26
|
Duan D, Koleske AJ. Phase separation of microtubule-binding proteins - implications for neuronal function and disease. J Cell Sci 2024; 137:jcs263470. [PMID: 39679446 PMCID: PMC11795294 DOI: 10.1242/jcs.263470] [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: 12/17/2024] Open
Abstract
Protein liquid-liquid phase separation (LLPS) is driven by intrinsically disordered regions and multivalent binding domains, both of which are common features of diverse microtubule (MT) regulators. Many in vitro studies have dissected the mechanisms by which MT-binding proteins (MBPs) regulate MT nucleation, stabilization and dynamics, and investigated whether LLPS plays a role in these processes. However, more recent in vivo studies have focused on how MBP LLPS affects biological functions throughout neuronal development. Dysregulation of MBP LLPS can lead to formation of aggregates - an underlying feature in many neurodegenerative diseases - such as the tau neurofibrillary tangles present in Alzheimer's disease. In this Review, we highlight progress towards understanding the regulation of MT dynamics through the lens of phase separation of MBPs and associated cytoskeletal regulators, from both in vitro and in vivo studies. We also discuss how LLPS of MBPs regulates neuronal development and maintains homeostasis in mature neurons.
Collapse
Affiliation(s)
- Daisy Duan
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06510, USA
| | - Anthony J. Koleske
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06510, USA
- Department of Neuroscience, Yale University, New Haven, CT 06510, USA
| |
Collapse
|
27
|
Kostović I. Development of the basic architecture of neocortical circuitry in the human fetus as revealed by the coupling spatiotemporal pattern of synaptogenesis along with microstructure and macroscale in vivo MR imaging. Brain Struct Funct 2024; 229:2339-2367. [PMID: 39102068 PMCID: PMC11612014 DOI: 10.1007/s00429-024-02838-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 07/12/2024] [Indexed: 08/06/2024]
Abstract
In humans, a quantifiable number of cortical synapses appears early in fetal life. In this paper, we present a bridge across different scales of resolution and the distribution of synapses across the transient cytoarchitectonic compartments: marginal zone (MZ), cortical plate (CP), subplate (SP), and in vivo MR images. The tissue of somatosensory cortex (7-26 postconceptional weeks (PCW)) was prepared for electron microscopy, and classified synapses with a determined subpial depth were used for creating histograms matched to the histological sections immunoreacted for synaptic markers and aligned to in vivo MR images (1.5 T) of corresponding fetal ages (maternal indication). Two time periods and laminar patterns of synaptogenesis were identified: an early and midfetal two-compartmental distribution (MZ and SP) and a late fetal three-compartmental distribution (CP synaptogenesis). During both periods, a voluminous, synapse-rich SP was visualized on the in vivo MR. Another novel finding concerns the phase of secondary expansion of the SP (13 PCW), where a quantifiable number of synapses appears in the upper SP. This lamina shows a T2 intermediate signal intensity below the low signal CP. In conclusion, the early fetal appearance of synapses shows early differentiation of putative genetic mechanisms underlying the synthesis, transport and assembly of synaptic proteins. "Pioneering" synapses are likely to play a morphogenetic role in constructing of fundamental circuitry architecture due to interaction between neurons. They underlie spontaneous, evoked, and resting state activity prior to ex utero experience. Synapses can also mediate genetic and environmental triggers, adversely altering the development of cortical circuitry and leading to neurodevelopmental disorders.
Collapse
Affiliation(s)
- Ivica Kostović
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia.
| |
Collapse
|
28
|
Tod P, Varga A, Román V, Lendvai B, Pálkovács R, Sperlágh B, Vizi ES. Tetrabenazine, a vesicular monoamine transporter 2 inhibitor, inhibits vesicular storage capacity and release of monoamine transmitters in mouse brain tissue. Br J Pharmacol 2024; 181:5094-5109. [PMID: 39304979 DOI: 10.1111/bph.17348] [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: 08/04/2023] [Revised: 07/10/2024] [Accepted: 08/23/2024] [Indexed: 11/22/2024] Open
Abstract
BACKGROUND AND PURPOSE Tetrabenazine (TBZ), used for treating hyperkinetic disorders, inhibits vesicular monoamine transporter-2 (VMAT-2), which sequesters monoamines into vesicles for exocytosis. However, our knowledge of the effect of TBZ on monoaminergic transmission is limited. Herein, we provide neurochemical evidence regarding the effect of VMAT-2 inhibition on vesicular neurotransmitter release from the prefrontal cortex (PFC) and striatum (STR) (brain regions involved in characteristic TBZ treatment side effects). The interaction between TBZ and MDMA was also assessed regarding motor behaviour in mice. EXPERIMENTAL APPROACH Vesicular storage capacity and release of [3H]-noradrenaline ([3H]-NA), [3H]-dopamine ([3H]-DA), [3H]-serotonin ([3H]-5-HT), and [3H]-acetylcholine ([3H]-ACh) was studied in mouse PFC and STR ex vivo slice preparations using electrical field stimulation. Additionally, locomotor activity was assessed in vehicle-treated mice and compared with that of MDMA, TBZ, and co-administered animals (n = 6) using the LABORAS system. KEY RESULTS TBZ lowered the storage capacity and inhibited the vesicular release of [3H]-NA and [3H]-DA from the PFC, and [3H]-DA and [3H]-5-HT from the STR in a concentration-dependent manner. Unlike vesamicol (vesicular ACh uptake inhibitor), TBZ failed to inhibit the vesicular release of [3H]-ACh from the PFC. When the vesicular storage of the investigated monoamines was inhibited by TBZ in the PFC and STR, MDMA induced the release of transmitters through transporter reversal; MDMA dose dependently increased locomotor activity in vivo. CONCLUSION AND IMPLICATIONS Our observations provide neurochemical evidence explaining the mechanism of VMAT-2 inhibitors in the brain and support the involvement of dopaminergic and noradrenergic transmission in hyperkinetic movement disorders.
