1
|
Song X, Baltanás-Copado J, Selvaraj M, Kokate SB, Kumpula EP, Corbalán-García S, Huiskonen JT. The mechanism underlying fascin-mediated bundling of actin filaments unveiled by cryo-electron tomography. J Struct Biol 2025; 217:108212. [PMID: 40403900 DOI: 10.1016/j.jsb.2025.108212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 03/23/2025] [Accepted: 05/19/2025] [Indexed: 05/24/2025]
Abstract
Fascins are crucial actin-binding proteins linked to carcinomas, such as cancer metastasis. Fascins crosslink unipolar actin filaments into linear and rigid parallel bundles, which play essential roles in the formation of filopodia, stereocilia and other membrane protrusions. However, the mechanism of how fascin bundles actin filaments has remained elusive. Here, we studied the organization of reconstituted fascin-actin bundles by cryo-electron tomography and determined the structure of the fascin-actin complex at 9 Å resolution by subtomogram averaging. Consistent with earlier findings, fascin molecules decorate adjacent actin filaments, positioned at regular intervals corresponding to the half-pitch of actin filaments. The fascin-actin complex structure allows us to verify the binding orientation of fascin between the two actin filaments. Fitting of the previously solved fascin crystal structure facilitates the analysis of the interaction surfaces. Our structural models serve as a blueprint to understand the detailed interactions between fascin and actins and provide new insights for the development of drugs targeting fascin proteins.
Collapse
Affiliation(s)
- Xiyong Song
- Institute of Biotechnology, Helsinki Institute of Life Science HiLIFE, University of Helsinki, Helsinki 00014, Finland
| | - Jesús Baltanás-Copado
- Departamento de Bioquímica y Biología Molecular-A, Facultad de Veterinaria, Regional Campus of International Excellence "Campus Mare Nostrum", Universidad de Murcia, Instituto Murciano de Investigación Biosanitaria, Murcia 30100, Spain
| | - Muniyandi Selvaraj
- Institute of Biotechnology, Helsinki Institute of Life Science HiLIFE, University of Helsinki, Helsinki 00014, Finland
| | - Shrikant B Kokate
- Institute of Biotechnology, Helsinki Institute of Life Science HiLIFE, University of Helsinki, Helsinki 00014, Finland
| | - Esa-Pekka Kumpula
- Institute of Biotechnology, Helsinki Institute of Life Science HiLIFE, University of Helsinki, Helsinki 00014, Finland
| | - Senena Corbalán-García
- Departamento de Bioquímica y Biología Molecular-A, Facultad de Veterinaria, Regional Campus of International Excellence "Campus Mare Nostrum", Universidad de Murcia, Instituto Murciano de Investigación Biosanitaria, Murcia 30100, Spain.
| | - Juha T Huiskonen
- Institute of Biotechnology, Helsinki Institute of Life Science HiLIFE, University of Helsinki, Helsinki 00014, Finland.
| |
Collapse
|
2
|
Heydari S, Liu J. High-throughput cryo-electron tomography enables multiscale visualization of the inner life of microbes. Curr Opin Struct Biol 2025; 93:103065. [PMID: 40381356 DOI: 10.1016/j.sbi.2025.103065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Revised: 04/21/2025] [Accepted: 04/22/2025] [Indexed: 05/20/2025]
Abstract
Cryo-electron tomography (cryo-ET) is an advanced and rapidly evolving imaging technique that enables three-dimensional visualization of biological structures in their native state. Although cryo-ET has historically faced significant challenges, including limited applications, tedious data acquisition, labor-intensive image processing, and lower resolution when compared with single particle cryo-electron microscopy (cryo-EM), recent breakthroughs in hardware and software development have significantly improved the entire cryo-ET workflow to enable higher throughput and resolution. These advances have accelerated discoveries in structural and cellular biology, particularly in microbiology, where cryo-ET has unveiled unprecedented insights into the inner life of microbes. This review presents pivotal advances propelling high-throughput cryo-ET and the visualization of microbial architecture. As innovations in imaging technologies, workflow automation, and computational methods continue progressing rapidly, cryo-ET is expected to be increasingly utilized across various fields of life sciences, shaping the future of biological research and biomedical discoveries.
