1
|
Sun R, Allen JP, Mao Z, Wilson L, Haider M, Alten B, Zhou Z, Wang X, Zhou Q. The postsynaptic density in excitatory synapses is composed of clustered, heterogeneous nanoblocks. J Cell Biol 2025; 224:e202406133. [PMID: 40145863 PMCID: PMC11948668 DOI: 10.1083/jcb.202406133] [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: 06/23/2024] [Revised: 12/05/2024] [Accepted: 03/13/2025] [Indexed: 03/28/2025] Open
Abstract
The nanoscale organization of proteins within synapses is critical for maintaining and regulating synaptic transmission and plasticity. Here, we used cryo-electron tomography (cryo-ET) to directly visualize the three-dimensional architecture and supramolecular organization of postsynaptic components in both synaptosomes and synapses from cultured neurons. Cryo-ET revealed that postsynaptic density (PSD) is composed of membrane-associated nanoblocks of various sizes. Subtomogram averaging from synaptosomes showed two types (type A and B) of postsynaptic receptor-like particles at resolutions of 24 and 26 Å, respectively. Furthermore, our analysis suggested that potential presynaptic release sites are closer to nanoblocks with type A/B receptor-like particles than to nanoblocks without type A/B receptor-like particles. The results of this study provide a more comprehensive understanding of synaptic ultrastructure and suggest that PSD is composed of clustering of various nanoblocks. These nanoblocks are heterogeneous in size, assembly, and distribution, which likely contribute to the dynamic nature of PSD in modulating synaptic strength.
Collapse
Affiliation(s)
- Rong Sun
- Department of Cell and Developmental Biology, Center for Structural Biology, Vanderbilt Kennedy Center, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
| | - James P. Allen
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
| | - Zhuqing Mao
- Department of Cell and Developmental Biology, Center for Structural Biology, Vanderbilt Kennedy Center, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
| | - Liana Wilson
- Department of Cell and Developmental Biology, Center for Structural Biology, Vanderbilt Kennedy Center, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
| | - Mariam Haider
- Department of Cell and Developmental Biology, Center for Structural Biology, Vanderbilt Kennedy Center, Vanderbilt University, Nashville, TN, USA
- Center for Structural Biology Cryo-EM Facility, Vanderbilt University, Nashville, TN, USA
| | - Baris Alten
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - Zimeng Zhou
- Department of Cell and Developmental Biology, Center for Structural Biology, Vanderbilt Kennedy Center, Vanderbilt University, Nashville, TN, USA
- School of Engineering, Vanderbilt University, Nashville, TN, USA
| | - Xinyi Wang
- Department of Cell and Developmental Biology, Center for Structural Biology, Vanderbilt Kennedy Center, Vanderbilt University, Nashville, TN, USA
- Peabody College, Vanderbilt University, Nashville, TN, USA
| | - Qiangjun Zhou
- Department of Cell and Developmental Biology, Center for Structural Biology, Vanderbilt Kennedy Center, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
2
|
Wang W, Han F, Qi Z, Yan C, Su B, Wang J. Phase Separation: Orchestrating Biological Adaptations to Environmental Fluctuations. Int J Mol Sci 2025; 26:4614. [PMID: 40429758 PMCID: PMC12110863 DOI: 10.3390/ijms26104614] [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: 03/24/2025] [Revised: 04/23/2025] [Accepted: 05/08/2025] [Indexed: 05/29/2025] Open
Abstract
Organisms have evolved various protective mechanisms to survive in complex and dynamic environments. Phase separation is a ubiquitous mechanism in plants, animals, and microorganisms. It facilitates the aggregation of biomolecules through weak interactions, forming membrane-less organelles that help organisms respond effectively to stress signals. These biomolecular condensates include DNA, RNA, and proteins. Proteins are categorized into scaffold and client proteins, whose coordinated actions ensure the compartmentalization of cellular signals, thereby regulating various biological processes. Studies indicate that, in response to stress, phase separation modulates gene expression, signal transduction, cytoskeleton dynamics, and protein homeostasis, ensuring the precise spatiotemporal control of cellular functions. These insights underscore the crucial role of phase separation in maintaining cellular integrity and adaptability.
Collapse
Affiliation(s)
- Wenxiu Wang
- National Key Laboratory of Agricultural Microbiology, Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
- Key Laboratory of Forage and Endemic Crop Biology, Ministry of Education, School of Life Science, Inner Mongolia University, Hohhot 010021, China
| | - Fangbing Han
- College of Agriculture, Henan University, Kaifeng 475004, China
| | - Zhi Qi
- Key Laboratory of Forage and Endemic Crop Biology, Ministry of Education, School of Life Science, Inner Mongolia University, Hohhot 010021, China
| | - Chunxia Yan
- Key Laboratory of Forage and Endemic Crop Biology, Ministry of Education, School of Life Science, Inner Mongolia University, Hohhot 010021, China
| | - Bodan Su
- National Key Laboratory of Agricultural Microbiology, Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Jin Wang
- National Key Laboratory of Agricultural Microbiology, Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| |
Collapse
|
3
|
Huhn C, Ho SY, Schulte C, Khayenko V, Hemmen K, Peulen TO, Wiessler AL, Bothe S, Bej A, Talucci I, Schönemann L, Werner C, Schindelin H, Strømgaard K, Villmann C, Heinze KG, Hruska M, Hell JW, Maric HM. eSylites: Synthetic Probes for Visualization and Topographic Mapping of Single Excitatory Synapses. J Am Chem Soc 2025; 147:15261-15280. [PMID: 40111234 DOI: 10.1021/jacs.5c00772] [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: 03/22/2025]
Abstract
The spatiotemporal organization of the postsynaptic density (PSD) is a fundamental determinant of synaptic transmission, information processing, and storage in the brain. The major bottleneck that prevents the direct and precise representation of the nanometer-scaled organization of excitatory glutamatergic synapses is the size of antibodies, nanobodies, and the genetically encoded fluorescent tags. Here, we introduce small, high affinity synthetic probes for simplified, high contrast visualization of excitatory synapses without the limitations of larger biomolecules. In vitro binding quantification together with microscopy-based evaluation identified eSylites, a series of fluorescent bivalent peptides comprising a dye, linker, and sequence composition that show remarkable cellular target selectivity. Applied on primary neurons or brain slices at nanomolar concentrations, eSylites specifically report PSD-95, the key orchestrator of glutamate receptor nanodomains juxtaposed to the presynaptic glutamate release sites that mediate fast synaptic transmission. The eSylite design minimizes a spatial dye offset and thereby enables visualization of PSD-95 with improved localization precision and further time-resolved discrimination. In particular, we find that individual dendritic spines can contain separate nanodomains enriched for either PSD-95 or its closest homologues, PSD-93 or SAP102. Collectively, these data establish eSylites as a broadly applicable tool for simplified excitatory synapse visualization, as well as a high-end microscopy compatible probe for resolving the PSD organization with unprecedented resolution.
Collapse
Affiliation(s)
- Christiane Huhn
- Rudolf Virchow Center for Integrative and Translational Bioimaging, Julius-Maximilians-Universität (JMU) Würzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany
- Biocenter, Department of Biotechnology and Biophysics, Julius-Maximilians-Universität (JMU) Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Sheng-Yang Ho
- Department of Pharmacology, University of California Davis, Davis, California 95616, United States
| | - Clemens Schulte
- Rudolf Virchow Center for Integrative and Translational Bioimaging, Julius-Maximilians-Universität (JMU) Würzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany
- Biocenter, Department of Biotechnology and Biophysics, Julius-Maximilians-Universität (JMU) Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Vladimir Khayenko
- Rudolf Virchow Center for Integrative and Translational Bioimaging, Julius-Maximilians-Universität (JMU) Würzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany
- Biocenter, Department of Biotechnology and Biophysics, Julius-Maximilians-Universität (JMU) Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Katherina Hemmen
- Rudolf Virchow Center for Integrative and Translational Bioimaging, Julius-Maximilians-Universität (JMU) Würzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany
| | - Thomas-Otavio Peulen
- Rudolf Virchow Center for Integrative and Translational Bioimaging, Julius-Maximilians-Universität (JMU) Würzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany
| | - Anna-Lena Wiessler
- Institute for Clinical Neurobiology, Julius-Maximilians-Universität (JMU) Würzburg, 97078 Würzburg, Germany
| | - Sebastian Bothe
- Rudolf Virchow Center for Integrative and Translational Bioimaging, Julius-Maximilians-Universität (JMU) Würzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany
- Institute of Pharmacy and Food Chemistry, Julius-Maximilians-Universität (JMU) Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Aritra Bej
- Department of Pharmacology, University of California Davis, Davis, California 95616, United States
| | - Ivan Talucci
- Rudolf Virchow Center for Integrative and Translational Bioimaging, Julius-Maximilians-Universität (JMU) Würzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany
| | - Lars Schönemann
- Rudolf Virchow Center for Integrative and Translational Bioimaging, Julius-Maximilians-Universität (JMU) Würzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany
| | - Christian Werner
- Biocenter, Department of Biotechnology and Biophysics, Julius-Maximilians-Universität (JMU) Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Hermann Schindelin
- Rudolf Virchow Center for Integrative and Translational Bioimaging, Julius-Maximilians-Universität (JMU) Würzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany
| | - Kristian Strømgaard
- Center for Biopharmaceuticals, Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Carmen Villmann
- Institute for Clinical Neurobiology, Julius-Maximilians-Universität (JMU) Würzburg, 97078 Würzburg, Germany
| | - Katrin G Heinze
- Rudolf Virchow Center for Integrative and Translational Bioimaging, Julius-Maximilians-Universität (JMU) Würzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany
| | - Martin Hruska
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, West Virginia 26506, United States
| | - Johannes W Hell
- Department of Pharmacology, University of California Davis, Davis, California 95616, United States
| | - Hans M Maric
- Rudolf Virchow Center for Integrative and Translational Bioimaging, Julius-Maximilians-Universität (JMU) Würzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany
- Biocenter, Department of Biotechnology and Biophysics, Julius-Maximilians-Universität (JMU) Würzburg, Am Hubland, 97074 Würzburg, Germany
| |
Collapse
|
4
|
Brill J, Clarke B, Hong I, Huganir RL. Dissociation of SYNGAP1 enzymatic and structural roles: Intrinsic excitability and seizure susceptibility. Proc Natl Acad Sci U S A 2025; 122:e2427288122. [PMID: 40294267 PMCID: PMC12067237 DOI: 10.1073/pnas.2427288122] [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: 12/31/2024] [Accepted: 03/12/2025] [Indexed: 04/30/2025] Open
Abstract
SYNGAP1 is a key Ras-GAP protein enriched at excitatory synapses, with mutations causing intellectual disability and epilepsy in humans. Recent studies have revealed that in addition to its role as a negative regulator of G-protein signaling through its GAP enzymatic activity, SYNGAP1 plays an important structural role through its interaction with postsynaptic density proteins. Here, we reveal that intrinsic excitability deficits and seizure phenotypes in heterozygous Syngap1 knockout (KO) mice are differentially dependent on Syngap1 GAP activity. Cortical excitatory neurons in heterozygous KO mice displayed reduced intrinsic excitability, including lower input resistance, and increased rheobase, a phenotype recapitulated in GAP-deficient Syngap1 mutants. However, seizure severity and susceptibility to pentylenetetrazol (PTZ)-induced seizures were significantly elevated in heterozygous KO mice but unaffected in GAP-deficient mutants, implicating the structural rather than enzymatic role of Syngap1 in seizure regulation. These findings highlight the complex interplay between SYNGAP1 structural and catalytic functions in neuronal physiology and disease.
Collapse
Affiliation(s)
- Julia Brill
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Blaise Clarke
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Ingie Hong
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Richard L. Huganir
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD21205
| |
Collapse
|
5
|
Shao X, Volk L. PICK1 links KIBRA and AMPA receptor subunit GluA2 in coiled-coil-driven supramolecular complexes. J Biol Chem 2025; 301:108397. [PMID: 40074086 DOI: 10.1016/j.jbc.2025.108397] [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: 11/08/2023] [Revised: 02/24/2025] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
The human memory-associated protein KIBRA regulates synaptic plasticity and trafficking of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)-type glutamate receptors, and is implicated in multiple neuropsychiatric and cognitive disorders. How KIBRA forms complexes with and regulates AMPA receptors remains unclear. Here, we show that KIBRA does not interact directly with the AMPA receptor subunit GluA2, but that protein interacting with C kinase 1 (PICK1), a key regulator of AMPA receptor trafficking, can serve as a bridge between KIBRA and GluA2. In contrast, KIBRA can form a complex with GluA1 independent of PICK1. We identified structural determinants of KIBRA-PICK1-AMPAR complexes by investigating interactions and cellular expression patterns of different combinations of KIBRA and PICK1 domain mutants. We find that the PICK1 BAR domain, a coiled-coil structure, is sufficient for interaction with KIBRA, whereas mutation of the PICK1 BAR domain disrupts KIBRA-PICK1-GluA2 complex formation. In addition, KIBRA recruits PICK1 into large supramolecular complexes, a process which requires KIBRA coiled-coil domains. These findings reveal molecular mechanisms by which KIBRA can organize key synaptic signaling complexes.
Collapse
Affiliation(s)
- Xin Shao
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Lenora Volk
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, Texas, USA; Neuroscience Graduate Program, UT Southwestern Medical Center, Dallas, Texas, USA; Department of Psychiatry, UT Southwestern Medical Center, Dallas, Texas, USA; Peter O'Donnell Jr Brain Institute Investigator, UT Southwestern Medical Center, Dallas, Texas, USA.
| |
Collapse
|
6
|
MacAinsh M, Muhammedkutty FNK, Prasad R, Zhou HX. Membrane Association of Intrinsically Disordered Proteins. Annu Rev Biophys 2025; 54:275-302. [PMID: 39952269 PMCID: PMC12055482 DOI: 10.1146/annurev-biophys-070124-092816] [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: 02/17/2025]
Abstract
It is now clear that membrane association of intrinsically disordered proteins or intrinsically disordered regions regulates many cellular processes, such as membrane targeting of Src family kinases and ion channel gating. Residue-specific characterization by nuclear magnetic resonance spectroscopy, molecular dynamics simulations, and other techniques has shown that polybasic motifs and amphipathic helices are the main drivers of membrane association; sequence-based prediction of residue-specific membrane association propensity has become possible. Membrane association facilitates protein-protein interactions and protein aggregation-these effects are due to reduced dimensionality but are similar to those afforded by condensate formation via liquid-liquid phase separation (LLPS). LLPS at the membrane surface provides a powerful means for recruiting and clustering proteins, as well as for membrane remodeling.