Collapse
Affiliation(s)
- Pál Tod
- Laboratory of Molecular Pharmacology, HUN-REN Institute of Experimental Medicine, Budapest, Hungary
| | - Anita Varga
- Pharmacology and Drug Safety Research, Gedeon Richter Plc, Budapest, Hungary
| | - Viktor Román
- Pharmacology and Drug Safety Research, Gedeon Richter Plc, Budapest, Hungary
| | - Balázs Lendvai
- Pharmacology and Drug Safety Research, Gedeon Richter Plc, Budapest, Hungary
| | - Roland Pálkovács
- Pharmacology and Drug Safety Research, Gedeon Richter Plc, Budapest, Hungary
| | - Beáta Sperlágh
- Laboratory of Molecular Pharmacology, HUN-REN Institute of Experimental Medicine, Budapest, Hungary
| | - E Sylvester Vizi
- Laboratory of Molecular Pharmacology, HUN-REN Institute of Experimental Medicine, Budapest, Hungary
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| |
Collapse
|
29
|
Özçete ÖD, Banerjee A, Kaeser PS. Mechanisms of neuromodulatory volume transmission. Mol Psychiatry 2024; 29:3680-3693. [PMID: 38789677 PMCID: PMC11540752 DOI: 10.1038/s41380-024-02608-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 05/07/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024]
Abstract
A wealth of neuromodulatory transmitters regulate synaptic circuits in the brain. Their mode of signaling, often called volume transmission, differs from classical synaptic transmission in important ways. In synaptic transmission, vesicles rapidly fuse in response to action potentials and release their transmitter content. The transmitters are then sensed by nearby receptors on select target cells with minimal delay. Signal transmission is restricted to synaptic contacts and typically occurs within ~1 ms. Volume transmission doesn't rely on synaptic contact sites and is the main mode of monoamines and neuropeptides, important neuromodulators in the brain. It is less precise than synaptic transmission, and the underlying molecular mechanisms and spatiotemporal scales are often not well understood. Here, we review literature on mechanisms of volume transmission and raise scientific questions that should be addressed in the years ahead. We define five domains by which volume transmission systems can differ from synaptic transmission and from one another. These domains are (1) innervation patterns and firing properties, (2) transmitter synthesis and loading into different types of vesicles, (3) architecture and distribution of release sites, (4) transmitter diffusion, degradation, and reuptake, and (5) receptor types and their positioning on target cells. We discuss these five domains for dopamine, a well-studied monoamine, and then compare the literature on dopamine with that on norepinephrine and serotonin. We include assessments of neuropeptide signaling and of central acetylcholine transmission. Through this review, we provide a molecular and cellular framework for volume transmission. This mechanistic knowledge is essential to define how neuromodulatory systems control behavior in health and disease and to understand how they are modulated by medical treatments and by drugs of abuse.
Collapse
Affiliation(s)
- Özge D Özçete
- Department of Neurobiology, Harvard Medical School, Boston, MA, 02115, USA
| | - Aditi Banerjee
- Department of Neurobiology, Harvard Medical School, Boston, MA, 02115, USA
| | - Pascal S Kaeser
- Department of Neurobiology, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
30
|
Miki T, Okamoto Y, Ueno-Umegai M, Toyofuku R, Hattori S, Sakaba T. Single-vesicle imaging reveals actin-dependent spatial restriction of vesicles at the active zone, essential for sustained transmission. Proc Natl Acad Sci U S A 2024; 121:e2402152121. [PMID: 39405348 PMCID: PMC11513904 DOI: 10.1073/pnas.2402152121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 09/10/2024] [Indexed: 10/25/2024] Open
Abstract
Synaptic-vesicle (SV) recruitment is thought to maintain reliable neurotransmitter release during high-frequency signaling. However, the mechanism underlying the SV reloading for sustained neurotransmission at central synapses remains unknown. To elucidate this, we performed direct observations of SV reloading and mobility at a single-vesicle level near the plasma membrane in cerebellar mossy fiber terminals using total internal reflection fluorescence microscopy, together with simultaneous recordings of membrane fusion by capacitance measurements. We found that actin disruption abolished the rapid SV recruitment and reduced sustained release. In contrast, induction of actin polymerization and stabilization did not affect vesicle recruitment and release, suggesting that the presence of actin filaments, rather than actin dynamics, was required for the rapid recruitment. Single-particle tracking experiments of quantum dot-labeled vesicles, which allows nanoscale resolution of vesicle mobility, revealed that actin disruption caused vesicles to diffuse more rapidly. Hidden Markov modeling with Bayesian inference revealed that SVs had two diffusion states under normal conditions: free-diffusing and trapped. After disruption of the actin filament, vesicles tended to have only the free-diffusing state. F-actin staining showed that actin filaments were localized outside the active zones (AZs) and surrounded some SV trajectories. Perturbation of SV mobility, possibly through interference with biomolecular condensates, also suggested that the restricted diffusion state determined the rate of SV recruitment. We propose that actin filaments confined SVs near the AZ to achieve rapid and efficient recruitment followed by priming and sustained synaptic transmission.
Collapse
Affiliation(s)
- Takafumi Miki
- Department of Cell Physiology, Graduate School of Medicine, Akita University, Akita010-8543, Japan
- Laboratory of Molecular Synaptic Function, Graduate School of Brain Science, Doshisha University, Kyoto610-0394, Japan
| | - Yuji Okamoto
- Department of Cell Physiology, Graduate School of Medicine, Akita University, Akita010-8543, Japan
| | | | - Rio Toyofuku
- Laboratory of Molecular Synaptic Function, Graduate School of Brain Science, Doshisha University, Kyoto610-0394, Japan
| | - Shun Hattori
- Department of Electronic Systems Engineering, Faculty of Advanced Engineering, The University of Shiga Prefecture, Hikone522-8533, Japan
| | - Takeshi Sakaba
- Laboratory of Molecular Synaptic Function, Graduate School of Brain Science, Doshisha University, Kyoto610-0394, Japan
| |
Collapse
|
31
|
Li Y, Badawi Y, Meriney SD. Age-Related Homeostatic Plasticity at Rodent Neuromuscular Junctions. Cells 2024; 13:1684. [PMID: 39451202 PMCID: PMC11506802 DOI: 10.3390/cells13201684] [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: 09/04/2024] [Revised: 10/03/2024] [Accepted: 10/08/2024] [Indexed: 10/26/2024] Open
Abstract
Motor ability decline remains a major threat to the quality of life of the elderly. Although the later stages of aging co-exist with degenerative pathologies, the long process of aging is more complicated than a simple and gradual degeneration. To combat senescence and the associated late-stage degeneration of the neuromuscular system, it is imperative to examine changes that occur during the long process of aging. Prior to late-stage degeneration, age-induced changes in the neuromuscular system trigger homeostatic plasticity. This unique phenomenon may be important for the maintenance of the neuromuscular system during the early stages of aging. In this review, we will focus on age-induced changes in neurotransmission at the neuromuscular junction, providing the potential mechanisms responsible for these changes. The goal is to highlight these key elements and their role in regulating neurotransmission, facilitating future research efforts to combat late-stage degeneration in the neuromuscular system by preserving the functional and structural integrity of these elements prior to the late stage of aging.
Collapse
Affiliation(s)
| | | | - Stephen D. Meriney
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA 15260, USA; (Y.L.); (Y.B.)
| |
Collapse
|
32
|
Slater CR. Neuromuscular Transmission in a Biological Context. Compr Physiol 2024; 14:5641-5702. [PMID: 39382166 DOI: 10.1002/cphy.c240001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/10/2024]
Abstract
Neuromuscular transmission is the process by which motor neurons activate muscle contraction and thus plays an essential role in generating the purposeful body movements that aid survival. While many features of this process are common throughout the Animal Kingdom, such as the release of transmitter in multimolecular "quanta," and the response to it by opening ligand-gated postsynaptic ion channels, there is also much diversity between and within species. Much of this diversity is associated with specialization for either slow, sustained movements such as maintain posture or fast but brief movements used during escape or prey capture. In invertebrates, with hydrostatic and exoskeletons, most motor neurons evoke graded depolarizations of the muscle which cause graded muscle contractions. By contrast, vertebrate motor neurons trigger action potentials in the muscle fibers which give rise to all-or-none contractions. The properties of neuromuscular transmission, in particular the intensity and persistence of transmitter release, reflect these differences. Neuromuscular transmission varies both between and within individual animals, which often have distinct tonic and phasic subsystems. Adaptive plasticity of neuromuscular transmission, on a range of time scales, occurs in many species. This article describes the main steps in neuromuscular transmission and how they vary in a number of "model" species, including C. elegans , Drosophila , zebrafish, mice, and humans. © 2024 American Physiological Society. Compr Physiol 14:5641-5702, 2024.
Collapse
|
33
|
Hilton BJ, Griffin JM, Fawcett JW, Bradke F. Neuronal maturation and axon regeneration: unfixing circuitry to enable repair. Nat Rev Neurosci 2024; 25:649-667. [PMID: 39164450 DOI: 10.1038/s41583-024-00849-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/19/2024] [Indexed: 08/22/2024]
Abstract
Mammalian neurons lose the ability to regenerate their central nervous system axons as they mature during embryonic or early postnatal development. Neuronal maturation requires a transformation from a situation in which neuronal components grow and assemble to one in which these components are fixed and involved in the machinery for effective information transmission and computation. To regenerate after injury, neurons need to overcome this fixed state to reactivate their growth programme. A variety of intracellular processes involved in initiating or sustaining neuronal maturation, including the regulation of gene expression, cytoskeletal restructuring and shifts in intracellular trafficking, have been shown to prevent axon regeneration. Understanding these processes will contribute to the identification of targets to promote repair after injury or disease.
Collapse
Affiliation(s)
- Brett J Hilton
- Department of Cellular and Physiological Sciences, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada.
- International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, British Columbia, Canada.
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada.
| | - Jarred M Griffin
- Laboratory for Axonal Growth and Regeneration, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - James W Fawcett
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, University of Cambridge, Cambridge, UK.
- Centre for Reconstructive Neuroscience, Institute for Experimental Medicine Czech Academy of Science (CAS), Prague, Czechia.
| | - Frank Bradke
- Laboratory for Axonal Growth and Regeneration, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.
| |
Collapse
|
34
|
Timalsina B, Lee S, Kaang BK. Advances in the labelling and selective manipulation of synapses. Nat Rev Neurosci 2024; 25:668-687. [PMID: 39174832 DOI: 10.1038/s41583-024-00851-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/23/2024] [Indexed: 08/24/2024]
Abstract
Synapses are highly specialized neuronal structures that are essential for neurotransmission, and they are dynamically regulated throughout the lifetime. Although accumulating evidence indicates that these structures are crucial for information processing and storage in the brain, their precise roles beyond neurotransmission are yet to be fully appreciated. Genetically encoded fluorescent tools have deepened our understanding of synaptic structure and function, but developing an ideal methodology to selectively visualize, label and manipulate synapses remains challenging. Here, we provide an overview of currently available synapse labelling techniques and describe their extension to enable synapse manipulation. We categorize these approaches on the basis of their conceptual bases and target molecules, compare their advantages and limitations and propose potential modifications to improve their effectiveness. These methods have broad utility, particularly for investigating mechanisms of synaptic function and synaptopathy.
Collapse
Affiliation(s)
- Binod Timalsina
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon, South Korea
| | - Sangkyu Lee
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon, South Korea
| | - Bong-Kiun Kaang
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon, South Korea.
| |
Collapse
|
35
|
Guo A, Wu Q, Yan X, Chen K, Liu Y, Liang D, Yang Y, Luo Q, Xiong M, Yu Y, Fei E, Chen F. Differential roles of lysosomal cholesterol transporters in the development of C. elegans NMJs. Life Sci Alliance 2024; 7:e202402584. [PMID: 39084875 PMCID: PMC11291935 DOI: 10.26508/lsa.202402584] [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: 01/10/2024] [Revised: 07/21/2024] [Accepted: 07/22/2024] [Indexed: 08/02/2024] Open
Abstract
Cholesterol homeostasis in neurons is critical for synapse formation and maintenance. Neurons with impaired cholesterol uptake undergo progressive synapse loss and eventual degeneration. To investigate the molecular mechanisms of neuronal cholesterol homeostasis and its role during synapse development, we studied motor neurons of Caenorhabditis elegans because these neurons rely on dietary cholesterol. Combining lipidomic analysis, we discovered that NCR-1, a lysosomal cholesterol transporter, promotes cholesterol absorption and synapse development. Loss of ncr-1 causes smaller synapses, and low cholesterol exacerbates the deficits. Moreover, NCR-1 deficiency hinders the increase in synapses under high cholesterol. Unexpectedly, NCR-2, the NCR-1 homolog, increases the use of cholesterol and sphingomyelins and impedes synapse formation. NCR-2 deficiency causes an increase in synapses regardless of cholesterol concentration. Inhibiting the degradation or synthesis of sphingomyelins can induce or suppress the synaptic phenotypes in ncr-2 mutants. Our findings indicate that neuronal cholesterol homeostasis is differentially controlled by two lysosomal cholesterol transporters and highlight the importance of neuronal cholesterol homeostasis in synapse development.