Collapse
Affiliation(s)
- Matthew MacAinsh
- Department of Chemistry, University of Illinois, Chicago, Illinois, USA;
| | | | - Ramesh Prasad
- Department of Chemistry, University of Illinois, Chicago, Illinois, USA;
| | - Huan-Xiang Zhou
- Department of Chemistry, University of Illinois, Chicago, Illinois, USA;
- Department of Physics, University of Illinois, Chicago, Illinois, USA
| |
Collapse
|
7
|
Pandey V, Hosokawa T, Hayashi Y, Urakubo H. Multiphasic protein condensation governed by shape and valency. Cell Rep 2025; 44:115504. [PMID: 40199325 DOI: 10.1016/j.celrep.2025.115504] [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: 02/05/2025] [Accepted: 03/12/2025] [Indexed: 04/10/2025] Open
Abstract
Liquid-liquid phase separation (LLPS) of biological macromolecules leads to the formation of various membraneless organelles. The multilayered and multiphasic form of LLPS can mediate complex cellular functions; however, the determinants of its topological features are not fully understood. Herein, we focus on synaptic proteins consisting of Ca2+/calmodulin-dependent protein kinase II (CaMKII) and its interacting partners and present a computational model that reproduces forms of LLPS, including a form of two-phase condensates, phase-in-phase (PIP) organization. The model analyses reveal that the PIP formation requires competitive binding between the proteins. The PIP forms only when CaMKII has high valency and a short linker length. Such CaMKII exhibits low surface tension, a modular structure, and slow diffusion, enabling it to stay in small biochemical domains for a long time, which is necessary for synaptic plasticity. Thus, the computational modeling reveals new structure-function relationships for CaMKII as a synaptic memory unit.
Collapse
Affiliation(s)
- Vikas Pandey
- Department of Biomedical Data Science, Fujita Health University School of Medicine, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi 470-1192, Japan; International Center for Brain Science (ICBS), Fujita Health University, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi 470-1192, Japan; National Institute for Physiological Sciences, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji, Okazaki, Aichi 444-8787, Japan
| | - Tomohisa Hosokawa
- Department of Pharmacology, Kyoto University Graduate School of Medicine, Kyoto 606-8501, Japan
| | - Yasunori Hayashi
- Department of Pharmacology, Kyoto University Graduate School of Medicine, Kyoto 606-8501, Japan
| | - Hidetoshi Urakubo
- Department of Biomedical Data Science, Fujita Health University School of Medicine, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi 470-1192, Japan; International Center for Brain Science (ICBS), Fujita Health University, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi 470-1192, Japan; National Institute for Physiological Sciences, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji, Okazaki, Aichi 444-8787, Japan.
| |
Collapse
|
8
|
Sun D, Zhao X, Wiegand T, Martin-Lemaitre C, Borianne T, Kleinschmidt L, Grill SW, Hyman AA, Weber C, Honigmann A. Assembly of tight junction belts by ZO1 surface condensation and local actin polymerization. Dev Cell 2025; 60:1234-1250.e6. [PMID: 39742662 DOI: 10.1016/j.devcel.2024.12.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 09/30/2024] [Accepted: 12/05/2024] [Indexed: 01/04/2025]
Abstract
Tight junctions play an essential role in sealing tissues, by forming belts of adhesion strands around cellular perimeters. Recent work has shown that the condensation of ZO1 scaffold proteins is required for tight junction assembly. However, the mechanisms by which junctional condensates initiate at cell-cell contacts and elongate around cell perimeters remain unknown. Combining biochemical reconstitutions and live-cell imaging of MDCKII tissue, we found that tight junction belt formation is driven by adhesion receptor-mediated ZO1 surface condensation coupled to local actin polymerization. Adhesion receptor oligomerization provides the signal for surface binding and local condensation of ZO1 at the cell membrane. Condensation produces a molecular scaffold that selectively enriches junctional proteins. Finally, ZO1 condensates directly facilitate local actin polymerization and filament bundling, driving the elongation into a continuous tight junction belt. More broadly, our work identifies how cells couple surface condensation with cytoskeleton organization to assemble and structure adhesion complexes.
Collapse
Affiliation(s)
- Daxiao Sun
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany; Technische Universität Dresden, Biotechnologisches Zentrum, Center for Molecular and Cellular Bioengineering (CMCB), Dresden, Germany.
| | - Xueping Zhao
- Department of Mathematical Sciences, University of Nottingham, Ningbo, China
| | - Tina Wiegand
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Cecilie Martin-Lemaitre
- Technische Universität Dresden, Biotechnologisches Zentrum, Center for Molecular and Cellular Bioengineering (CMCB), Dresden, Germany
| | - Tom Borianne
- Technische Universität Dresden, Biotechnologisches Zentrum, Center for Molecular and Cellular Bioengineering (CMCB), Dresden, Germany
| | - Lennart Kleinschmidt
- Technische Universität Dresden, Biotechnologisches Zentrum, Center for Molecular and Cellular Bioengineering (CMCB), Dresden, Germany
| | - Stephan W Grill
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany; Cluster of Excellence Physics of Life, TU Dresden, Dresden, Germany
| | - Anthony A Hyman
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany; Cluster of Excellence Physics of Life, TU Dresden, Dresden, Germany
| | - Christoph Weber
- Faculty of Mathematics, Natural Sciences, and Materials Engineering, Institute of Physics, University of Augsburg, Augsburg, Germany.
| | - Alf Honigmann
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany; Technische Universität Dresden, Biotechnologisches Zentrum, Center for Molecular and Cellular Bioengineering (CMCB), Dresden, Germany; Cluster of Excellence Physics of Life, TU Dresden, Dresden, Germany.
| |
Collapse
|
9
|
Arsenault J, Kong T, Saghian R, Weng OY, Pathak SS, Yang C, Chao OY, Rakhaminov G, Forman-Kay JD, Ditlev JA, Yang YM, Wang LY. Essential lipids enrich membrane-associated condensates to rescue synaptic morpho-functional deficits in a mouse model of autism. Cell Rep 2025; 44:115573. [PMID: 40232934 DOI: 10.1016/j.celrep.2025.115573] [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: 09/23/2024] [Revised: 01/14/2025] [Accepted: 03/26/2025] [Indexed: 04/17/2025] Open
Abstract
Synaptic proteins form intracellular condensates with their scaffolds, but it is unknown whether and how essential lipids transform dynamic cytosolic condensates into stable, functional macromolecular assemblies at the membrane. We show that docosahexaenoic acid (DHA), independent of canonical fatty acid receptor 4 signaling, facilitates the re-localization of cytosolic "full-droplet" condensates composed of the key synaptic elements PSD95 and Kv1.2 to the plasma membrane as "half-droplets." To exploit the therapeutic potential of DHA in vivo, we briefly place juvenile wild-type and Fmr1 KO mice, modeling human fragile X syndrome (FXS), under DHA-enriched or -depleted diets. DHA reverses the inhibitory overtone by promoting the re-localization of presynaptic PSD95-Kv1.2 condensates to interneuron terminal membranes and corrects morpho-functional synaptic defects and stereotypic behaviors. These findings reveal an unexpected role of essential lipids in translocating dynamic condensates into stable synaptic condensates, providing long-lasting benefits for rectifying excitation-inhibition imbalance in FXS and potentially other neurodevelopmental disorders.
Collapse
Affiliation(s)
- Jason Arsenault
- Program in Neurosciences & Mental Health, SickKids Research Institute, Toronto, ON M5G 1X8, Canada; Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Tian Kong
- Program in Neurosciences & Mental Health, SickKids Research Institute, Toronto, ON M5G 1X8, Canada; Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Rayan Saghian
- Program in Neurosciences & Mental Health, SickKids Research Institute, Toronto, ON M5G 1X8, Canada; Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Octavia Yifang Weng
- Program in Neurosciences & Mental Health, SickKids Research Institute, Toronto, ON M5G 1X8, Canada; Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Salil Saurav Pathak
- Department of Biomedical Sciences, University of Minnesota Medical School, 1035 University Drive, Duluth, MN 55812, USA
| | - Chengye Yang
- Program in Neurosciences & Mental Health, SickKids Research Institute, Toronto, ON M5G 1X8, Canada; Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Owen Y Chao
- Department of Biomedical Sciences, University of Minnesota Medical School, 1035 University Drive, Duluth, MN 55812, USA
| | - Gaddy Rakhaminov
- Program in Molecular Medicine, SickKids Research Institute, Toronto, ON M5G 1X8, Canada; Department of Biochemistry, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Julie D Forman-Kay
- Program in Molecular Medicine, SickKids Research Institute, Toronto, ON M5G 1X8, Canada; Department of Biochemistry, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Jonathon A Ditlev
- Program in Molecular Medicine, SickKids Research Institute, Toronto, ON M5G 1X8, Canada; Department of Biochemistry, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada; Program in Cell Biology, SickKids Research Institute, Toronto, ON M5G 1X8, Canada.
| | - Yi-Mei Yang
- Department of Biomedical Sciences, University of Minnesota Medical School, 1035 University Drive, Duluth, MN 55812, USA.
| | - Lu-Yang Wang
- Program in Neurosciences & Mental Health, SickKids Research Institute, Toronto, ON M5G 1X8, Canada; Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
10
|
Nixon-Abell J, Ruggeri FS, Qamar S, Herling TW, Czekalska MA, Shen Y, Wang G, King C, Fernandopulle MS, Sneideris T, Watson JL, Pillai VVS, Meadows W, Henderson JW, Chambers JE, Wagstaff JL, Williams SH, Coyle H, Šneiderienė G, Lu Y, Zhang S, Marciniak SJ, Freund SMV, Derivery E, Ward ME, Vendruscolo M, Knowles TPJ, St George-Hyslop P. ANXA11 biomolecular condensates facilitate protein-lipid phase coupling on lysosomal membranes. Nat Commun 2025; 16:2814. [PMID: 40118863 PMCID: PMC11928461 DOI: 10.1038/s41467-025-58142-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 03/12/2025] [Indexed: 03/24/2025] Open
Abstract
Phase transitions of cellular proteins and lipids play a key role in governing the organisation and coordination of intracellular biology. Recent work has raised the intriguing prospect that phase transitions in proteins and lipids can be co-regulated. Here we investigate this possibility in the ribonucleoprotein (RNP) granule-ANXA11-lysosome ensemble, where ANXA11 tethers RNP granules to lysosomal membranes to enable their co-trafficking. We show that changes to the protein phase state within this system, driven by the low complexity ANXA11 N-terminus, induces a coupled phase state change in the lipids of the underlying membrane. We identify the ANXA11 interacting proteins ALG2 and CALC as potent regulators of ANXA11-based phase coupling and demonstrate their influence on the nanomechanical properties of the ANXA11-lysosome ensemble and its capacity to engage RNP granules. The phenomenon of protein-lipid phase coupling we observe within this system serves as a potential regulatory mechanism in RNA trafficking and offers an important template to understand other examples across the cell whereby biomolecular condensates closely juxtapose organellar membranes.
Collapse
Grants
- Wellcome Trust
- F30 AG060722 NIA NIH HHS
- FC001029 Arthritis Research UK
- Canadian Institutes of Health Research (406915 Foundation Grant and Canadian Consortium on Neurodegeneration in Aging); Wellcome Career Development Award 227745/Z/23/Z (JNA), Wellcome Trust Collaborative Award 203249/Z/16/Z (PStGH, MEV, TPK), a US Alzheimer Society Zenith Grant ZEN-18-529769 (PStGH), a National Institute on Aging grant F30AG060722 (MEW), the NIH-Oxford-Cambridge Scholars Program (MSF), the El-Hibri Foundation (MSF), the Dutch Ministry of Education – Sector Plan Beta for science and technology (FSR), an Ernest Oppenheimer Early Career Research Fellowship (TWH), the Polish Ministry of Science and Higher Education (by Mobilnosc Plus V, decision number 1623/MOB/V/2017/0, MAC), and the Medical Research Council as part of UKRI (MC_UP_1201/13 to ED & MC_U105184326 to LMB NMR facility), and the Human Frontier Science Program (Career Development Award CDA00034/2017-C to ED). The CIMR microscopy core is supported by a Wellcome Trust Strategic Award 100140, and a Wellcome Trust equipment grant 093026. The Francis Crick Institute receives its core funding from Cancer Research UK (FC001029), the UK Medical Research Council (FC001029), and the Wellcome Trust (FC001029).
Collapse
Affiliation(s)
- Jonathon Nixon-Abell
- Department of Clinical Neurosciences, Cambridge Institute for Medical Research, Clinical School, University of Cambridge, Cambridge, UK.
| | - Francesco S Ruggeri
- Physical Chemistry and Soft matter, Wageningen University & Research, Stippeneng, The Netherlands
- Yusuf Hamied Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, UK
| | - Seema Qamar
- Department of Clinical Neurosciences, Cambridge Institute for Medical Research, Clinical School, University of Cambridge, Cambridge, UK
| | - Therese W Herling
- Yusuf Hamied Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, UK
| | - Magdalena A Czekalska
- Yusuf Hamied Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, UK
| | - Yi Shen
- Yusuf Hamied Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, UK
- School of Chemical and Biomolecular Engineering, The University of Sydney, Sydney, NSW, Australia
- The University of Sydney Nano Institute, The University of Sydney, Sydney, NSW, Australia
| | - Guozhen Wang
- Department of Clinical Neurosciences, Cambridge Institute for Medical Research, Clinical School, University of Cambridge, Cambridge, UK
- School of Medicine, Zhejiang University, Hangzhou, China
| | - Christopher King
- National institute for Neurological Disorder and Stroke, NIH, Bethesda, MD, USA
| | - Michael S Fernandopulle
- Department of Clinical Neurosciences, Cambridge Institute for Medical Research, Clinical School, University of Cambridge, Cambridge, UK
- National institute for Neurological Disorder and Stroke, NIH, Bethesda, MD, USA
- Medical Scientist Training Program, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Tomas Sneideris
- Yusuf Hamied Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, UK
| | - Joseph L Watson
- Cell Biology Division, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Visakh V S Pillai
- Physical Chemistry and Soft matter, Wageningen University & Research, Stippeneng, The Netherlands
| | - William Meadows
- Department of Clinical Neurosciences, Cambridge Institute for Medical Research, Clinical School, University of Cambridge, Cambridge, UK
| | - James W Henderson
- Department of Clinical Neurosciences, Cambridge Institute for Medical Research, Clinical School, University of Cambridge, Cambridge, UK
| | - Joseph E Chambers
- Department of Clinical Neurosciences, Cambridge Institute for Medical Research, Clinical School, University of Cambridge, Cambridge, UK
| | - Jane L Wagstaff
- Structure Studies Division, NMR Facility, MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge Biomedical Campus, Cambridge, UK
| | - Sioned H Williams
- Department of Clinical Neurosciences, Cambridge Institute for Medical Research, Clinical School, University of Cambridge, Cambridge, UK
| | - Helena Coyle
- Department of Clinical Neurosciences, Cambridge Institute for Medical Research, Clinical School, University of Cambridge, Cambridge, UK
| | - Greta Šneiderienė
- Yusuf Hamied Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, UK
| | - Yuqian Lu
- Yusuf Hamied Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, UK
| | - Shuyuan Zhang
- Yusuf Hamied Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, UK
| | - Stefan J Marciniak
- Department of Clinical Neurosciences, Cambridge Institute for Medical Research, Clinical School, University of Cambridge, Cambridge, UK
| | - Stefan M V Freund
- Structure Studies Division, NMR Facility, MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge Biomedical Campus, Cambridge, UK
| | - Emmanuel Derivery
- Cell Biology Division, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Michael E Ward
- National institute for Neurological Disorder and Stroke, NIH, Bethesda, MD, USA
| | - Michele Vendruscolo
- Yusuf Hamied Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, UK
| | - Tuomas P J Knowles
- Yusuf Hamied Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, UK
| | - Peter St George-Hyslop
- Department of Medicine (Division of Neurology), Temerty Faculty of Medicine, University Health Network, University of Toronto, Toronto, ON, Canada.
- Carol and Gene Ludwig Center for Research on Neurodegeneration, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Department of Neurology, Columbia University Irvine Medical Center, New York, NY, USA.
| |
Collapse
|
11
|
Holland J, Nott TJ, Aarts DGAL. Intrinsic hydrophobicity of IDP-based biomolecular condensates drives their partial drying on membrane surfaces. J Chem Phys 2025; 162:115101. [PMID: 40094245 PMCID: PMC11919390 DOI: 10.1063/5.0253522] [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: 12/17/2024] [Accepted: 02/19/2025] [Indexed: 03/19/2025] Open
Abstract
The localization of biomolecular condensates to intracellular membrane surfaces has emerged as an important feature of sub-cellular organization. In this work, we study the wetting behavior of biomolecular condensates on various substrates. We use confocal microscopy to measure the contact angles of model condensates formed by intrinsically disordered protein Ddx4N. We show the importance of taking optical aberrations into account, as these impact apparent contact angle measurements. Ddx4N condensates are seen to partially dry (contact angles above 90°) a model membrane, with little dependence on the magnitude of charge on, or tyrosine content of, Ddx4N. Further contact angle measurements on surfaces of varying hydrophilicity reveal a preference of Ddx4N condensates for hydrophobic surfaces, suggesting an intrinsic repulsion between protein condensates and hydrophilic membrane surfaces. This observation is in line with previous studies relating protein adsorption to surface hydrophilicity. Our work advances the understanding of the molecular details governing the localization of biomolecular condensates.
Collapse
Affiliation(s)
- J. Holland
- Department of Chemistry, Physical and Theoretical Chemistry Laboratory, University of Oxford, Oxford OX1 3QZ, United Kingdom
| | - T. J. Nott
- Department of Chemistry, King’s College London, Britannia House, 7 Trinity Street, London SE1 1DB, United Kingdom
| | - D. G. A. L. Aarts
- Department of Chemistry, Physical and Theoretical Chemistry Laboratory, University of Oxford, Oxford OX1 3QZ, United Kingdom
| |
Collapse
|
12
|
Wan Y, Hudson R, Smith J, Forman-Kay JD, Ditlev JA. Protein interactions, calcium, phosphorylation, and cholesterol modulate CFTR cluster formation on membranes. Proc Natl Acad Sci U S A 2025; 122:e2424470122. [PMID: 40063811 PMCID: PMC11929494 DOI: 10.1073/pnas.2424470122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 01/07/2025] [Indexed: 03/25/2025] Open
Abstract
The cystic fibrosis transmembrane conductance regulator (CFTR) is a chloride channel whose dysfunction leads to intracellular accumulation of chloride ions, dehydration of cell surfaces, and subsequent damage to airway and ductal organs. Beyond its function as a chloride channel, interactions between CFTR, epithelium sodium channel, and solute carrier (SLC) transporter family membrane proteins and cytoplasmic proteins, including calmodulin and Na+/H+ exchanger regulatory factor-1 (NHERF-1), coregulate ion homeostasis. CFTR has also been observed to form mesoscale membrane clusters. However, the contributions of multivalent protein and lipid interactions to cluster formation are not well understood. Using a combination of computational modeling and biochemical reconstitution assays, we demonstrate that multivalent interactions with CFTR protein binding partners, calcium, and membrane cholesterol can induce mesoscale CFTR cluster formation on model membranes. Phosphorylation of the intracellular domains of CFTR also promotes mesoscale cluster formation in the absence of calcium, indicating that multiple mechanisms can contribute to CFTR cluster formation. Our findings reveal that coupling of multivalent protein and lipid interactions promotes CFTR cluster formation consistent with membrane-associated biological phase separation.
Collapse
Affiliation(s)
- Yimei Wan
- Department of Biochemistry, University of Toronto, Toronto, ONM5S 1A8, Canada
- Program in Molecular Medicine, Hospital for Sick Children, Toronto, ONM5G 0A4, Canada
| | - Rhea Hudson
- Program in Molecular Medicine, Hospital for Sick Children, Toronto, ONM5G 0A4, Canada
| | - Jordyn Smith
- Program in Molecular Medicine, Hospital for Sick Children, Toronto, ONM5G 0A4, Canada
| | - Julie D. Forman-Kay
- Department of Biochemistry, University of Toronto, Toronto, ONM5S 1A8, Canada
- Program in Molecular Medicine, Hospital for Sick Children, Toronto, ONM5G 0A4, Canada
| | - Jonathon A. Ditlev
- Department of Biochemistry, University of Toronto, Toronto, ONM5S 1A8, Canada
- Program in Molecular Medicine, Hospital for Sick Children, Toronto, ONM5G 0A4, Canada
- Program in Cell and Systems Biology, Hospital for Sick Children, Toronto, ONM5G 0A4, Canada
| |
Collapse
|
13
|
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
|
14
|
Sanfeliu-Cerdán N, Krieg M. The mechanobiology of biomolecular condensates. BIOPHYSICS REVIEWS 2025; 6:011310. [PMID: 40160200 PMCID: PMC11952833 DOI: 10.1063/5.0236610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 02/10/2025] [Indexed: 04/02/2025]
Abstract
The central goal of mechanobiology is to understand how the mechanical forces and material properties of organelles, cells, and tissues influence biological processes and functions. Since the first description of biomolecular condensates, it was hypothesized that they obtain material properties that are tuned to their functions inside cells. Thus, they represent an intriguing playground for mechanobiology. The idea that biomolecular condensates exhibit diverse and adaptive material properties highlights the need to understand how different material states respond to external forces and whether these responses are linked to their physiological roles within the cell. For example, liquids buffer and dissipate, while solids store and transmit mechanical stress, and the relaxation time of a viscoelastic material can act as a mechanical frequency filter. Hence, a liquid-solid transition of a condensate in the force transmission pathway can determine how mechanical signals are transduced within and in-between cells, affecting differentiation, neuronal network dynamics, and behavior to external stimuli. Here, we first review our current understanding of the molecular drivers and how rigidity phase transitions are set forth in the complex cellular environment. We will then summarize the technical advancements that were necessary to obtain insights into the rich and fascinating mechanobiology of condensates, and finally, we will highlight recent examples of physiological liquid-solid transitions and their connection to specific cellular functions. Our goal is to provide a comprehensive summary of the field on how cells harness and regulate condensate mechanics to achieve specific functions.
Collapse
Affiliation(s)
- Neus Sanfeliu-Cerdán
- ICFO - Institut de Ciències Fotòniques, Castelldefels, The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Michael Krieg
- ICFO - Institut de Ciències Fotòniques, Castelldefels, The Barcelona Institute of Science and Technology, Barcelona, Spain
| |
Collapse
|
15
|
Ding M, Wang D, Chen H, Kesner B, Grimm NB, Weissbein U, Lappala A, Jiang J, Rivera C, Lou J, Li P, Lee JT. A biophysical basis for the spreading behavior and limited diffusion of Xist. Cell 2025; 188:978-997.e25. [PMID: 39824183 PMCID: PMC11863002 DOI: 10.1016/j.cell.2024.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 11/04/2024] [Accepted: 12/06/2024] [Indexed: 01/20/2025]
Abstract
Xist RNA initiates X inactivation as it spreads in cis across the chromosome. Here, we reveal a biophysical basis for its cis-limited diffusion. Xist RNA and HNRNPK together drive a liquid-liquid phase separation (LLPS) that encapsulates the chromosome. HNRNPK droplets pull on Xist and internalize the RNA. Once internalized, Xist induces a further phase transition and "softens" the HNRNPK droplet. Xist alters the condensate's deformability, adhesiveness, and wetting properties in vitro. Other Xist-interacting proteins are internalized and entrapped within the droplet, resulting in a concentration of Xist and protein partners within the condensate. We attribute LLPS to HNRNPK's RGG and Xist's repeat B (RepB) motifs. Mutating these motifs causes Xist diffusion, disrupts polycomb recruitment, and precludes the required mixing of chromosomal compartments for Xist's migration. Thus, we hypothesize that phase transitions in HNRNPK condensates allow Xist to locally concentrate silencing factors and to spread through internal channels of the HNRNPK-encapsulated chromosome.
Collapse
Affiliation(s)
- Mingrui Ding
- State Key Laboratory of Membrane Biology, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Danni Wang
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Hui Chen
- Key Laboratory of RNA Biology, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Barry Kesner
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Niklas-Benedikt Grimm
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Universitat Pompeu Fabra (UPF), Barcelona, Spain; Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Uri Weissbein
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Anna Lappala
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Jiying Jiang
- State Key Laboratory of Membrane Biology, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Carlos Rivera
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Jizhong Lou
- Key Laboratory of RNA Biology, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Pilong Li
- State Key Laboratory of Membrane Biology, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Tsinghua-Peking Center for Life Sciences, Beijing 100084, China.
| | - Jeannie T Lee
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Genetics, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
16
|
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
|
17
|
Gendreizig D, Kalarikkal A, Holtbrügge SL, Mukherjee S, Galazzo L, Kucher S, Rosspeintner A, Schäfer LV, Bordignon E. A Combined Approach to Extract Rotational Dynamics of Globular Proteins Undergoing Liquid-Liquid Phase Separation. J Phys Chem B 2025; 129:1185-1196. [PMID: 39815790 DOI: 10.1021/acs.jpcb.4c06259] [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: 01/18/2025]
Abstract
The formation of protein condensates (droplets) via liquid-liquid phase separation (LLPS) is a commonly observed phenomenon in vitro. Changing the environmental properties with cosolutes, molecular crowders, protein partners, temperature, pressure, etc. has been shown to favor or disfavor the formation of protein droplets by fine-tuning the water-water, water-protein, and protein-protein interactions. Therefore, these environmental properties and their spatiotemporal fine-tuning are likely to be important also in a cellular context at the existing protein expression levels. One of the key physicochemical properties of biomolecules impacted by molecular crowding is diffusion, which determines the viscoelastic behavior of the condensates. Here, we investigate the change in the rotational diffusion of γD-crystallin, undergoing LLPS in vitro in aqueous solutions in the absence and presence of cosolutes. We studied its rotational dynamics using molecular dynamics simulations (MD), electron paramagnetic resonance (EPR) spectroscopy, and fluorescence spectroscopy. MD simulations performed under dilute and crowded conditions show that the rotational diffusion of crystallin in water is retarded by 1 to 2 orders of magnitude in the condensed phase. To obtain the rotational dynamics in the dilute phase, we used fluorescence anisotropy and to extract the retardation factor in the condensed phase, we used spin-labeled γD-crystallin proteins as EPR viscosity nanoprobes. Aided by a viscosity nanoruler calibrated with solutions at increasing sucrose concentrations, we validated the rotational diffusion retardation predicted by MD simulations. This study underlines the predictive power of MD simulations and showcases the use of a sensitive EPR nanoprobe to extract the viscosity of biomolecular condensates.
Collapse
Affiliation(s)
- Dominik Gendreizig
- Department of Physical Chemistry, Sciences II, University of Geneva, 30 Quai Ernest Ansermet, Geneva 1211, Switzerland
| | - Abhishek Kalarikkal
- Faculty of Chemistry and Biochemistry, Ruhr University Bochum, Bochum 44801, Germany
| | - Simon L Holtbrügge
- Center for Theoretical Chemistry, Faculty of Chemistry and Biochemistry, Ruhr University Bochum, Bochum 44801, Germany
| | - Saumyak Mukherjee
- Center for Theoretical Chemistry, Faculty of Chemistry and Biochemistry, Ruhr University Bochum, Bochum 44801, Germany
| | - Laura Galazzo
- Department of Physical Chemistry, Sciences II, University of Geneva, 30 Quai Ernest Ansermet, Geneva 1211, Switzerland
- Faculty of Chemistry and Biochemistry, Ruhr University Bochum, Bochum 44801, Germany
| | - Svetlana Kucher
- Department of Physical Chemistry, Sciences II, University of Geneva, 30 Quai Ernest Ansermet, Geneva 1211, Switzerland
| | - Arnulf Rosspeintner
- Department of Physical Chemistry, Sciences II, University of Geneva, 30 Quai Ernest Ansermet, Geneva 1211, Switzerland
| | - Lars V Schäfer
- Center for Theoretical Chemistry, Faculty of Chemistry and Biochemistry, Ruhr University Bochum, Bochum 44801, Germany
| | - Enrica Bordignon
- Department of Physical Chemistry, Sciences II, University of Geneva, 30 Quai Ernest Ansermet, Geneva 1211, Switzerland
| |
Collapse
|
18
|
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
|
19
|
Wang C, Quan Y, Jiang J, Yu H, Liu J, Tang W, Li X, Wang S, Huo D, Jiang GL, Yang Y, Ding Q. Protein Coronation-Induced Cancer Staging-Dependent Multilevel Cytotoxicity: An All-Humanized Study in Blood Vessel Organoids. ACS NANO 2025; 19:345-368. [PMID: 39743836 DOI: 10.1021/acsnano.4c07783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
The protein corona effect refers to the phenomenon wherein nanomaterials in the bloodstream are coated by serum proteins, yet how protein coronated nanomaterials interact with blood vessels and its toxicity implications remain poorly understood. In this study, we investigated protein corona-related vessel toxicity by using an all-humanized assay integrating blood vessel organoids and patient-derived serum. Initially, we screened various nanomaterials to discern how parameters including size, morphology, hydrophobicity, surface charge, and chirality-dependent protein corona difference influence their uptake by vessel organoids. For nanomaterials showing substantial differences in vessel uptake, their protein corona was analyzed by using label-free mass spectra. Our findings revealed the involvement of cancer staging-related cytoskeleton components in mediating preferential uptake by cells, including endothelial and mural cells. Additionally, a transcriptome study was conducted to elucidate the influence of nanomaterials. We confirmed that protein coronated nanomaterials provoke remodeling at both transcriptional and translational levels, impacting pathways such as PI3K-Akt/Hippo/Wnt, and membraneless organelle integrity, respectively. Our study further demonstrated that the remodeling potential of patient-derived protein coronated nanomaterials can be harnessed to synergize with antiangiogenesis therapeutics to improve the outcomes. We anticipate that this study will provide guidance for the safe use of nanomedicine in the future.