Collapse
Affiliation(s)
- Amin Guo
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Qi Wu
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Xin Yan
- School of Life Sciences, Nanchang University, Nanchang, China
| | - Kanghua Chen
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yuxiang Liu
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Dingfa Liang
- Queen Mary School of Nanchang University, Jiangxi Medical College, Nanchang, China
| | - Yuxiao Yang
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Qunfeng Luo
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Mingtao Xiong
- Institute of Biomedical Innovation, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yong Yu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Erkang Fei
- Institute of Biomedical Innovation, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Fei Chen
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| |
Collapse
|
36
|
Mohammadkhani A, Qiao M, Borgland SL. Distinct Neuromodulatory Effects of Endogenous Orexin and Dynorphin Corelease on Projection-Defined Ventral Tegmental Dopamine Neurons. J Neurosci 2024; 44:e0682242024. [PMID: 39187377 PMCID: PMC11426376 DOI: 10.1523/jneurosci.0682-24.2024] [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: 04/09/2024] [Revised: 08/11/2024] [Accepted: 08/16/2024] [Indexed: 08/28/2024] Open
Abstract
Dopamine (DA) neurons in the ventral tegmental area (VTA) respond to motivationally relevant cues, and circuit-specific signaling drives different aspects of motivated behavior. Orexin (ox; also known as hypocretin) and dynorphin (dyn) are coexpressed lateral hypothalamic (LH) neuropeptides that project to the VTA. These peptides have opposing effects on the firing activity of VTADA neurons via orexin 1 (Ox1R) or kappa opioid (KOR) receptors. Given that Ox1R activation increases VTADA firing, and KOR decreases firing, it is unclear how the coreleased peptides contribute to the net activity of DA neurons. We tested if optical stimulation of LHox/dyn neuromodulates VTADA neuronal activity via peptide release and if the effects of optically driven LHox/dyn release segregate based on VTADA projection targets including the basolateral amygdala (BLA) or the lateral or medial shell of the nucleus accumbens (lAcbSh, mAchSh). Using a combination of circuit tracing, optogenetics, and patch-clamp electrophysiology in male and female orexincre mice, we showed a diverse response of LHox/dyn optical stimulation on VTADA neuronal firing, which is not mediated by fast transmitter release and is blocked by antagonists to KOR and Ox1R signaling. Additionally, where optical stimulation of LHox/dyn inputs in the VTA inhibited firing of the majority of BLA-projecting VTADA neurons, optical stimulation of LHox/dyn inputs in the VTA bidirectionally affects firing of either lAcbSh- or mAchSh-projecting VTADA neurons. These findings indicate that LHox/dyn corelease may influence the output of the VTA by balancing ensembles of neurons within each population which contribute to different aspects of reward seeking.
Collapse
Affiliation(s)
- Aida Mohammadkhani
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, The University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Min Qiao
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, The University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Stephanie L Borgland
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, The University of Calgary, Calgary, AB T2N 4N1, Canada
| |
Collapse
|
37
|
Marshall-Phelps KL, Almeida R. Axonal neurotransmitter release in the regulation of myelination. Biosci Rep 2024; 44:BSR20231616. [PMID: 39230890 PMCID: PMC11427734 DOI: 10.1042/bsr20231616] [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: 04/24/2024] [Revised: 08/30/2024] [Accepted: 09/04/2024] [Indexed: 09/05/2024] Open
Abstract
Myelination of axons is a key determinant of fast action potential propagation, axonal health and circuit function. Previously considered a static structure, it is now clear that myelin is dynamically regulated in response to neuronal activity in the central nervous system (CNS). However, how activity-dependent signals are conveyed to oligodendrocytes remains unclear. Here, we review the potential mechanisms by which neurons could communicate changing activity levels to myelin, with a focus on the accumulating body of evidence to support activity-dependent vesicular signalling directly onto myelin sheaths. We discuss recent in vivo findings of activity-dependent fusion of neurotransmitter vesicles from non-synaptic axonal sites, and how modulation of this vesicular fusion regulates the stability and growth of myelin sheaths. We also consider the potential mechanisms by which myelin could sense and respond to axon-derived signals to initiate remodelling, and the relevance of these adaptations for circuit function. We propose that axonal vesicular signalling represents an important and underappreciated mode of communication by which neurons can transmit activity-regulated signals to myelinating oligodendrocytes and, potentially, more broadly to other cell types in the CNS.
Collapse
Affiliation(s)
- Katy L.H. Marshall-Phelps
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, U.K
- MS Society Edinburgh Centre for MS Research, University of Edinburgh, Edinburgh, U.K
| | - Rafael G. Almeida
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, U.K
- MS Society Edinburgh Centre for MS Research, University of Edinburgh, Edinburgh, U.K
| |
Collapse
|
38
|
Glausier JR, Bouchet-Marquis C, Maier M, Banks-Tibbs T, Wu K, Ning J, Melchitzky D, Lewis DA, Freyberg Z. Volume electron microscopy reveals 3D synaptic nanoarchitecture in postmortem human prefrontal cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.26.582174. [PMID: 38463986 PMCID: PMC10925168 DOI: 10.1101/2024.02.26.582174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Synaptic function is directly reflected in quantifiable ultrastructural features using electron microscopy (EM) approaches. This coupling of synaptic function and ultrastructure suggests that in vivo synaptic function can be inferred from EM analysis of ex vivo human brain tissue. To investigate this, we employed focused ion beam-scanning electron microscopy (FIB-SEM), a volume EM (VEM) approach, to generate ultrafine-resolution, three-dimensional (3D) micrographic datasets of postmortem human dorsolateral prefrontal cortex (DLPFC), a region with cytoarchitectonic characteristics distinct to human brain. Synaptic, sub-synaptic, and organelle measures were highly consistent with findings from experimental models that are free from antemortem or postmortem effects. Further, 3D neuropil reconstruction revealed a unique, ultrastructurally-complex, spiny dendritic shaft that exhibited features characteristic of heightened synaptic communication, integration, and plasticity. Altogether, our findings provide critical proof-of-concept data demonstrating that ex vivo VEM analysis is an effective approach to infer in vivo synaptic functioning in human brain.