Collapse
Affiliation(s)
- Chan Wang
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, P. R. China
| | - Yingyi Quan
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing 211166, P. R. China
| | - Jiang Jiang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing 211166, P. R. China
| | - Han Yu
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, P. R. China
| | - Jia Liu
- Department of Medicinal Chemistry, School of Pharmacy, Nanjing Medical University, Nanjing 211166, P. R. China
| | - Wei Tang
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, P. R. China
| | - Xinyue Li
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, P. R. China
| | - Shouju Wang
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 211166, P. R. China
| | - Da Huo
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Department of Pharmaceutics, Nanjing Medical University, Nanjing 211169, P. R. China
| | - Guang-Liang Jiang
- Department of Urology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P. R. China
| | - Yang Yang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing 211166, P. R. China
| | - Qingqing Ding
- Department of Geriatric Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 211166, P. R. China
| |
Collapse
|
20
|
Brill J, Clarke B, Hong I, Huganir RL. Dissociation of SYNGAP1 Enzymatic and Structural Roles: Intrinsic Excitability and Seizure Susceptibility. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.14.633019. [PMID: 39868300 PMCID: PMC11761602 DOI: 10.1101/2025.01.14.633019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
SYNGAP1 is a key Ras-GAP protein enriched at excitatory synapses, with mutations causing intellectual disability and epilepsy in humans. Recent studies have revealed that in addition to its role as a negative regulator of G-protein signaling through its GAP enzymatic activity, SYNGAP1 plays an important structural role through its interaction with post-synaptic density proteins. Here, we reveal that intrinsic excitability deficits and seizure phenotypes in heterozygous Syngap1 knockout (KO) mice are differentially dependent on Syngap1 GAP activity. Cortical excitatory neurons in heterozygous KO mice displayed reduced intrinsic excitability, including lower input resistance, and increased rheobase, a phenotype recapitulated in GAP-deficient Syngap1 mutants. However, seizure severity and susceptibility to pentylenetetrazol (PTZ)-induced seizures were significantly elevated in heterozygous KO mice but unaffected in GAP-deficient mutants, implicating the structural rather than enzymatic role of Syngap1 in seizure regulation. These findings highlight the complex interplay between SYNGAP1 structural and catalytic functions in neuronal physiology and disease.
Collapse
Affiliation(s)
- Julia Brill
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Blaise Clarke
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ingie Hong
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Richard L. Huganir
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
21
|
Carnicer‐Lombarte A, Malliaras GG, Barone DG. The Future of Biohybrid Regenerative Bioelectronics. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2408308. [PMID: 39564751 PMCID: PMC11756040 DOI: 10.1002/adma.202408308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 09/13/2024] [Indexed: 11/21/2024]
Abstract
Biohybrid regenerative bioelectronics are an emerging technology combining implantable devices with cell transplantation. Once implanted, biohybrid regenerative devices integrate with host tissue. The combination of transplant and device provides an avenue to both replace damaged or dysfunctional tissue, and monitor or control its function with high precision. While early challenges in the fusion of the biological and technological components limited development of biohybrid regenerative technologies, progress in the field has resulted in a rapidly increasing number of applications. In this perspective the great potential of this emerging technology for the delivery of therapy is discussed, including both recent research progress and potential new directions. Then the technology barriers are discussed that will need to be addressed to unlock the full potential of biohybrid regenerative devices.
Collapse
Affiliation(s)
| | - George G. Malliaras
- Department of EngineeringElectrical Engineering DivisionUniversity of CambridgeCambridgeCB3 0FAUK
| | - Damiano G. Barone
- Department of EngineeringElectrical Engineering DivisionUniversity of CambridgeCambridgeCB3 0FAUK
- Department of Neurosurgery, Houston MethodistHouston77030USA
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeCB2 0QQUK
| |
Collapse
|
22
|
Li M, Huang W, Duan L, Sun F. Control Intracellular Protein Condensates with Light. ACS Synth Biol 2024; 13:3799-3811. [PMID: 39622001 DOI: 10.1021/acssynbio.4c00305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024]
Abstract
Protein phase transitions are gaining traction among biologists for their wide-ranging roles in biological regulation. However, achieving precise control over these phenomena in vivo remains a formidable task. Optogenetic techniques present us with a potential means to control protein phase behavior with spatiotemporal precision. This review delves into the design of optogenetic tools, particularly those aimed at manipulating protein phase transitions in complex biological systems. We begin by discussing the pivotal roles of subcellular phase transitions in physiological and pathological processes. Subsequently, we offer a thorough examination of the evolution of optogenetic tools and their applications in regulating these protein phase behaviors. Furthermore, we highlight the tailored design of optogenetic tools for controlling protein phase transitions and the construction of synthetic condensates using these innovative techniques. In the long run, the development of optogenetic tools not only holds the potential to elucidate the roles of protein phase transitions in various physiological processes but also to antagonize pathological ones to reinstate cellular homeostasis, thus bringing about novel therapeutic strategies. The integration of optogenetic techniques into the study of protein phase transitions represents a significant step forward in our understanding and manipulation of biology at the subcellular level.
Collapse
Affiliation(s)
- Manjia Li
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Hong Kong SAR, China
| | - Weiqi Huang
- College of Computer and Information Sciences, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China
| | - Liting Duan
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, China
| | - Fei Sun
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Hong Kong SAR, China
- Greater Bay Biomedical InnoCenter, Shenzhen Bay Laboratory, Shenzhen 518036, China
- Research Institute of Tsinghua, Pearl River Delta, Guangzhou 510530, China
| |
Collapse
|
23
|
Wiegand T, Liu J, Vogeley L, LuValle-Burke I, Geisler J, Fritsch AW, Hyman AA, Grill SW. Actin polymerization counteracts prewetting of N-WASP on supported lipid bilayers. Proc Natl Acad Sci U S A 2024; 121:e2407497121. [PMID: 39630867 PMCID: PMC11648614 DOI: 10.1073/pnas.2407497121] [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: 04/15/2024] [Accepted: 10/27/2024] [Indexed: 12/07/2024] Open
Abstract
Cortical condensates, transient punctate-like structures rich in actin and the actin nucleation pathway member Neural Wiskott-Aldrich syndrome protein (N-WASP), form during activation of the actin cortex in the Caenorhabditis elegans oocyte. Their emergence and spontaneous dissolution is linked to a phase separation process driven by chemical kinetics. However, the mechanisms that drive the onset of cortical condensate formation near membranes remain unexplored. Here, using a reconstituted phase separation assay of cortical condensate proteins, we demonstrate that the key component, N-WASP, can collectively undergo surface condensation on supported lipid bilayers via a prewetting transition. Actin partitions into the condensates, where it polymerizes and counteracts the N-WASP prewetting transition. Taken together, the dynamics of condensate-assisted cortex formation appear to be controlled by a balance between surface-assisted condensate formation and polymer-driven condensate dissolution. This opens perspectives for understanding how the formation of complex intracellular structures is affected and controlled by phase separation.
Collapse
Affiliation(s)
- Tina Wiegand
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden01307, Germany
- Max Planck Institute for the Physics of Complex Systems, Dresden01187, Germany
| | - Jinghui Liu
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden01307, Germany
- Center for Systems Biology Dresden, Dresden01307, Germany
| | - Lutz Vogeley
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden01307, Germany
| | - Isabel LuValle-Burke
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden01307, Germany
- Max Planck Institute for the Physics of Complex Systems, Dresden01187, Germany
| | - Jan Geisler
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden01307, Germany
- Max Planck School Matter to Life, Heidelberg69120, Germany
| | - Anatol W. Fritsch
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden01307, Germany
- Max Planck Institute for the Physics of Complex Systems, Dresden01187, Germany
| | - Anthony A. Hyman
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden01307, Germany
- Cluster of Excellence Physics of Life, Technische Universität Dresden, Dresden01307, Germany
| | - Stephan W. Grill
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden01307, Germany
- Cluster of Excellence Physics of Life, Technische Universität Dresden, Dresden01307, Germany
| |
Collapse
|
24
|
Yang W, Wang Y, Liu G, Wang Y, Wu C. TPM4 condensates glycolytic enzymes and facilitates actin reorganization under hyperosmotic stress. Cell Discov 2024; 10:120. [PMID: 39622827 PMCID: PMC11612400 DOI: 10.1038/s41421-024-00744-2] [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: 07/12/2024] [Accepted: 10/20/2024] [Indexed: 12/06/2024] Open
Abstract
Actin homeostasis is fundamental for cell structure and consumes a large portion of cellular ATP. It has been documented in the literature that certain glycolytic enzymes can interact with actin, indicating an intricate interplay between the cytoskeleton and cellular metabolism. Here we report that hyperosmotic stress triggers actin severing and subsequent phase separation of the actin-binding protein tropomyosin 4 (TPM4). TPM4 condensates recruit glycolytic enzymes such as HK2, PFKM, and PKM2, while wetting actin filaments. Notably, the condensates of TPM4 and glycolytic enzymes are enriched of NADH and ATP, suggestive of their functional importance in cell metabolism. At cellular level, actin filament assembly is enhanced upon hyperosmotic stress and TPM4 condensation, while depletion of TPM4 impairs osmolarity-induced actin reorganization. At tissue level, colocalized condensates of TPM4 and glycolytic enzymes are observed in renal tissues subjected to hyperosmotic stress. Together, our findings suggest that stress-induced actin perturbation may act on TPM4 to organize glycolytic hubs that tether energy production to cytoskeletal reorganization.
Collapse
Affiliation(s)
- Wenzhong Yang
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- International Cancer Institute, Peking University, Beijing, China
- Institute of Advanced Clinical Medicine, State Key Laboratory of Molecular Oncology, Beijing, China
| | - Yuan Wang
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- International Cancer Institute, Peking University, Beijing, China
- Institute of Advanced Clinical Medicine, State Key Laboratory of Molecular Oncology, Beijing, China
| | - Geyao Liu
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- International Cancer Institute, Peking University, Beijing, China
- Institute of Advanced Clinical Medicine, State Key Laboratory of Molecular Oncology, Beijing, China
| | - Yan Wang
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- International Cancer Institute, Peking University, Beijing, China
- Institute of Advanced Clinical Medicine, State Key Laboratory of Molecular Oncology, Beijing, China
| | - Congying Wu
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China.
- International Cancer Institute, Peking University, Beijing, China.
- Institute of Advanced Clinical Medicine, State Key Laboratory of Molecular Oncology, Beijing, China.
| |
Collapse
|
25
|
Pasquale EB. Eph receptor signaling complexes in the plasma membrane. Trends Biochem Sci 2024; 49:1079-1096. [PMID: 39537538 PMCID: PMC11967910 DOI: 10.1016/j.tibs.2024.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 09/24/2024] [Accepted: 10/04/2024] [Indexed: 11/16/2024]
Abstract
Eph receptor tyrosine kinases, together with their cell surface-anchored ephrin ligands, constitute an important cell-cell communication system that regulates physiological and pathological processes in most cell types. This review focuses on the multiple mechanisms by which Eph receptors initiate signaling via the formation of protein complexes in the plasma membrane. Upon ephrin binding, Eph receptors assemble into oligomers that can further aggregate into large complexes. Eph receptors also mediate ephrin-independent signaling through interplay with intracellular kinases or other cell-surface receptors. The distinct characteristics of Eph receptor family members, as well as their conserved domain structure, provide a framework for understanding their functional differences and redundancies. Possible areas of interest for future investigations of Eph receptor signaling complexes are also highlighted.
Collapse
Affiliation(s)
- Elena B Pasquale
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA.
| |
Collapse
|
26
|
Subedi S, Nag N, Shukla H, Padhi AK, Tripathi T. Comprehensive analysis of liquid-liquid phase separation propensities of HSV-1 proteins and their interaction with host factors. J Cell Biochem 2024; 125:e30480. [PMID: 37796176 DOI: 10.1002/jcb.30480] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 09/08/2023] [Accepted: 09/17/2023] [Indexed: 10/06/2023]
Abstract
In recent years, it has been shown that the liquid-liquid phase separation (LLPS) of virus proteins plays a crucial role in their life cycle. It promotes the formation of viral replication organelles, concentrating viral components for efficient replication and facilitates the assembly of viral particles. LLPS has emerged as a crucial process in the replication and assembly of herpes simplex virus-1 (HSV-1). Recent studies have identified several HSV-1 proteins involved in LLPS, including the myristylated tegument protein UL11 and infected cell protein 4; however, a complete proteome-level understanding of the LLPS-prone HSV-1 proteins is not available. We provide a comprehensive analysis of the HSV-1 proteome and explore the potential of its proteins to undergo LLPS. By integrating sequence analysis, prediction algorithms and an array of tools and servers, we identified 10 HSV-1 proteins that exhibit high LLPS potential. By analysing the amino acid sequences of the LLPS-prone proteins, we identified specific sequence motifs and enriched amino acid residues commonly found in LLPS-prone regions. Our findings reveal a diverse range of LLPS-prone proteins within the HSV-1, which are involved in critical viral processes such as replication, transcriptional regulation and assembly of viral particles. This suggests that LLPS might play a crucial role in facilitating the formation of specialized viral replication compartments and the assembly of HSV-1 virion. The identification of LLPS-prone proteins in HSV-1 opens up new avenues for understanding the molecular mechanisms underlying viral pathogenesis. Our work provides valuable insights into the LLPS landscape of HSV-1, highlighting potential targets for further experimental validation and enhancing our understanding of viral replication and pathogenesis.
Collapse
Affiliation(s)
- Sushma Subedi
- Molecular and Structural Biophysics Laboratory, Department of Biochemistry, North-Eastern Hill University, Shillong, India
| | - Niharika Nag
- Molecular and Structural Biophysics Laboratory, Department of Biochemistry, North-Eastern Hill University, Shillong, India
| | - Harish Shukla
- Molecular and Structural Biophysics Laboratory, Department of Biochemistry, North-Eastern Hill University, Shillong, India
| | - Aditya K Padhi
- Laboratory for Computational Biology & Biomolecular Design, School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi, India
| | - Timir Tripathi
- Molecular and Structural Biophysics Laboratory, Department of Biochemistry, North-Eastern Hill University, Shillong, India
- Department of Zoology, North-Eastern Hill University, Shillong, India
| |
Collapse
|
27
|
Li M, Yang X, Zhang D, Tian Y, Jia ZC, Liu WH, Hao RR, Chen YS, Chen MX, Liu YG. A story of two kingdoms: unravelling the intricacies of protein phase separation in plants and animals. Crit Rev Biotechnol 2024:1-21. [PMID: 39592156 DOI: 10.1080/07388551.2024.2425989] [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: 03/27/2024] [Revised: 08/17/2024] [Accepted: 10/20/2024] [Indexed: 11/28/2024]
Abstract
The biomolecular condensates (BCs) formed by proteins through phase separation provide the necessary space and raw materials for the orderly progression of cellular activities, and on this basis, various membraneless organelles (MLOs) are formed. The occurrence of eukaryotic phase separation is driven by multivalent interactions from intrinsically disordered regions (IDRs) and/or specific protein/nucleic acid binding domains and is regulated by various environmental factors. In plant and animal cells, the MLOs involved in gene expression regulation, stress response, and mitotic control display similar functions and mechanisms. In contrast, the phase separation related to reproductive development and immune regulation differs significantly between the two kingdoms owing to their distinct cell structures and nutritional patterns. In addition, animals and plants each exhibit unique protein phase separation activities, such as neural regulation and light signal response. By comparing the similarities and differences in the formation mechanism and functional regulation of known protein phase separation, we elucidated its importance in the evolution, differentiation, and environmental adaptation of both animals and plants. The significance of studying protein phase separation for enhancing biological quality of life has been further emphasized.
Collapse
Affiliation(s)
- Min Li
- State Key Laboratory of Wheat Improvement, College of Life Science, Shandong Agricultural University, Taian, China
| | - Xue Yang
- State Key Laboratory of Wheat Improvement, College of Life Science, Shandong Agricultural University, Taian, China
| | - Di Zhang
- Department of Biology, Hong Kong Baptist University, and State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Yuan Tian
- State Key Laboratory of Wheat Improvement, College of Life Science, Shandong Agricultural University, Taian, China
- State Key Laboratory of Tree Genetics and Breeding, Co-Innovation Center for Sustainable Forestry in Southern China, Key Laboratory of State Forestry and Grassland Administration on Subtropical Forest Biodiversity Conservation, College of Life Sciences, Nanjing Forestry University, Nanjing, China
| | - Zi-Chang Jia
- State Key Laboratory of Wheat Improvement, College of Life Science, Shandong Agricultural University, Taian, China
| | - Wen-Hui Liu
- State Key Laboratory of Wheat Improvement, College of Life Science, Shandong Agricultural University, Taian, China
| | - Rui-Rui Hao
- State Key Laboratory of Tree Genetics and Breeding, Co-Innovation Center for Sustainable Forestry in Southern China, Key Laboratory of State Forestry and Grassland Administration on Subtropical Forest Biodiversity Conservation, College of Life Sciences, Nanjing Forestry University, Nanjing, China
| | - Yun-Sheng Chen
- Clinical Laboratory, Shenzhen Children's Hospital, Shenzhen, China
| | - Mo-Xian Chen
- State Key Laboratory of Tree Genetics and Breeding, Co-Innovation Center for Sustainable Forestry in Southern China, Key Laboratory of State Forestry and Grassland Administration on Subtropical Forest Biodiversity Conservation, College of Life Sciences, Nanjing Forestry University, Nanjing, China
| | - Ying-Gao Liu
- State Key Laboratory of Wheat Improvement, College of Life Science, Shandong Agricultural University, Taian, China
| |
Collapse
|
28
|
Mu C, Liu P, Liu L, Wang Y, Liu K, Li X, Li G, Cheng J, Bu M, Chen H, Tang M, Yao Y, Guan J, Ma T, Zhou Z, Wu Q, Li J, Guo H, Xia K, Hu Z, Peng X, Lang B, Li F, Chen XW, Xu Z, Yuan L. KCTD10 p.C124W variant contributes to schizophrenia by attenuating LLPS-mediated synapse formation. Proc Natl Acad Sci U S A 2024; 121:e2400464121. [PMID: 39565307 PMCID: PMC11621769 DOI: 10.1073/pnas.2400464121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 10/23/2024] [Indexed: 11/21/2024] Open
Abstract
KCTD10, a member of the potassium channel tetramerization domain (KCTD) family, is implicated in neuropsychiatric disorders and functions as a substrate recognition component within the RING-type ubiquitin ligase complex. A rare de novo variant of KCTD10, p.C124W, was identified in schizophrenia cases, yet its underlying pathogenesis remains unexplored. Here, we demonstrate that heterozygous KCTD10 C124W mice display pronounced synaptic abnormalities and exhibit schizophrenia-like behaviors. Mechanistically, we reveal that KCTD10 undergoes liquid-liquid phase separation (LLPS), a process orchestrated by its intrinsically disordered region (IDR). p.C124W mutation disrupts this LLPS capability, leading to diminished degradation of RHOB and subsequent excessive accumulation in the postsynaptic density fractions. Notably, neither IDR deletion nor p.C124W mutation in KCTD10 mitigates the synaptic abnormalities caused by Kctd10 deficiency. Thus, our findings implicate that LLPS may be associated with the pathogenesis of KCTD10-associated brain disorders and highlight the potential of targeting RHOB as a therapeutic strategy for diseases linked to mutations in KCTD10 or RHOB.
Collapse
Affiliation(s)
- Chenjun Mu
- Center for Medical Genetics, Hunan Key Laboratory of Medical Genetics, Key Lab of Rare Pediatric Diseases of Ministry of Education, School of Life Science, Central South University, Changsha, Hunan410078, China
| | - Pan Liu
- Center for Medical Genetics, Hunan Key Laboratory of Medical Genetics, Key Lab of Rare Pediatric Diseases of Ministry of Education, School of Life Science, Central South University, Changsha, Hunan410078, China
| | - Liang Liu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing100053, China
| | - Yaqing Wang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Beijing100101, China
| | - Kefu Liu
- Center for Medical Genetics, Hunan Key Laboratory of Medical Genetics, Key Lab of Rare Pediatric Diseases of Ministry of Education, School of Life Science, Central South University, Changsha, Hunan410078, China
| | - Xiangyu Li
- Center for Medical Genetics, Hunan Key Laboratory of Medical Genetics, Key Lab of Rare Pediatric Diseases of Ministry of Education, School of Life Science, Central South University, Changsha, Hunan410078, China
| | - Guozhong Li
- Center for Medical Genetics, Hunan Key Laboratory of Medical Genetics, Key Lab of Rare Pediatric Diseases of Ministry of Education, School of Life Science, Central South University, Changsha, Hunan410078, China
| | - Jianbo Cheng
- Center for Medical Genetics, Hunan Key Laboratory of Medical Genetics, Key Lab of Rare Pediatric Diseases of Ministry of Education, School of Life Science, Central South University, Changsha, Hunan410078, China
| | - Mengyao Bu
- Center for Medical Genetics, Hunan Key Laboratory of Medical Genetics, Key Lab of Rare Pediatric Diseases of Ministry of Education, School of Life Science, Central South University, Changsha, Hunan410078, China
| | - Han Chen
- Center for Medical Genetics, Hunan Key Laboratory of Medical Genetics, Key Lab of Rare Pediatric Diseases of Ministry of Education, School of Life Science, Central South University, Changsha, Hunan410078, China
| | - Manpei Tang
- Center for Medical Genetics, Hunan Key Laboratory of Medical Genetics, Key Lab of Rare Pediatric Diseases of Ministry of Education, School of Life Science, Central South University, Changsha, Hunan410078, China
| | - Yuanhang Yao
- Center for Life Sciences, Institute of Molecular Medicine, School of Future Technology, Peking University, Beijing100871, China
| | - Jun Guan
- Center for Medical Genetics, Hunan Key Laboratory of Medical Genetics, Key Lab of Rare Pediatric Diseases of Ministry of Education, School of Life Science, Central South University, Changsha, Hunan410078, China
| | - Tiantian Ma
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Beijing100101, China
| | - Zhengrong Zhou
- Department of Basic Medical Sciences, Neuroscience Center, Shantou University Medical College, Shantou, Guangdong515041, China
| | - Qingfeng Wu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Beijing100101, China
| | - Jiada Li
- Center for Medical Genetics, Hunan Key Laboratory of Medical Genetics, Key Lab of Rare Pediatric Diseases of Ministry of Education, School of Life Science, Central South University, Changsha, Hunan410078, China
- Hunan Key Laboratory of Animal Models for Human Diseases, School of Life Science, Central South University, Changsha, Hunan410078, China
| | - Hui Guo
- Center for Medical Genetics, Hunan Key Laboratory of Medical Genetics, Key Lab of Rare Pediatric Diseases of Ministry of Education, School of Life Science, Central South University, Changsha, Hunan410078, China
- Hunan Key Laboratory of Animal Models for Human Diseases, School of Life Science, Central South University, Changsha, Hunan410078, China
| | - Kun Xia
- Center for Medical Genetics, Hunan Key Laboratory of Medical Genetics, Key Lab of Rare Pediatric Diseases of Ministry of Education, School of Life Science, Central South University, Changsha, Hunan410078, China
- Hunan Key Laboratory of Animal Models for Human Diseases, School of Life Science, Central South University, Changsha, Hunan410078, China
| | - Zhengmao Hu
- Center for Medical Genetics, Hunan Key Laboratory of Medical Genetics, Key Lab of Rare Pediatric Diseases of Ministry of Education, School of Life Science, Central South University, Changsha, Hunan410078, China
- Hunan Key Laboratory of Animal Models for Human Diseases, School of Life Science, Central South University, Changsha, Hunan410078, China
| | - Xiaoqing Peng
- Center for Medical Genetics, Hunan Key Laboratory of Medical Genetics, Key Lab of Rare Pediatric Diseases of Ministry of Education, School of Life Science, Central South University, Changsha, Hunan410078, China
- Hunan Key Laboratory of Animal Models for Human Diseases, School of Life Science, Central South University, Changsha, Hunan410078, China
| | - Bing Lang
- Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, Hunan410011, China
| | - Faxiang Li
- Center for Medical Genetics, Hunan Key Laboratory of Medical Genetics, Key Lab of Rare Pediatric Diseases of Ministry of Education, School of Life Science, Central South University, Changsha, Hunan410078, China
- Hunan Key Laboratory of Animal Models for Human Diseases, School of Life Science, Central South University, Changsha, Hunan410078, China
| | - Xiao-wei Chen
- Center for Life Sciences, Institute of Molecular Medicine, School of Future Technology, Peking University, Beijing100871, China
| | - Zhiheng Xu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Beijing100101, China
| | - Ling Yuan
- Center for Medical Genetics, Hunan Key Laboratory of Medical Genetics, Key Lab of Rare Pediatric Diseases of Ministry of Education, School of Life Science, Central South University, Changsha, Hunan410078, China
- Hunan Key Laboratory of Animal Models for Human Diseases, School of Life Science, Central South University, Changsha, Hunan410078, China
| |
Collapse
|
29
|
Guzikowski NJ, Kavalali ET. Functional specificity of liquid-liquid phase separation at the synapse. Nat Commun 2024; 15:10103. [PMID: 39572583 PMCID: PMC11582584 DOI: 10.1038/s41467-024-54423-7] [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: 03/12/2024] [Accepted: 11/08/2024] [Indexed: 11/24/2024] Open
Abstract
The mechanisms that enable synapses to achieve temporally and spatially precise signaling at nano-scale while being fluid with the cytosol are poorly understood. Liquid-liquid phase separation (LLPS) is emerging as a key principle governing subcellular organization; however, the impact of synaptic LLPS on neurotransmission is unclear. Here, using rat primary hippocampal cultures, we show that robust disruption of neuronal LLPS with aliphatic alcohols severely dysregulates action potential-dependent neurotransmission, while spontaneous neurotransmission persists. Synaptic LLPS maintains synaptic vesicle pool clustering and recycling as well as the precise organization of active zone RIM1/2 and Munc13 nanoclusters, thus supporting fast evoked Ca2+-dependent release. These results indicate although LLPS is necessary within the nanodomain of the synapse, the disruption of this nano-organization largely spares spontaneous neurotransmission. Therefore, properties of in vitro micron sized liquid condensates translate to the nano-structure of the synapse in a functionally specific manner regulating action potential-evoked release.
Collapse
Affiliation(s)
- Natalie J Guzikowski
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
| | - Ege T Kavalali
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA.
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
30
|
Scorrano G, Di Francesco L, Di Ludovico A, Chiarelli F, Matricardi S. Exploring the Landscape of Pre- and Post-Synaptic Pediatric Disorders with Epilepsy: A Narrative Review on Molecular Mechanisms Involved. Int J Mol Sci 2024; 25:11982. [PMID: 39596051 PMCID: PMC11593774 DOI: 10.3390/ijms252211982] [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: 10/07/2024] [Revised: 11/03/2024] [Accepted: 11/06/2024] [Indexed: 11/28/2024] Open
Abstract
Neurodevelopmental disorders (NDDs) are a group of conditions affecting brain development, with variable degrees of severity and heterogeneous clinical features. They include intellectual disability (ID), autism spectrum disorder (ASD), attention-deficit/hyperactivity disorder (ADHD), often coexisting with epilepsy, extra-neurological comorbidities, and multisystemic involvement. In recent years, next-generation sequencing (NGS) technologies allowed the identification of several gene pathogenic variants etiologically related to these disorders in a large cohort of affected children. These genes encode proteins involved in synaptic homeostasis, such as SNARE proteins, implicated in calcium-triggered pre-synaptic release of neurotransmitters, or channel subunit proteins, such as post-synaptic ionotropic glutamate receptors involved in the brain's fast excitatory neurotransmission. In this narrative review, we dissected emerged molecular mechanisms related to NDDs and epilepsy due to defects in pre- and post-synaptic transmission. We focused on the most recently discovered SNAREopathies and AMPA-related synaptopathies.
Collapse
Affiliation(s)
- Giovanna Scorrano
- Department of Pediatrics, University of Chieti-Pescara, Sant’Annunziata Hospital, 66100 Chieti, Italy; (G.S.); (A.D.L.); (F.C.)
| | - Ludovica Di Francesco
- Department of Neonatology, University of L’Aquila, San Salvatore Hospital, 67100 L’Aquila, Italy;
| | - Armando Di Ludovico
- Department of Pediatrics, University of Chieti-Pescara, Sant’Annunziata Hospital, 66100 Chieti, Italy; (G.S.); (A.D.L.); (F.C.)
| | - Francesco Chiarelli
- Department of Pediatrics, University of Chieti-Pescara, Sant’Annunziata Hospital, 66100 Chieti, Italy; (G.S.); (A.D.L.); (F.C.)
| | - Sara Matricardi
- Department of Pediatrics, University of Chieti-Pescara, Sant’Annunziata Hospital, 66100 Chieti, Italy; (G.S.); (A.D.L.); (F.C.)
| |
Collapse
|
31
|
Kim N, Yun H, Lee H, Yoo JY. Interplay between membranes and biomolecular condensates in the regulation of membrane-associated cellular processes. Exp Mol Med 2024; 56:2357-2364. [PMID: 39482532 PMCID: PMC11612285 DOI: 10.1038/s12276-024-01337-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 08/04/2024] [Accepted: 08/11/2024] [Indexed: 11/03/2024] Open
Abstract
Liquid‒liquid phase separation (LLPS) has emerged as a key mechanism for organizing cellular spaces independent of membranes. Biomolecular condensates, which assemble through LLPS, exhibit distinctive liquid droplet-like behavior and can exchange constituents with their surroundings. The regulation of condensate phases, including transitions from a liquid state to gel or irreversible aggregates, is important for their physiological functions and for controlling pathological progression, as observed in neurodegenerative diseases and cancer. While early studies on biomolecular condensates focused primarily on those in fluidic environments such as the cytosol, recent discoveries have revealed their existence in close proximity to, on, or even comprising membranes. The aim of this review is to provide an overview of the properties of membrane-associated condensates in a cellular context and their biological functions in relation to membranes.