Collapse
Affiliation(s)
- Jill R. Glausier
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA
| | | | - Matthew Maier
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA
| | - Tabitha Banks-Tibbs
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA
- College of Medicine, The Ohio State University, Columbus, OH
| | - Ken Wu
- Materials and Structural Analysis, Thermo Fisher Scientific, Hillsboro, OR
| | - Jiying Ning
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA
| | | | - David A. Lewis
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA
| | - Zachary Freyberg
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
39
|
Kim DI, Park S, Park S, Ye M, Chen JY, Kang SJ, Jhang J, Hunker AC, Zweifel LS, Caron KM, Vaughan JM, Saghatelian A, Palmiter RD, Han S. Presynaptic sensor and silencer of peptidergic transmission reveal neuropeptides as primary transmitters in pontine fear circuit. Cell 2024; 187:5102-5117.e16. [PMID: 39043179 PMCID: PMC11380597 DOI: 10.1016/j.cell.2024.06.035] [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: 01/13/2023] [Revised: 11/17/2023] [Accepted: 06/25/2024] [Indexed: 07/25/2024]
Abstract
Neurons produce and release neuropeptides to communicate with one another. Despite their importance in brain function, circuit-based mechanisms of peptidergic transmission are poorly understood, primarily due to the lack of tools for monitoring and manipulating neuropeptide release in vivo. Here, we report the development of two genetically encoded tools for investigating peptidergic transmission in behaving mice: a genetically encoded large dense core vesicle (LDCV) sensor that detects presynaptic neuropeptide release and a genetically encoded silencer that specifically degrades neuropeptides inside LDCVs. Using these tools, we show that neuropeptides, not glutamate, encode the unconditioned stimulus in the parabrachial-to-amygdalar threat pathway during Pavlovian threat learning. We also show that neuropeptides play important roles in encoding positive valence and suppressing conditioned threat response in the amygdala-to-parabrachial endogenous opioidergic circuit. These results show that our sensor and silencer for presynaptic peptidergic transmission are reliable tools to investigate neuropeptidergic systems in awake, behaving animals.
Collapse
Affiliation(s)
- Dong-Il Kim
- Peptide Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Sekun Park
- Howard Hughes Medical Institute, Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Seahyung Park
- Peptide Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Mao Ye
- Peptide Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Jane Y Chen
- Howard Hughes Medical Institute, Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Sukjae J Kang
- Peptide Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Jinho Jhang
- Peptide Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Avery C Hunker
- Department of Pharmacology, University of Washington, Seattle, WA 98195, USA
| | - Larry S Zweifel
- Department of Pharmacology, University of Washington, Seattle, WA 98195, USA
| | - Kathleen M Caron
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Joan M Vaughan
- Peptide Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Alan Saghatelian
- Peptide Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Richard D Palmiter
- Howard Hughes Medical Institute, Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Sung Han
- Peptide Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA; Center for Neuroscience Imaging Research, Institute for Basic Science, Suwon 16419, Republic of Korea; Department of Biomedical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| |
Collapse
|
40
|
Emperador-Melero J, Andersen JW, Metzbower SR, Levy AD, Dharmasri PA, de Nola G, Blanpied TA, Kaeser PS. Distinct active zone protein machineries mediate Ca 2+ channel clustering and vesicle priming at hippocampal synapses. Nat Neurosci 2024; 27:1680-1694. [PMID: 39160372 PMCID: PMC11682530 DOI: 10.1038/s41593-024-01720-5] [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: 10/27/2023] [Accepted: 06/28/2024] [Indexed: 08/21/2024]
Abstract
Action potentials trigger neurotransmitter release at the presynaptic active zone with spatiotemporal precision. This is supported by protein machinery that mediates synaptic vesicle priming and clustering of CaV2 Ca2+ channels nearby. One model posits that scaffolding proteins directly tether vesicles to CaV2s; however, here we find that at mouse hippocampal synapses, CaV2 clustering and vesicle priming are executed by separate machineries. CaV2 nanoclusters are positioned at variable distances from those of the priming protein Munc13. The active zone organizer RIM anchors both proteins but distinct interaction motifs independently execute these functions. In transfected cells, Liprin-α and RIM form co-assemblies that are separate from CaV2-organizing complexes. At synapses, Liprin-α1-Liprin-α4 knockout impairs vesicle priming but not CaV2 clustering. The cell adhesion protein PTPσ recruits Liprin-α, RIM and Munc13 into priming complexes without co-clustering CaV2s. We conclude that active zones consist of distinct machineries to organize CaV2s and prime vesicles, and Liprin-α and PTPσ specifically support priming site assembly.
Collapse
Affiliation(s)
| | | | - Sarah R Metzbower
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Aaron D Levy
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Poorna A Dharmasri
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Giovanni de Nola
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Thomas A Blanpied
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Pascal S Kaeser
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
41
|
Sermon JJ, Wiest C, Tan H, Denison T, Duchet B. Evoked resonant neural activity long-term dynamics can be reproduced by a computational model with vesicle depletion. Neurobiol Dis 2024; 199:106565. [PMID: 38880431 PMCID: PMC11300885 DOI: 10.1016/j.nbd.2024.106565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 06/04/2024] [Accepted: 06/11/2024] [Indexed: 06/18/2024] Open
Abstract
Subthalamic deep brain stimulation (DBS) robustly generates high-frequency oscillations known as evoked resonant neural activity (ERNA). Recently the importance of ERNA has been demonstrated through its ability to predict the optimal DBS contact in the subthalamic nucleus in patients with Parkinson's disease. However, the underlying mechanisms of ERNA are not well understood, and previous modelling efforts have not managed to reproduce the wealth of published data describing the dynamics of ERNA. Here, we aim to present a minimal model capable of reproducing the characteristics of the slow ERNA dynamics published to date. We make biophysically-motivated modifications to the Kuramoto model and fit its parameters to the slow dynamics of ERNA obtained from data. Our results demonstrate that it is possible to reproduce the slow dynamics of ERNA (over hundreds of seconds) with a single neuronal population, and, crucially, with vesicle depletion as one of the key mechanisms behind the ERNA frequency decay in our model. We further validate the proposed model against experimental data from Parkinson's disease patients, where it captures the variations in ERNA frequency and amplitude in response to variable stimulation frequency, amplitude, and to stimulation pulse bursting. We provide a series of predictions from the model that could be the subject of future studies for further validation.
Collapse
Affiliation(s)
- James J Sermon
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK; MRC Brain Networks Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Christoph Wiest
- MRC Brain Networks Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Huiling Tan
- MRC Brain Networks Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Timothy Denison
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK; MRC Brain Networks Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Benoit Duchet
- MRC Brain Networks Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK.
| |
Collapse
|
42
|
Hoagland A, Newman ZL, Cai Z, Isacoff EY. Circuit firing homeostasis following synaptic perturbation ensures robust behavior. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.27.609984. [PMID: 39253468 PMCID: PMC11383027 DOI: 10.1101/2024.08.27.609984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
Homeostatic regulation of excitability and synaptic transmission ensures stable neural circuit output under changing conditions. We find that pre- or postsynaptic weakening of motor neuron (MN) to muscle glutamatergic transmission in Drosophila larva has little impact on locomotion, suggesting non-synaptic compensatory mechanisms. In vivo imaging of MN to muscle synaptic transmission and MN activity both show that synaptic weakening increases activity in tonic type Ib MNs, but not in the phasic type Is MN that innervate the same muscles. Additionally, an inhibitory class of pre-MNs that innervates type Ib-but not Is-MNs decreases activity. Our experiments suggest that weakening of MN evoked synaptic release onto the muscle is compensated for by an increase in MN firing due to a combined cell-autonomous increase in excitability and decreased inhibitory central drive. Selectivity for type Ib MNs may serve to restore tonic drive while absence of firing adjustment in the convergent Is MN can maintain the contraction wave dynamics needed for locomotion.