Collapse
Affiliation(s)
- Nari Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea.
| | - Hyeri Yun
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Hojin Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Joo-Yeon Yoo
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea.
| |
Collapse
|
32
|
Koja Y, Arakawa T, Yoritaka Y, Joshima Y, Kobayashi H, Toda K, Takeda S. Basic design of artificial membrane-less organelles using condensation-prone proteins in plant cells. Commun Biol 2024; 7:1396. [PMID: 39462114 PMCID: PMC11514006 DOI: 10.1038/s42003-024-07102-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 10/18/2024] [Indexed: 10/28/2024] Open
Abstract
Membrane-less organelles, formed by the condensation of biomolecules, play a pivotal role in eukaryotes. Artificial membrane-less organelles and condensates are effective tools for the creation of new cellular functions. However, it is poorly understood how to control the properties that affect condensate function, particularly in plants. Here, we report the construction of model artificial condensates using the condensation-prone proteins OsJAZ2 and AtFCA in a transient assay using rice (Oryza sativa) cells, and how condensate properties, such as subcellular localization, protein mobility, and size can be altered. We showed that proteins of interest can be recruited to condensates using nanobodies or chemically induced dimerization. Furthermore, by combining two types of condensation-prone proteins, we demonstrated that artificial hybrid condensates with heterogeneous material properties could be constructed. Finally, we showed that modified artificial condensates can be constructed in transgenic Arabidopsis thaliana plants. These results provide a framework for the basic design of synthetic membrane-less organelles in plants.
Collapse
Affiliation(s)
- Yoshito Koja
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Takuya Arakawa
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Yusuke Yoritaka
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Yu Joshima
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
- Bioscience and Biotechnology Center, Nagoya University, Nagoya, Japan
| | - Hazuki Kobayashi
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Kenta Toda
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Shin Takeda
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan.
- Bioscience and Biotechnology Center, Nagoya University, Nagoya, Japan.
| |
Collapse
|
33
|
Yu H, Wang W. Modulation of heteromeric glycine receptor function through high concentration clustering. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.17.618879. [PMID: 39464082 PMCID: PMC11507885 DOI: 10.1101/2024.10.17.618879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Ion channels are targeted by many drugs for treating neurological, musculoskeletal, renal and other diseases. These drugs bind to and alter the function of individual channels to achieve desired therapeutic effects. However, many ion channels function in high concentration clusters in their native environment. It is unclear if and how clustering modulates ion channel function. Human heteromeric glycine receptors (GlyRs) are the major inhibitory neurotransmitter receptors in the spinal cord and are active targets for developing chronic pain medications. We show that the α2β heteromeric GlyR assembles with the master postsynaptic scaffolding gephyrin (GPHN) into micron-sized clustered at the plasma membrane after heterologous expression. The inhibitory trans- synaptic adhesion protein neuroligin-2 (NL2) further increases both the cluster sizes and GlyR concentration. The apparent glycine affinity increases monotonically as a function of GlyR concentration but not with cluster size. We also show that ligand re-binding to adjacent GlyRs alters kinetics but not chemical equilibrium. A positively charged N- terminus sequence of the GlyR β subunit was further identified essential for glycine affinity modulation through clustering. Taken together, we propose a mechanism where clustering enhances local electrostatic potential, which in turn concentrates ions and ligands, modulating the function of GlyR. This mechanism is likely universal across ion channel clusters found ubiquitously in biology and provides new perspectives in possible pharmaceutical development.
Collapse
|
34
|
Zhu M, Xu H, Jin Y, Kong X, Xu B, Liu Y, Yu H. Synaptotagmin-1 undergoes phase separation to regulate its calcium-sensitive oligomerization. J Cell Biol 2024; 223:e202311191. [PMID: 38980206 PMCID: PMC11232894 DOI: 10.1083/jcb.202311191] [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: 12/01/2023] [Revised: 04/17/2024] [Accepted: 06/11/2024] [Indexed: 07/10/2024] Open
Abstract
Synaptotagmin-1 (Syt1) is a calcium sensor that regulates synaptic vesicle fusion in synchronous neurotransmitter release. Syt1 interacts with negatively charged lipids and the SNARE complex to control the fusion event. However, it remains incompletely understood how Syt1 mediates Ca2+-trigged synaptic vesicle fusion. Here, we discovered that Syt1 undergoes liquid-liquid phase separation (LLPS) to form condensates both in vitro and in living cells. Syt1 condensates play a role in vesicle attachment to the PM and efficiently recruit SNAREs and complexin, which may facilitate the downstream synaptic vesicle fusion. We observed that Syt1 condensates undergo a liquid-to-gel-like phase transition, reflecting the formation of Syt1 oligomers. The phase transition can be blocked or reversed by Ca2+, confirming the essential role of Ca2+ in Syt1 oligomer disassembly. Finally, we showed that the Syt1 mutations causing Syt1-associated neurodevelopmental disorder impair the Ca2+-driven phase transition. These findings reveal that Syt1 undergoes LLPS and a Ca2+-sensitive phase transition, providing new insights into Syt1-mediated vesicle fusion.
Collapse
Affiliation(s)
- Min Zhu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Han Xu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Yulei Jin
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Xiaoxu Kong
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Bingkuan Xu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Yinghui Liu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Haijia Yu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| |
Collapse
|
35
|
Reshetniak S, Bogaciu CA, Bonn S, Brose N, Cooper BH, D'Este E, Fauth M, Fernández-Busnadiego R, Fiosins M, Fischer A, Georgiev SV, Jakobs S, Klumpp S, Köster S, Lange F, Lipstein N, Macarrón-Palacios V, Milovanovic D, Moser T, Müller M, Opazo F, Outeiro TF, Pape C, Priesemann V, Rehling P, Salditt T, Schlüter O, Simeth N, Steinem C, Tchumatchenko T, Tetzlaff C, Tirard M, Urlaub H, Wichmann C, Wolf F, Rizzoli SO. The synaptic vesicle cluster as a controller of pre- and postsynaptic structure and function. J Physiol 2024. [PMID: 39367860 DOI: 10.1113/jp286400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 09/11/2024] [Indexed: 10/07/2024] Open
Abstract
The synaptic vesicle cluster (SVC) is an essential component of chemical synapses, which provides neurotransmitter-loaded vesicles during synaptic activity, at the same time as also controlling the local concentrations of numerous exo- and endocytosis cofactors. In addition, the SVC hosts molecules that participate in other aspects of synaptic function, from cytoskeletal components to adhesion proteins, and affects the location and function of organelles such as mitochondria and the endoplasmic reticulum. We argue here that these features extend the functional involvement of the SVC in synapse formation, signalling and plasticity, as well as synapse stabilization and metabolism. We also propose that changes in the size of the SVC coalesce with changes in the postsynaptic compartment, supporting the interplay between pre- and postsynaptic dynamics. Thereby, the SVC could be seen as an 'all-in-one' regulator of synaptic structure and function, which should be investigated in more detail, to reveal molecular mechanisms that control synaptic function and heterogeneity.
Collapse
Affiliation(s)
- Sofiia Reshetniak
- Institute for Neuro- and Sensory Physiology and Biostructural Imaging of Neurodegeneration (BIN) Center, University Medical Center Göttingen, Göttingen, Germany
| | - Cristian A Bogaciu
- Institute for Neuro- and Sensory Physiology and Biostructural Imaging of Neurodegeneration (BIN) Center, University Medical Center Göttingen, Göttingen, Germany
| | - Stefan Bonn
- Institute of Medical Systems Biology, Center for Molecular Neurobiology Hamburg, Hamburg, Germany
| | - Nils Brose
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Benjamin H Cooper
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Elisa D'Este
- Optical Microscopy Facility, Max Planck Institute for Medical Research, Heidelberg, Germany
| | - Michael Fauth
- Georg-August-University Göttingen, Faculty of Physics, Institute for the Dynamics of Complex Systems, Friedrich-Hund-Platz 1, Göttingen, Germany
| | - Rubén Fernández-Busnadiego
- Institute of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Maksims Fiosins
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| | - André Fischer
- German Center for Neurodegenerative Diseases, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Svilen V Georgiev
- Institute for Neuro- and Sensory Physiology and Biostructural Imaging of Neurodegeneration (BIN) Center, University Medical Center Göttingen, Göttingen, Germany
| | - Stefan Jakobs
- Research Group Structure and Dynamics of Mitochondria, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Stefan Klumpp
- Theoretical Biophysics Group, Institute for the Dynamics of Complex Systems, Georg-August University Göttingen, Göttingen, Germany
| | - Sarah Köster
- Institute for X-Ray Physics, Georg-August University Göttingen, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Felix Lange
- Research Group Structure and Dynamics of Mitochondria, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Noa Lipstein
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| | | | - Dragomir Milovanovic
- Laboratory of Molecular Neuroscience, German Center for Neurodegenerative Diseases, Berlin, Germany
| | - Tobias Moser
- Institute for Auditory Neuroscience, University Medical Center Göttingen, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Marcus Müller
- Institute for Theoretical Physics, Georg-August University Göttingen, Göttingen, Germany
| | - Felipe Opazo
- Institute for Neuro- and Sensory Physiology and Biostructural Imaging of Neurodegeneration (BIN) Center, University Medical Center Göttingen, Göttingen, Germany
| | - Tiago F Outeiro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany
| | - Constantin Pape
- Institute of Computer Science, Georg-August University Göttingen, Göttingen, Germany
| | - Viola Priesemann
- Georg-August-University Göttingen, Faculty of Physics, Institute for the Dynamics of Complex Systems, Friedrich-Hund-Platz 1, Göttingen, Germany
- Max-Planck Institute for Dynamics and Self-Organization, Am Fassberg 17, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Peter Rehling
- Department of Cellular Biochemistry, University Medical Center Göttingen, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Tim Salditt
- Institute for X-Ray Physics, Georg-August University Göttingen, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Oliver Schlüter
- Clinic for Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
| | - Nadja Simeth
- Institute of Organic and Biomolecular Chemistry, Georg-August University Göttingen, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Claudia Steinem
- Institute of Organic and Biomolecular Chemistry, Georg-August University Göttingen, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Tatjana Tchumatchenko
- Institute of Experimental Epileptology and Cognition Research, University of Bonn Medical Center, Bonn, Germany
| | - Christian Tetzlaff
- Institute for Neuro- and Sensory Physiology and Biostructural Imaging of Neurodegeneration (BIN) Center, University Medical Center Göttingen, Göttingen, Germany
| | - Marilyn Tirard
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Henning Urlaub
- Bioanalytical Mass Spectrometry, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Carolin Wichmann
- Institute for Auditory Neuroscience University Medical Center Göttingen, Göttingen, Germany
- Biostructural Imaging of Neurodegeneration (BIN) Center, University Medical Center Göttingen, Göttingen, Germany
| | - Fred Wolf
- Max-Planck-Institute for Dynamics and Self-Organization, 37077 Göttingen and Institute for Dynamics of Biological Networks, Georg-August University Göttingen, Göttingen, Germany
| | - Silvio O Rizzoli
- Institute for Neuro- and Sensory Physiology and Biostructural Imaging of Neurodegeneration (BIN) Center, University Medical Center Göttingen, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| |
Collapse
|
36
|
Lv W, Wang Y. Neural Influences on Tumor Progression Within the Central Nervous System. CNS Neurosci Ther 2024; 30:e70097. [PMID: 39469896 PMCID: PMC11519750 DOI: 10.1111/cns.70097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 09/21/2024] [Accepted: 10/13/2024] [Indexed: 10/30/2024] Open
Abstract
For decades, researchers have studied how brain tumors, the immune system, and drugs interact. With the advances in cancer neuroscience, which centers on defining and therapeutically targeting nervous system-cancer interactions, both within the local tumor microenvironment (TME) and on a systemic level, the subtle relationship between neurons and tumors in the central nervous system (CNS) has been deeply studied. Neurons, as the executors of brain functional activities, have been shown to significantly influence the emergence and development of brain tumors, including both primary and metastatic tumors. They engage with tumor cells via chemical or electrical synapses, directly regulating tumors or via intricate coupling networks, and also contribute to the TME through paracrine signaling, secreting proteins that exert regulatory effects. For instance, in a study involving a mouse model of glioblastoma, the authors observed a 42% increase in tumor volume when neuronal activity was stimulated, compared to controls (p < 0.01), indicating a direct correlation between neural activity and tumor growth. These thought-provoking results offer promising new strategies for brain tumor therapies, highlighting the potential of neuronal modulation to curb tumor progression. Future strategies may focus on developing drugs to inhibit or neutralize proteins and other bioactive substances secreted by neurons, break synaptic connections and interactions between infiltrating cells and tumor cells, as well as disrupt electrical coupling within glioma cell networks. By harnessing the insights gained from this research, we aspire to usher in a new era of brain tumor therapies that are both more potent and precise.
Collapse
Affiliation(s)
- Wenhao Lv
- Affiliated Hospital of Hangzhou Normal UniversityHangzhou Normal UniversityHangzhouZhejiangChina
- School of PharmacyHangzhou Normal UniversityHangzhouZhejiangChina
| | - Yongjie Wang
- School of PharmacyHangzhou Normal UniversityHangzhouZhejiangChina
| |
Collapse
|
37
|
Yu L, Li Y, Lv Y, Gu B, Cai J, Liu QS, Zhao L. Treadmill Exercise Facilitates Synaptic Plasticity in APP/PS1 Mice by Regulating Hippocampal AMPAR Activity. Cells 2024; 13:1608. [PMID: 39404372 PMCID: PMC11475322 DOI: 10.3390/cells13191608] [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: 09/03/2024] [Revised: 09/22/2024] [Accepted: 09/23/2024] [Indexed: 10/19/2024] Open
Abstract
Accumulating evidence underscores exercise as a straightforward and cost-effective lifestyle intervention capable of mitigating the risk and slowing the emergence and progression of Alzheimer's disease (AD). However, the intricate cellular and molecular mechanisms mediating these exercise-induced benefits in AD remain elusive. The present study delved into the impact of treadmill exercise on memory retrieval performance, hippocampal synaptic plasticity, synaptic morphology, and the expression and activity of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic receptors (AMPARs) in 6-month-old APP/PS1 mice. APP/PS1 mice (4-month-old males) were randomly assigned to either a treadmill exercise group or a sedentary group, with C57BL/6J mice (4-month-old males) as the control group (both exercise and sedentary). The exercise regimen spanned 8 weeks. Our findings revealed that 8-week treadmill exercise reversed memory retrieval impairment in step-down fear conditioning in 6-month-old APP/PS1 mice. Additionally, treadmill exercise enhanced basic synaptic strength, short-term potentiation (STP), and long-term potentiation (LTP) of the hippocampus in these mice. Moreover, treadmill exercise correlated with an augmentation in synapse numbers, refinement of synaptic structures, and heightened expression and activity of AMPARs. Our findings suggest that treadmill exercise improves behavioral performance and facilitates synaptic transmission by increasing structural synaptic plasticity and the activity of AMPARs in the hippocampus of 6-month-old APP/PS1 mice, which is involved in pre- and postsynaptic processes.
Collapse
Affiliation(s)
- Laikang Yu
- Department of Strength and Conditioning Assessment and Monitoring, Beijing Sport University, Beijing 100084, China;
- Beijing Key Laboratory of Sports Performance and Skill Assessment, Beijing Sport University, Beijing 100084, China; (Y.L.); (Y.L.); (B.G.); (J.C.)
| | - Yan Li
- Beijing Key Laboratory of Sports Performance and Skill Assessment, Beijing Sport University, Beijing 100084, China; (Y.L.); (Y.L.); (B.G.); (J.C.)
| | - Yuanyuan Lv
- Beijing Key Laboratory of Sports Performance and Skill Assessment, Beijing Sport University, Beijing 100084, China; (Y.L.); (Y.L.); (B.G.); (J.C.)
- China Institute of Sport and Health Science, Beijing Sport University, Beijing 100084, China
| | - Boya Gu
- Beijing Key Laboratory of Sports Performance and Skill Assessment, Beijing Sport University, Beijing 100084, China; (Y.L.); (Y.L.); (B.G.); (J.C.)
| | - Jiajia Cai
- Beijing Key Laboratory of Sports Performance and Skill Assessment, Beijing Sport University, Beijing 100084, China; (Y.L.); (Y.L.); (B.G.); (J.C.)
| | - Qing-Song Liu
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI 53226, USA;
| | - Li Zhao
- Beijing Key Laboratory of Sports Performance and Skill Assessment, Beijing Sport University, Beijing 100084, China; (Y.L.); (Y.L.); (B.G.); (J.C.)
| |
Collapse
|
38
|
Wu Z, Zhao Z. Heterogating Gel Iontronics: A Revolution in Biointerfaces and Ion Signal Transmission. Gels 2024; 10:594. [PMID: 39330196 PMCID: PMC11431666 DOI: 10.3390/gels10090594] [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: 08/26/2024] [Revised: 09/13/2024] [Accepted: 09/13/2024] [Indexed: 09/28/2024] Open
Abstract
Currently, existing iontronic systems are limited and struggle to process electronic-to-multi-ionic transport, resulting in interchange inefficiencies and incompatibilities between artificial ion devices and biological tissue interfaces. The development of heterogating gel iontronics offers a significant advancement in bridging this gap, drawing inspiration from the complex ionic transmission mechanisms found in biological synapses within neural networks. These heterogating gels utilize a biphasic architecture, where the heterointerface effect constructs ionic transfer energy barriers, enabling distinct signal transmission among different ions. In systems with multiple ion species, heterogating gel iontronics allow for precise control of ion transmission, realizing hierarchical and selective cross-stage signal transmission as a neuromorphic function. This perspective highlights the vast potential of heterogating iontronics in applications such as biosensing, neuroprosthetics, and ion separation technologies. Meanwhile, it also addresses the current challenges, including scaling production, ensuring biocompatibility, and integrating with existing technologies, which are crucial for future development. The advancement of heterogating gels is expected to promote the integration between abiotic and biotic systems, with broad implications for smart sensors, bioneural devices, and beyond.
Collapse
Affiliation(s)
| | - Ziguang Zhao
- School of Future Technology, University of Chinese Academy of Sciences, Beijing 100190, China;
| |
Collapse
|
39
|
Chattaraj A, Baltaci Z, Chung S, Mayer BJ, Loew LM, Ditlev JA. Measurement of solubility product reveals the interplay of oligomerization and self-association for defining condensate formation. Mol Biol Cell 2024; 35:ar122. [PMID: 39046778 PMCID: PMC11449392 DOI: 10.1091/mbc.e24-01-0030] [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: 01/26/2024] [Revised: 06/06/2024] [Accepted: 07/16/2024] [Indexed: 07/25/2024] Open
Abstract
Cellular condensates often consist of 10s to 100s of distinct interacting molecular species. Because of the complexity of these interactions, predicting the point at which they will undergo phase separation is daunting. Using experiments and computation, we therefore studied a simple model system consisting of polySH3 and polyPRM designed for pentavalent heterotypic binding. We tested whether the peak solubility product, or the product of the dilute phase concentration of each component, is a predictive parameter for the onset of phase separation. Titrating up equal total concentrations of each component showed that the maximum solubility product does approximately coincide with the threshold for phase separation in both experiments and models. However, we found that measurements of dilute phase concentration include small oligomers and monomers; therefore, a quantitative comparison of the experiments and models required inclusion of small oligomers in the model analysis. Even with the inclusion of small polyPRM and polySH3 oligomers, models did not predict experimental results. This led us to perform dynamic light scattering experiments, which revealed homotypic binding of polyPRM. Addition of this interaction to our model recapitulated the experimentally observed asymmetry. Thus, comparing experiments with simulation reveals that the solubility product can be predictive of the interactions underlying phase separation, even if small oligomers and low affinity homotypic interactions complicate the analysis.
Collapse
Affiliation(s)
- Aniruddha Chattaraj
- R. D. Berlin Center for Cell Analysis and Modeling, University of Connecticut School of Medicine, Farmington, CT 06030
| | - Zeynep Baltaci
- Program in Molecular Medicine, Toronto, ON M5G 1E8, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Steve Chung
- Program in Molecular Medicine, Toronto, ON M5G 1E8, Canada
| | - Bruce J. Mayer
- R. D. Berlin Center for Cell Analysis and Modeling, University of Connecticut School of Medicine, Farmington, CT 06030
- Genetics and Genome Sciences, University of Connecticut School of Medicine, Farmington, CT 06030
| | - Leslie M. Loew
- R. D. Berlin Center for Cell Analysis and Modeling, University of Connecticut School of Medicine, Farmington, CT 06030
| | - Jonathon A. Ditlev
- Program in Molecular Medicine, Toronto, ON M5G 1E8, Canada
- Program in Cell Biology, Hospital for Sick Children, Toronto, ON M5G 1E8, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A1, Canada
| |
Collapse
|
40
|
Ruan K, Bai G, Fang Y, Li D, Li T, Liu X, Lu B, Lu Q, Songyang Z, Sun S, Wang Z, Zhang X, Zhou W, Zhang H. Biomolecular condensates and disease pathogenesis. SCIENCE CHINA. LIFE SCIENCES 2024; 67:1792-1832. [PMID: 39037698 DOI: 10.1007/s11427-024-2661-3] [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: 05/01/2024] [Accepted: 06/21/2024] [Indexed: 07/23/2024]
Abstract
Biomolecular condensates or membraneless organelles (MLOs) formed by liquid-liquid phase separation (LLPS) divide intracellular spaces into discrete compartments for specific functions. Dysregulation of LLPS or aberrant phase transition that disturbs the formation or material states of MLOs is closely correlated with neurodegeneration, tumorigenesis, and many other pathological processes. Herein, we summarize the recent progress in development of methods to monitor phase separation and we discuss the biogenesis and function of MLOs formed through phase separation. We then present emerging proof-of-concept examples regarding the disruption of phase separation homeostasis in a diverse array of clinical conditions including neurodegenerative disorders, hearing loss, cancers, and immunological diseases. Finally, we describe the emerging discovery of chemical modulators of phase separation.
Collapse
Affiliation(s)
- Ke Ruan
- The First Affiliated Hospital & School of Life Sciences, Ministry of Education Key Laboratory for Membrane-less Organelles & Cellular Dynamics, Hefei National Research Center for Interdisciplinary Sciences at the Microscale, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.
| | - Ge Bai
- Nanhu Brain-computer Interface Institute, Hangzhou, 311100, China.
- Department of Neurology of Second Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| | - Yanshan Fang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 201210, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Dan Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, 200030, China.
| | - Tingting Li
- Department of Biomedical Informatics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China.
| | - Xingguo Liu
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
- Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 510000, China.
| | - Boxun Lu
- Neurology Department at Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, School of Life Sciences, Fudan University, Shanghai, 200433, China.
| | - Qing Lu
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, 200030, China.
| | - Zhou Songyang
- State Key Laboratory of Biocontrol, MOE Key Laboratory of Gene Function and Regulation and Guangzhou Key Laboratory of Healthy Aging Research, School of Life Sciences, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510275, China.
| | - Shuguo Sun
- Department of Human Anatomy, Histology and Embryology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Zheng Wang
- The Second Affiliated Hospital, School of Basic Medical Sciences, Institute of Biomedical Innovation, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China.
| | - Xin Zhang
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, China.
| | - Wen Zhou
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China.
| | - Hong Zhang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
41
|
Bagheri Y, Rouches M, Machta B, Veatch SL. Prewetting couples membrane and protein phase transitions to greatly enhance coexistence in models and cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.26.609758. [PMID: 39253471 PMCID: PMC11383005 DOI: 10.1101/2024.08.26.609758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
Both membranes and biopolymers can individually separate into coexisting liquid phases. Here we explore biopolymer prewetting at membranes, a phase transition that emerges when these two thermodynamic systems are coupled. In reconstitution, we couple short poly-L-Lysine and poly-L-Glutamic Acid polyelectrolytes to membranes of saturated lipids, unsaturated lipids, and cholesterol, and detect coexisting prewet and dry surface phases well outside of the region of coexistence for each individual system. Notability, polyelectrolyte prewetting is highly sensitive to membrane lipid composition, occurring at 10 fold lower polymer concentration in a membrane close to its phase transition compared to one without a phase transition. In cells, protein prewetting is achieved using an optogenetic tool that enables titration of condensing proteins and tethering to the plasma membrane inner leaflet. Here we show that protein prewetting occurs for conditions well outside those where proteins condense in the cytoplasm, and that the stability of prewet domains is sensitive to perturbations of plasma membrane composition and structure. Our work presents an example of how thermodynamic phase transitions can impact cellular structure outside their individual coexistence regions, suggesting new possible roles for phase-separation-prone systems in cell biology.
Collapse
Affiliation(s)
- Yousef Bagheri
- Program in Biophysics, University of Michigan, Ann Arbor, MI USA
| | - Mason Rouches
- Department of Molecular Biophysics & Biochemistry, Yale University, New Haven CT USA
| | | | - Sarah L. Veatch
- Program in Biophysics, University of Michigan, Ann Arbor, MI USA
| |
Collapse
|
42
|
Chen R, Shi X, Yao X, Gao T, Huang G, Ning D, Cao Z, Xu Y, Liang W, Tian SZ, Zhu Q, Fang L, Zheng M, Hu Y, Cui H, Chen W. Specific multivalent molecules boost CRISPR-mediated transcriptional activation. Nat Commun 2024; 15:7222. [PMID: 39174527 PMCID: PMC11341856 DOI: 10.1038/s41467-024-51694-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 08/15/2024] [Indexed: 08/24/2024] Open
Abstract
CRISPR/Cas-based transcriptional activators can be enhanced by intrinsically disordered regions (IDRs). However, the underlying mechanisms are still debatable. Here, we examine 12 well-known IDRs by fusing them to the dCas9-VP64 activator, of which only seven can augment activation, albeit independently of their phase separation capabilities. Moreover, modular domains (MDs), another class of multivalent molecules, though ineffective in enhancing dCas9-VP64 activity on their own, show substantial enhancement in transcriptional activation when combined with dCas9-VP64-IDR. By varying the number of gRNA binding sites and fusing dCas9-VP64 with different IDRs/MDs, we uncover that optimal, rather than maximal, cis-trans cooperativity enables the most robust activation. Finally, targeting promoter-enhancer pairs yields synergistic effects, which can be further amplified via enhancing chromatin interactions. Overall, our study develops a versatile platform for efficient gene activation and sheds important insights into CRIPSR-based transcriptional activators enhanced with multivalent molecules.
Collapse
Affiliation(s)
- Rui Chen
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
- Innovative Center for RNA Therapeutics (ICRT), School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
- Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen, China
| | - Xinyao Shi
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Xiangrui Yao
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Tong Gao
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Guangyu Huang
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Duo Ning
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Zemin Cao
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Youxin Xu
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Weizheng Liang
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
- Central Laboratory, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei, China
| | - Simon Zhongyuan Tian
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
- Innovative Center for RNA Therapeutics (ICRT), School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Qionghua Zhu
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
- Innovative Center for RNA Therapeutics (ICRT), School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
- Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen, China
| | - Liang Fang
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
- Innovative Center for RNA Therapeutics (ICRT), School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
- Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen, China
| | - Meizhen Zheng
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
- Innovative Center for RNA Therapeutics (ICRT), School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Yuhui Hu
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, China
- Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Huanhuan Cui
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China.
- Innovative Center for RNA Therapeutics (ICRT), School of Life Sciences, Southern University of Science and Technology, Shenzhen, China.
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China.
- Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen, China.
| | - Wei Chen
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China.
- Innovative Center for RNA Therapeutics (ICRT), School of Life Sciences, Southern University of Science and Technology, Shenzhen, China.
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China.
- Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen, China.
| |
Collapse
|
43
|
Agarwal A, Chandran A, Raza F, Ungureanu IM, Hilcenko C, Stott K, Bright NA, Morone N, Warren AJ, Lautenschläger J. VAMP2 regulates phase separation of α-synuclein. Nat Cell Biol 2024; 26:1296-1308. [PMID: 38951707 PMCID: PMC11322000 DOI: 10.1038/s41556-024-01451-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 05/30/2024] [Indexed: 07/03/2024]
Abstract
α-Synuclein (αSYN), a pivotal synaptic protein implicated in synucleinopathies such as Parkinson's disease and Lewy body dementia, undergoes protein phase separation. We reveal that vesicle-associated membrane protein 2 (VAMP2) orchestrates αSYN phase separation both in vitro and in cells. Electrostatic interactions, specifically mediated by VAMP2 via its juxtamembrane domain and the αSYN C-terminal region, drive phase separation. Condensate formation is specific for R-SNARE VAMP2 and dependent on αSYN lipid membrane binding. Our results delineate a regulatory mechanism for αSYN phase separation in cells. Furthermore, we show that αSYN condensates sequester vesicles and attract complexin-1 and -2, thus supporting a role in synaptic physiology and pathophysiology.
Collapse
Affiliation(s)
- Aishwarya Agarwal
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Aswathy Chandran
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Farheen Raza
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- Protein and Cellular Sciences, GSK, Stevenage, UK
| | - Irina-Maria Ungureanu
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- Department of Clinical Neurosciences, UK Dementia Research Institute, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Christine Hilcenko
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- Wellcome Trust-Medical Research Council Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK
- Department of Haematology, University of Cambridge, School of Clinical Medicine, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK
| | - Katherine Stott
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Nicholas A Bright
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | | | - Alan J Warren
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- Wellcome Trust-Medical Research Council Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK
- Department of Haematology, University of Cambridge, School of Clinical Medicine, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK
| | - Janin Lautenschläger
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK.
| |
Collapse
|
44
|
Kusumi A, Tsunoyama TA, Suzuki KGN, Fujiwara TK, Aladag A. Transient, nano-scale, liquid-like molecular assemblies coming of age. Curr Opin Cell Biol 2024; 89:102394. [PMID: 38963953 DOI: 10.1016/j.ceb.2024.102394] [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: 02/08/2024] [Revised: 06/02/2024] [Accepted: 06/06/2024] [Indexed: 07/06/2024]
Abstract
This review examines the dynamic mechanisms underlying cellular signaling, communication, and adhesion via transient, nano-scale, liquid-like molecular assemblies on the plasma membrane (PM). Traditional views posit that stable, solid-like molecular complexes perform these functions. However, advanced imaging reveals that many signaling and scaffolding proteins only briefly reside in these molecular complexes and that micron-scale protein assemblies on the PM, including cell adhesion structures and synapses, are likely made of archipelagoes of nanoliquid protein islands. Borrowing the concept of liquid-liquid phase separation to form micron-scale biocondensates, we propose that these nano-scale oligomers and assemblies are enabled by multiple weak but specific molecular interactions often involving intrinsically disordered regions. The signals from individual nanoliquid signaling complexes would occur as pulses. Single-molecule imaging emerges as a crucial technique for characterizing these transient nanoliquid assemblies on the PM, suggesting a shift toward a model where the fluidity of interactions underpins signal regulation and integration.