Collapse
|
43
|
Gao C, Xiong R, Zhang ZY, Peng H, Gu YK, Xu W, Yang WT, Liu Y, Gao J, Yin Y. Hybrid nanostructures for neurodegenerative disease theranostics: the art in the combination of biomembrane and non-biomembrane nanostructures. Transl Neurodegener 2024; 13:43. [PMID: 39192378 DOI: 10.1186/s40035-024-00436-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 08/07/2024] [Indexed: 08/29/2024] Open
Abstract
The diagnosis of neurodegenerative diseases (NDDs) remains challenging, and existing therapeutic approaches demonstrate little efficacy. NDD drug delivery can be achieved through the utilization of nanostructures, hence enabling multimodal NDD theranostics. Nevertheless, both biomembrane and non-biomembrane nanostructures possess intrinsic shortcomings that must be addressed by hybridization to create novel nanostructures with versatile applications in NDD theranostics. Hybrid nanostructures display improved biocompatibility, inherent targeting capabilities, intelligent responsiveness, and controlled drug release. This paper provides a concise overview of the latest developments in hybrid nanostructures for NDD theranostics and emphasizes various engineering methodologies for the integration of diverse nanostructures, including liposomes, exosomes, cell membranes, and non-biomembrane nanostructures such as polymers, metals, and hydrogels. The use of a combination technique can significantly augment the precision, intelligence, and efficacy of hybrid nanostructures, therefore functioning as a more robust theranostic approach for NDDs. This paper also addresses the issues that arise in the therapeutic translation of hybrid nanostructures and explores potential future prospects in this field.
Collapse
Affiliation(s)
- Chao Gao
- Department of Neurology, Second Affiliated Hospital (Shanghai Changzheng Hospital) of Naval Medical University, Shanghai, 200003, China
| | - Ran Xiong
- Department of Neurology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Zhi-Yu Zhang
- Department of Health Management, Second Affliated Hospital (Shanghai Changzheng Hospital) of Naval Medical University, Shanghai, 200003, China
| | - Hua Peng
- Department of Neurology, Second Affiliated Hospital (Shanghai Changzheng Hospital) of Naval Medical University, Shanghai, 200003, China
| | - Yuan-Kai Gu
- Department of Neurology, Second Affiliated Hospital (Shanghai Changzheng Hospital) of Naval Medical University, Shanghai, 200003, China
| | - Wei Xu
- Department of Neurology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Wei-Ting Yang
- Department of Neurology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Yan Liu
- Department of Clinical Pharmacy, Xinhua Hospital, Clinical Pharmacy Innovation Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, 200000, China.
- Clinical Pharmacy Innovation Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| | - Jie Gao
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China.
- Shanghai Key Laboratory of Nautical Medicine and Translation of Drugs and Medical Devices, Shanghai, 200433, China.
| | - You Yin
- Department of Neurology, Second Affiliated Hospital (Shanghai Changzheng Hospital) of Naval Medical University, Shanghai, 200003, China.
- Department of Neurology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
| |
Collapse
|
44
|
Fesce R. Old innovations and shifted paradigms in cellular neuroscience. Front Cell Neurosci 2024; 18:1460219. [PMID: 39234031 PMCID: PMC11371623 DOI: 10.3389/fncel.2024.1460219] [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: 07/05/2024] [Accepted: 08/06/2024] [Indexed: 09/06/2024] Open
Abstract
Once upon a time the statistics of quantal release were fashionable: "n" available vesicles (fusion sites), each with probability "p" of releasing a quantum. The story was not so simple, a nice paradigm to be abandoned. Biophysicists, experimenting with "black films," explained the astonishing rapidity of spike-induced release: calcium can trigger the fusion of lipidic vesicles with a lipid bilayer, by masking the negative charges of the membranes. The idea passed away, buried by the discovery of NSF, SNAPs, SNARE proteins and synaptotagmin, Munc, RIM, complexin. Electrophysiology used to be a field for few adepts. Then came patch clamp, and multielectrode arrays and everybody became electrophysiologists. Now, optogenetics have blossomed, and the whole field has changed again. Nice surprise for me, when Alvarez de Toledo demonstrated that release of transmitters could occur through the transient opening of a pore between the vesicle and the plasma-membrane, no collapse of the vesicle in the membrane needed: my mentor Bruno Ceccarelli had cherished this idea ("kiss and run") and tried to prove it for 20 years. The most impressive developments have probably regarded IT, computers and all their applications; machine learning, AI, and the truly spectacular innovations in brain imaging, especially functional ones, have transformed cognitive neurosciences into a new extraordinarily prolific field, and certainly let us imagine that we may finally understand what is going on in our brains. Cellular neuroscience, on the other hand, though the large public has been much less aware of the incredible amount of information the scientific community has acquired on the cellular aspects of neuronal function, may indeed help us to eventually understand the mechanistic detail of how the brain work. But this is no more in the past, this is the future.
Collapse
Affiliation(s)
- Riccardo Fesce
- Department of Biomedical Sciences, Humanitas University Medical School, Pieve Emanuele, Italy
| |
Collapse
|
45
|
Mueller BD, Merrill SA, Von Diezmann L, Jorgensen EM. Using Localization Microscopy to Quantify Calcium Channels at Presynaptic Boutons. Bio Protoc 2024; 14:e5049. [PMID: 39210951 PMCID: PMC11349493 DOI: 10.21769/bioprotoc.5049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 07/07/2024] [Accepted: 07/11/2024] [Indexed: 09/04/2024] Open
Abstract
Calcium channels at synaptic boutons are critical for synaptic function, but their number and distribution are poorly understood. This gap in knowledge is primarily due to the resolution limits of fluorescence microscopy. In the last decade, the diffraction limit of light was surpassed, and fluorescent molecules can now be localized with nanometer precision. Concurrently, new gene editing strategies allowed direct tagging of the endogenous calcium channel genes-expressed in the correct cells and at physiological levels. Further, the repurposing of self-labeling enzymes to attach fluorescent dyes to proteins improved photon yields enabling efficient localization of single molecules. Here, we describe tagging strategies, localization microscopy, and data analysis for calcium channel localization. In this case, we are imaging calcium channels fused with SNAP or HALO tags in live anesthetized C. elegans nematodes, but the analysis is relevant for any super-resolution preparations. We describe how to process images into localizations and protein clusters into confined nanodomains. Finally, we discuss strategies for estimating the number of calcium channels present at synaptic boutons. Key features • Super-resolution imaging of live anesthetized C. elegans. • Three-color super-resolution reconstruction of synapses. • Nanodomains and the distribution of proteins. • Quantification of the number of proteins at synapses from single-molecule localization data.