Collapse
Affiliation(s)
- Akihiro Kusumi
- Membrane Cooperativity Unit, Okinawa Institute of Science and Technology Graduate University, Onna-son, Okinawa 904-0495, Japan; Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto 606-8501, Japan.
| | - Taka A Tsunoyama
- Membrane Cooperativity Unit, Okinawa Institute of Science and Technology Graduate University, Onna-son, Okinawa 904-0495, Japan
| | - Kenichi G N Suzuki
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto 606-8501, Japan; Institute for Glyco-core Research (iGCORE), Gifu University, Gifu 501-1193, Japan; National Cancer Center Research Institute, Tokyo 104-0045, Japan
| | - Takahiro K Fujiwara
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto 606-8501, Japan
| | - Amine Aladag
- Membrane Cooperativity Unit, Okinawa Institute of Science and Technology Graduate University, Onna-son, Okinawa 904-0495, Japan; Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto 606-8501, Japan
| |
Collapse
|
45
|
Nowacka A, Getz AM, Bessa-Neto D, Choquet D. Activity-dependent diffusion trapping of AMPA receptors as a key step for expression of early LTP. Philos Trans R Soc Lond B Biol Sci 2024; 379:20230220. [PMID: 38853553 PMCID: PMC11343219 DOI: 10.1098/rstb.2023.0220] [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: 02/24/2024] [Revised: 04/10/2024] [Accepted: 04/11/2024] [Indexed: 06/11/2024] Open
Abstract
This review focuses on the activity-dependent diffusion trapping of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs) as a crucial mechanism for the expression of early long-term potentiation (LTP), a process central to learning and memory. Despite decades of research, the precise mechanisms by which LTP induction leads to an increase in AMPAR responses at synapses have been elusive. We review the different hypotheses that have been put forward to explain the increased AMPAR responsiveness during LTP. We discuss the dynamic nature of AMPAR complexes, including their constant turnover and activity-dependent modifications that affect their synaptic accumulation. We highlight a hypothesis suggesting that AMPARs are diffusively trapped at synapses through activity-dependent interactions with protein-based binding slots in the post-synaptic density (PSD), offering a potential explanation for the increased synaptic strength during LTP. Furthermore, we outline the challenges still to be addressed before we fully understand the functional roles and molecular mechanisms of AMPAR dynamic nanoscale organization in LTP. This article is part of a discussion meeting issue 'Long-term potentiation: 50 years on'.
Collapse
Affiliation(s)
- Agata Nowacka
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, BordeauxF-33000, France
| | - Angela M. Getz
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, BordeauxF-33000, France
- University of Bordeaux, CNRS, INSERM, Bordeaux Imaging Center, BIC, UMS 3420, US 4, BordeauxF-33000, France
| | - Diogo Bessa-Neto
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, BordeauxF-33000, France
| | - Daniel Choquet
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, BordeauxF-33000, France
- University of Bordeaux, CNRS, INSERM, Bordeaux Imaging Center, BIC, UMS 3420, US 4, BordeauxF-33000, France
| |
Collapse
|
46
|
Yang C, Huang YT, Yao YF, Fu JY, Long YS. Hippocampal proteome comparison of infant and adult Fmr1 deficiency mice reveals adult-related changes associated with postsynaptic density. J Proteomics 2024; 303:105202. [PMID: 38797434 DOI: 10.1016/j.jprot.2024.105202] [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: 05/15/2024] [Accepted: 05/17/2024] [Indexed: 05/29/2024]
Abstract
Deficiency in fragile X mental retardation 1 (Fmr1) leads to loss of its encoded protein FMRP and causes fragile X syndrome (FXS) by dysregulating its target gene expression in an age-related fashion. Using comparative proteomic analysis, this study identified 105 differentially expressed proteins (DEPs) in the hippocampus of postnatal day 7 (P7) Fmr1-/y mice and 306 DEPs of P90 Fmr1-/y mice. We found that most DEPs in P90 hippocampus were not changed in P7 hippocampus upon FMRP absence, and some P90 DEPs exhibited diverse proteophenotypes with abnormal expression of protein isoform or allele variants. Bioinformatic analyses showed that the P7 DEPs were mainly enriched in fatty acid metabolism and oxidoreductase activity and nutrient responses; whereas the P90 PEPs (especially down-regulated DEPs) were primarily enriched in postsynaptic density (PSD), neuronal projection development and synaptic plasticity. Interestingly, 25 of 30 down-regulated PSD proteins present in the most enriched protein to protein interaction network, and 6 of them (ANK3, ATP2B2, DST, GRIN1, SHANK2 and SYNGAP1) are both FMRP targets and autism candidates. Therefore, this study suggests age-dependent alterations in hippocampal proteomes upon loss of FMRP that may be associated with the pathogenesis of FXS and its related disorders. SIGNIFICANCE: It is well known that loss of FMRP resulted from Fmr1 deficiency leads to fragile X syndrome (FXS), a common neurodevelopmental disorder accompanied by intellectual disability and autism spectrum disorder (ASD). FMRP exhibits distinctly spatiotemporal patterns in the hippocampus between early development and adulthood, which lead to distinct dysregulations of gene expression upon loss of FMRP at the two age stages potentially linked to age-related phenotypes. Therefore, comparison of hippocampal proteomes between infancy and adulthood is valuable to provide insights into the early causations and adult-dependent consequences for FXS and ASD. Using a comparative proteomic analysis, this study identified 105 and 306 differentially expressed proteins (DEPs) in the hippocampi of postnatal day 7 (P7) and P90 Fmr1-/y mice, respectively. Few overlapping DEPs were identified between P7 and P90 stages, and the P7 DEPs were mainly enriched in the regulation of fatty acid metabolism and oxidoreduction, whereas the P90 DEPs were preferentially enriched in the regulation of synaptic formation and plasticity. Particularly, the up-regulated P90 proteins are primarily involved in immune responses and neurodegeneration, and the down-regulated P90 proteins are associated with postsynaptic density, neuron projection and synaptic plasticity. Our findings suggest that distinctly changed proteins in FMRP-absence hippocampus between infancy and adulthood may contribute to age-dependent pathogenesis of FXS and ASD.
Collapse
Affiliation(s)
- Cui Yang
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Yu-Ting Huang
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Yi-Fei Yao
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Jun-Yi Fu
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China.
| | - Yue-Sheng Long
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China.
| |
Collapse
|
47
|
Miski M, Weber Á, Fekete-Molnár K, Keömley-Horváth BM, Csikász-Nagy A, Gáspári Z. Simulated complexes formed from a set of postsynaptic proteins suggest a localised effect of a hypomorphic Shank mutation. BMC Neurosci 2024; 25:32. [PMID: 38971749 PMCID: PMC11227168 DOI: 10.1186/s12868-024-00880-1] [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: 02/09/2024] [Accepted: 06/28/2024] [Indexed: 07/08/2024] Open
Abstract
BACKGROUND The postsynaptic density is an elaborate protein network beneath the postsynaptic membrane involved in the molecular processes underlying learning and memory. The postsynaptic density is built up from the same major proteins but its exact composition and organization differs between synapses. Mutations perturbing protein: protein interactions generally occurring in this network might lead to effects specific for cell types or processes, the understanding of which can be especially challenging. RESULTS In this work we use systems biology-based modeling of protein complex distributions in a simplified set of major postsynaptic proteins to investigate the effect of a hypomorphic Shank mutation perturbing a single well-defined interaction. We use data sets with widely variable abundances of the constituent proteins. Our results suggest that the effect of the mutation is heavily dependent on the overall availability of all the protein components of the whole network and no trivial correspondence between the expression level of the directly affected proteins and overall complex distribution can be observed. CONCLUSIONS Our results stress the importance of context-dependent interpretation of mutations. Even the weakening of a generally occurring protein: protein interaction might have well-defined effects, and these can not easily be predicted based only on the abundance of the proteins directly affected. Our results provide insight on how cell-specific effects can be exerted by a mutation perturbing a generally occurring interaction even when the wider interaction network is largely similar.
Collapse
Affiliation(s)
- Marcell Miski
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary
| | - Áron Weber
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary
- Cytocast Hungary Kft, Budapest, Hungary
| | - Krisztina Fekete-Molnár
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary
| | - Bence Márk Keömley-Horváth
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary
- Cytocast Hungary Kft, Budapest, Hungary
| | - Attila Csikász-Nagy
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary.
- Cytocast Hungary Kft, Budapest, Hungary.
| | - Zoltán Gáspári
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary.
| |
Collapse
|
48
|
Chuang HY, He RY, Huang YA, Hsu WT, Cheng YJ, Guo ZR, Wali N, Hwang IS, Shie JJ, Huang JJT. Engineered droplet-forming peptide as photocontrollable phase modulator for fused in sarcoma protein. Nat Commun 2024; 15:5686. [PMID: 38971830 PMCID: PMC11227587 DOI: 10.1038/s41467-024-50025-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 06/27/2024] [Indexed: 07/08/2024] Open
Abstract
The assembly and disassembly of biomolecular condensates are crucial for the subcellular compartmentalization of biomolecules in the control of cellular reactions. Recently, a correlation has been discovered between the phase transition of condensates and their maturation (aggregation) process in diseases. Therefore, modulating the phase of condensates to unravel the roles of condensation has become a matter of interest. Here, we create a peptide-based phase modulator, JSF1, which forms droplets in the dark and transforms into amyloid-like fibrils upon photoinitiation, as evidenced by their distinctive nanomechanical and dynamic properties. JSF1 is found to effectively enhance the condensation of purified fused in sarcoma (FUS) protein and, upon light exposure, induce its fibrilization. We also use JSF1 to modulate the biophysical states of FUS condensates in live cells and elucidate the relationship between FUS phase transition and FUS proteinopathy, thereby shedding light on the effect of protein phase transition on cellular function and malfunction.
Collapse
Affiliation(s)
- Hao-Yu Chuang
- Institute of Chemistry, Academia Sinica, Taipei, 115, Taiwan
- Chemical Biology and Molecular Biophysics, Taiwan International Graduate Program, Academia Sinica, Taipei, 115, Taiwan
- Department of Chemistry, National Tsing Hua University, Hsinchu, 300, Taiwan
| | - Ruei-Yu He
- Institute of Chemistry, Academia Sinica, Taipei, 115, Taiwan
| | - Yung-An Huang
- Institute of Chemistry, Academia Sinica, Taipei, 115, Taiwan
| | - Wan-Ting Hsu
- Institute of Chemistry, Academia Sinica, Taipei, 115, Taiwan
| | - Ya-Jen Cheng
- Neuroscience Program of Academia Sinica, Academia Sinica, Taipei, 115, Taiwan
- Institute of Molecular Biology, Academia Sinica, Taipei, 115, Taiwan
| | - Zheng-Rong Guo
- Institute of Physics, Academia Sinica, Taipei, 115, Taiwan
| | - Niaz Wali
- Institute of Chemistry, Academia Sinica, Taipei, 115, Taiwan
| | | | - Jiun-Jie Shie
- Institute of Chemistry, Academia Sinica, Taipei, 115, Taiwan
| | - Joseph Jen-Tse Huang
- Institute of Chemistry, Academia Sinica, Taipei, 115, Taiwan.
- Neuroscience Program of Academia Sinica, Academia Sinica, Taipei, 115, Taiwan.
- Sustainable Chemical Science and Technology, Taiwan International Graduate Program, Academia Sinica, Taipei, 115, Taiwan.
- Department of Applied Chemistry, National Chiayi University, Chiayi City, 600, Taiwan.
| |
Collapse
|
49
|
Lee CT, Bell M, Bonilla-Quintana M, Rangamani P. Biophysical Modeling of Synaptic Plasticity. Annu Rev Biophys 2024; 53:397-426. [PMID: 38382115 DOI: 10.1146/annurev-biophys-072123-124954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2024]
Abstract
Dendritic spines are small, bulbous compartments that function as postsynaptic sites and undergo intense biochemical and biophysical activity. The role of the myriad signaling pathways that are implicated in synaptic plasticity is well studied. A recent abundance of quantitative experimental data has made the events associated with synaptic plasticity amenable to quantitative biophysical modeling. Spines are also fascinating biophysical computational units because spine geometry, signal transduction, and mechanics work in a complex feedback loop to tune synaptic plasticity. In this sense, ideas from modeling cell motility can inspire us to develop multiscale approaches for predictive modeling of synaptic plasticity. In this article, we review the key steps in postsynaptic plasticity with a specific focus on the impact of spine geometry on signaling, cytoskeleton rearrangement, and membrane mechanics. We summarize the main experimental observations and highlight how theory and computation can aid our understanding of these complex processes.
Collapse
Affiliation(s)
- Christopher T Lee
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, California, USA;
| | - Miriam Bell
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, California, USA;
| | - Mayte Bonilla-Quintana
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, California, USA;
| | - Padmini Rangamani
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, California, USA;
| |
Collapse
|
50
|
Carricaburu E, Benner O, Burlingham SR, Dos Santos Passos C, Hobaugh N, Karr CH, Chanda S. Gephyrin promotes autonomous assembly and synaptic localization of GABAergic postsynaptic components without presynaptic GABA release. Proc Natl Acad Sci U S A 2024; 121:e2315100121. [PMID: 38889143 PMCID: PMC11214061 DOI: 10.1073/pnas.2315100121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 05/17/2024] [Indexed: 06/20/2024] Open
Abstract
Synapses containing γ-aminobutyric acid (GABA) constitute the primary centers for inhibitory neurotransmission in our nervous system. It is unclear how these synaptic structures form and align their postsynaptic machineries with presynaptic terminals. Here, we monitored the cellular distribution of several GABAergic postsynaptic proteins in a purely glutamatergic neuronal culture derived from human stem cells, which virtually lacks any vesicular GABA release. We found that several GABAA receptor (GABAAR) subunits, postsynaptic scaffolds, and major cell-adhesion molecules can reliably coaggregate and colocalize at even GABA-deficient subsynaptic domains, but remain physically segregated from glutamatergic counterparts. Genetic deletions of both Gephyrin and a Gephyrin-associated guanosine di- or triphosphate (GDP/GTP) exchange factor Collybistin severely disrupted the coassembly of these postsynaptic compositions and their proper apposition with presynaptic inputs. Gephyrin-GABAAR clusters, developed in the absence of GABA transmission, could be subsequently activated and even potentiated by delayed supply of vesicular GABA. Thus, molecular organization of GABAergic postsynapses can initiate via a GABA-independent but Gephyrin-dependent intrinsic mechanism.
Collapse
Affiliation(s)
- Etta Carricaburu
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO80523
| | - Orion Benner
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO80523
| | - Scott R. Burlingham
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO80523
| | | | - Natalia Hobaugh
- Biological Sciences Division, University of Chicago, Chicago, IL60637
| | - Charles H. Karr
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO80523
| | - Soham Chanda
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO80523
- Molecular, Cellular and Integrated Neurosciences Program, Colorado State University, Fort Collins, CO80523
- Cell and Molecular Biology Program, Colorado State University, Fort Collins, CO80523
| |
Collapse
|