Collapse
Affiliation(s)
- Brian D. Mueller
- School of Biological Sciences, University of Utah, Salt Lake City, UT, USA
- Howard Hughes Medical Institute, Salt Lake City, UT, USA
| | - Sean A. Merrill
- School of Biological Sciences, University of Utah, Salt Lake City, UT, USA
- Howard Hughes Medical Institute, Salt Lake City, UT, USA
| | - Lexy Von Diezmann
- Dept. of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN, USA
| | - Erik M. Jorgensen
- School of Biological Sciences, University of Utah, Salt Lake City, UT, USA
- Howard Hughes Medical Institute, Salt Lake City, UT, USA
| |
Collapse
|
46
|
Peng J, Liang D, Zhang Z. Palmitoylation of synaptic proteins: roles in functional regulation and pathogenesis of neurodegenerative diseases. Cell Mol Biol Lett 2024; 29:108. [PMID: 39127627 DOI: 10.1186/s11658-024-00625-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 07/30/2024] [Indexed: 08/12/2024] Open
Abstract
Palmitoylation is a type of lipid modification that plays an important role in various aspects of neuronal function. Over the past few decades, several studies have shown that the palmitoylation of synaptic proteins is involved in neurotransmission and synaptic functions. Palmitoyl acyltransferases (PATs), which belong to the DHHC family, are major players in the regulation of palmitoylation. Dysregulated palmitoylation of synaptic proteins and mutated/dysregulated DHHC proteins are associated with several neurodegenerative diseases, such as Alzheimer's disease (AD), Huntington's disease (HD), and Parkinson's disease (PD). In this review, we summarize the recent discoveries on the subcellular distribution of DHHC proteins and analyze their expression patterns in different brain cells. In particular, this review discusses how palmitoylation of synaptic proteins regulates synaptic vesicle exocytotic fusion and the localization, clustering, and transport of several postsynaptic receptors, as well as the role of palmitoylation of other proteins in regulating synaptic proteins. Additionally, some of the specific known associations of these factors with neurodegenerative disorders are explored, with a few suggestions for the development of therapeutic strategies. Finally, this review provides possible directions for future research to reveal detailed and specific mechanisms underlying the roles of synaptic protein palmitoylation.
Collapse
Affiliation(s)
- Jiaying Peng
- Shenzhen Key Laboratory of Marine Bioresources and Ecology, Brain Disease and Big Data Research Institute, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Danchan Liang
- Shenzhen Key Laboratory of Marine Bioresources and Ecology, Brain Disease and Big Data Research Institute, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Zhonghao Zhang
- Shenzhen Key Laboratory of Marine Bioresources and Ecology, Brain Disease and Big Data Research Institute, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China.
- Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China.
| |
Collapse
|
47
|
Jevon D, Cottle L, Hallahan N, Harwood R, Samra JS, Gill AJ, Loudovaris T, Thomas HE, Thorn P. Capillary contact points determine beta cell polarity, control secretion and are disrupted in the db/db mouse model of diabetes. Diabetologia 2024; 67:1683-1697. [PMID: 38814445 PMCID: PMC11343897 DOI: 10.1007/s00125-024-06180-x] [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: 01/11/2024] [Accepted: 03/26/2024] [Indexed: 05/31/2024]
Abstract
AIMS/HYPOTHESIS Almost all beta cells contact one capillary and insulin granule fusion is targeted to this region. However, there are reports of beta cells contacting more than one capillary. We therefore set out to determine the proportion of beta cells with multiple contacts and the impact of this on cell structure and function. METHODS We used pancreatic slices in mice and humans to better maintain cell and islet structure than in isolated islets. Cell structure was assayed using immunofluorescence and 3D confocal microscopy. Live-cell two-photon microscopy was used to map granule fusion events in response to glucose stimulation. RESULTS We found that 36% and 22% of beta cells in islets from mice and humans, respectively, have separate contact with two capillaries. These contacts establish a distinct form of cell polarity with multiple basal regions. Both capillary contact points are enriched in presynaptic scaffold proteins, and both are a target for insulin granule fusion. Cells with two capillary contact points have a greater capillary contact area and secrete more, with analysis showing that, independent of the number of contact points, increased contact area is correlated with increased granule fusion. Using db/db mice as a model for type 2 diabetes, we observed changes in islet capillary organisation that significantly reduced total islet capillary surface area, and reduced area of capillary contact in single beta cells. CONCLUSIONS/INTERPRETATION Beta cells that contact two capillaries are a significant subpopulation of beta cells within the islet. They have a distinct form of cell polarity and both contact points are specialised for secretion. The larger capillary contact area of cells with two contact points is correlated with increased secretion. In the db/db mouse, changes in capillary structure impact beta cell capillary contact, implying that this is a new factor contributing to disease progression.
Collapse
Affiliation(s)
- Dillon Jevon
- Charles Perkins Centre, School of Medical Sciences, University of Sydney, Camperdown, NSW, Australia
| | - Louise Cottle
- Charles Perkins Centre, School of Medical Sciences, University of Sydney, Camperdown, NSW, Australia
| | - Nicole Hallahan
- Charles Perkins Centre, School of Medical Sciences, University of Sydney, Camperdown, NSW, Australia
| | - Richard Harwood
- Charles Perkins Centre, Sydney Microscopy and Microanalysis, University of Sydney, Camperdown, NSW, Australia
| | - Jaswinder S Samra
- The University of Sydney Northern Clinical School, Sydney, NSW, Australia
- Upper Gastrointestinal Surgical Unit, Royal North Shore Hospital, St Leonards, NSW, Australia
| | - Anthony J Gill
- The University of Sydney Northern Clinical School, Sydney, NSW, Australia
- Department of Anatomical Pathology, Royal North Shore Hospital, St Leonards, NSW, Australia
- Cancer Diagnosis and Pathology Research Group, Kolling Institute of Medical Research, St Leonards, NSW, Australia
| | | | - Helen E Thomas
- St Vincent's Institute, Fitzroy, VIC, Australia
- Department of Medicine, St Vincent's Hospital, University of Melbourne, Fitzroy, VIC, Australia
| | - Peter Thorn
- Charles Perkins Centre, School of Medical Sciences, University of Sydney, Camperdown, NSW, Australia.
| |
Collapse
|
48
|
Held RG, Liang J, Brunger AT. Nanoscale architecture of synaptic vesicles and scaffolding complexes revealed by cryo-electron tomography. Proc Natl Acad Sci U S A 2024; 121:e2403136121. [PMID: 38923992 PMCID: PMC11228483 DOI: 10.1073/pnas.2403136121] [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: 02/17/2024] [Accepted: 05/16/2024] [Indexed: 06/28/2024] Open
Abstract
The spatial distribution of proteins and their arrangement within the cellular ultrastructure regulates the opening of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors in response to glutamate release at the synapse. Fluorescence microscopy imaging revealed that the postsynaptic density (PSD) and scaffolding proteins in the presynaptic active zone (AZ) align across the synapse to form a trans-synaptic "nanocolumn," but the relation to synaptic vesicle release sites is uncertain. Here, we employ focused-ion beam (FIB) milling and cryoelectron tomography to image synapses under near-native conditions. Improved image contrast, enabled by FIB milling, allows simultaneous visualization of supramolecular nanoclusters within the AZ and PSD and synaptic vesicles. Surprisingly, membrane-proximal synaptic vesicles, which fuse to release glutamate, are not preferentially aligned with AZ or PSD nanoclusters. These synaptic vesicles are linked to the membrane by peripheral protein densities, often consistent in size and shape with Munc13, as well as globular densities bridging the synaptic vesicle and plasma membrane, consistent with prefusion complexes of SNAREs, synaptotagmins, and complexin. Monte Carlo simulations of synaptic transmission events using biorealistic models guided by our tomograms predict that clustering AMPARs within PSD nanoclusters increases the variability of the postsynaptic response but not its average amplitude. Together, our data support a model in which synaptic strength is tuned at the level of single vesicles by the spatial relationship between scaffolding nanoclusters and single synaptic vesicle fusion sites.
Collapse
Affiliation(s)
- Richard G. Held
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA94305
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA94305
- Department of Structural Biology, Stanford University, Stanford, CA94305
- Department of Photon Science, Stanford University, Stanford, CA94305
- HHMI, Stanford University, Stanford, CA94305
| | - Jiahao Liang
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA94305
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA94305
- Department of Structural Biology, Stanford University, Stanford, CA94305
- Department of Photon Science, Stanford University, Stanford, CA94305
- HHMI, Stanford University, Stanford, CA94305
| | - Axel T. Brunger
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA94305
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA94305
- Department of Structural Biology, Stanford University, Stanford, CA94305
- Department of Photon Science, Stanford University, Stanford, CA94305
- HHMI, Stanford University, Stanford, CA94305
| |
Collapse
|
49
|
Bucher ML, Dicent J, Duarte Hospital C, Miller GW. Neurotoxicology of dopamine: Victim or assailant? Neurotoxicology 2024; 103:175-188. [PMID: 38857676 PMCID: PMC11694735 DOI: 10.1016/j.neuro.2024.06.001] [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: 04/13/2024] [Revised: 06/02/2024] [Accepted: 06/03/2024] [Indexed: 06/12/2024]
Abstract
Since the identification of dopamine as a neurotransmitter in the mid-20th century, investigators have examined the regulation of dopamine homeostasis at a basic biological level and in human disorders. Genetic animal models that manipulate the expression of proteins involved in dopamine homeostasis have provided key insight into the consequences of dysregulated dopamine. As a result, we have come to understand the potential of dopamine to act as an endogenous neurotoxin through the generation of reactive oxygen species and reactive metabolites that can damage cellular macromolecules. Endogenous factors, such as genetic variation and subcellular processes, and exogenous factors, such as environmental exposures, have been identified as contributors to the dysregulation of dopamine homeostasis. Given the variety of dysregulating factors that impact dopamine homeostasis and the potential for dopamine itself to contribute to further cellular dysfunction, dopamine can be viewed as both the victim and an assailant of neurotoxicity. Parkinson's disease has emerged as the exemplar case study of dopamine dysregulation due to the genetic and environmental factors known to contribute to disease risk, and due to the evidence of dysregulated dopamine as a pathologic and pathogenic feature of the disease. This review, inspired by the talk, "Dopamine in Durham: location, location, location" presented by Dr. Miller for the Jacob Hooisma Memorial Lecture at the International Neurotoxicology Association meeting in 2023, offers a primer on dopamine toxicity covering endogenous and exogenous factors that disrupt dopamine homeostasis and the actions of dopamine as an endogenous neurotoxin.
Collapse
Affiliation(s)
- Meghan L Bucher
- Department of Environmental Health Sciences, Mailman School of Public Health at Columbia University, New York, NY 10032, USA
| | - Jocelyn Dicent
- Department of Environmental Health Sciences, Mailman School of Public Health at Columbia University, New York, NY 10032, USA
| | - Carolina Duarte Hospital
- Department of Environmental Health Sciences, Mailman School of Public Health at Columbia University, New York, NY 10032, USA
| | - Gary W Miller
- Department of Environmental Health Sciences, Mailman School of Public Health at Columbia University, New York, NY 10032, USA; Department of Molecular Pharmacology and Therapeutics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
50
|
Stockwell I, Watson JF, Greger IH. Tuning synaptic strength by regulation of AMPA glutamate receptor localization. Bioessays 2024; 46:e2400006. [PMID: 38693811 PMCID: PMC7616278 DOI: 10.1002/bies.202400006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 04/19/2024] [Accepted: 04/23/2024] [Indexed: 05/03/2024]
Abstract
Long-term potentiation (LTP) of excitatory synapses is a leading model to explain the concept of information storage in the brain. Multiple mechanisms contribute to LTP, but central amongst them is an increased sensitivity of the postsynaptic membrane to neurotransmitter release. This sensitivity is predominantly determined by the abundance and localization of AMPA-type glutamate receptors (AMPARs). A combination of AMPAR structural data, super-resolution imaging of excitatory synapses, and an abundance of electrophysiological studies are providing an ever-clearer picture of how AMPARs are recruited and organized at synaptic junctions. Here, we review the latest insights into this process, and discuss how both cytoplasmic and extracellular receptor elements cooperate to tune the AMPAR response at the hippocampal CA1 synapse.
Collapse
Affiliation(s)
- Imogen Stockwell
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Jake F. Watson
- Institute of Science and Technology, Technology (IST) Austria, Klosterneuburg, Austria
| | - Ingo H. Greger
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, UK
| |
Collapse
|