Collapse
Affiliation(s)
- Samira Heydari
- Microbial Sciences Institute, Yale University, West Haven, CT 06516, USA; Department of Microbial Pathogenesis, Yale School of Medicine, New Haven, CT 06536, USA
| | - Jun Liu
- Microbial Sciences Institute, Yale University, West Haven, CT 06516, USA; Department of Microbial Pathogenesis, Yale School of Medicine, New Haven, CT 06536, USA.
| |
Collapse
|
3
|
Zhan X, Zeng X, Uddin MR, Xu M. AITom: AI-guided cryo-electron tomography image analyses toolkit. J Struct Biol 2025; 217:108207. [PMID: 40378936 DOI: 10.1016/j.jsb.2025.108207] [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: 12/17/2024] [Revised: 04/20/2025] [Accepted: 04/28/2025] [Indexed: 05/19/2025]
Abstract
Cryo-electron tomography (cryo-ET) is an essential tool in structural biology, uniquely capable of visualizing three-dimensional macromolecular complexes within their native cellular environments, thereby providing profound molecular-level insights. Despite its significant promise, cryo-ET faces persistent challenges in the systematic localization, identification, segmentation, and structural recovery of three-dimensional subcellular components, necessitating the development of efficient and accurate large-scale image analysis methods. In response to these complexities, this paper introduces AITom, an open-source artificial intelligence platform tailored for cryo-ET researchers. AITom integrates a comprehensive suite of public and proprietary algorithms, supporting both traditional template-based and template-free approaches, alongside state-of-the-art deep learning methodologies for cryo-ET data analysis. By incorporating diverse computational strategies, AITom enables researchers to more effectively tackle the complexities inherent in cryo-ET, facilitating precise analysis and interpretation of complex biological structures. Furthermore, AITom provides extensive tutorials for each analysis module, offering valuable guidance to users in utilizing its comprehensive functionalities.
Collapse
Affiliation(s)
- Xueying Zhan
- Computational Biology Department, Carnegie Mellon University, Pittsburgh, PA, United States
| | - Xiangrui Zeng
- Computational Biology Department, Carnegie Mellon University, Pittsburgh, PA, United States
| | - Mostofa Rafid Uddin
- Computational Biology Department, Carnegie Mellon University, Pittsburgh, PA, United States
| | - Min Xu
- Computational Biology Department, Carnegie Mellon University, Pittsburgh, PA, United States.
| |
Collapse
|
4
|
Zhou H, Hutchings J, Shiozaki M, Zhao X, Doolittle LK, Yang S, Yan R, Jean N, Riggi M, Yu Z, Villa E, Rosen MK. Quantitative spatial analysis of chromatin biomolecular condensates using cryoelectron tomography. Proc Natl Acad Sci U S A 2025; 122:e2426449122. [PMID: 40327693 PMCID: PMC12088439 DOI: 10.1073/pnas.2426449122] [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: 12/17/2024] [Accepted: 03/31/2025] [Indexed: 05/08/2025] Open
Abstract
Phase separation is an important mechanism to generate certain biomolecular condensates and organize the cell interior. Condensate formation and function remain incompletely understood due to difficulties in visualizing the condensate interior at high resolution. Here, we analyzed the structure of biochemically reconstituted chromatin condensates through cryoelectron tomography. We found that traditional blotting methods of sample preparation were inadequate, and high-pressure freezing plus focused ion beam milling was essential to maintain condensate integrity. To identify densely packed molecules within the condensate, we integrated deep learning-based segmentation with context-aware template matching. Our approaches were developed on chromatin condensates and were also effective on condensed regions of in situ native chromatin. Using these methods, we determined the average structure of nucleosomes to 6.1 and 12 Å resolution in reconstituted and native systems, respectively, found that nucleosomes form heterogeneous interaction networks in both cases, and gained insight into the molecular origins of surface tension in chromatin condensates. Our methods should be applicable to biomolecular condensates containing large and distinctive components in both biochemical reconstitutions and certain cellular systems.
Collapse
Affiliation(s)
- Huabin Zhou
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX75390
- HHMI, University of Texas Southwestern Medical Center, Dallas, TX75390
| | - Joshua Hutchings
- School of Biological Sciences, University of California, San Diego, La Jolla, CA92093
| | | | | | - Lynda K. Doolittle
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX75390
- HHMI, University of Texas Southwestern Medical Center, Dallas, TX75390
| | - Shixin Yang
- Janelia Research Campus, HHMI, Ashburn, VA20147
| | - Rui Yan
- Janelia Research Campus, HHMI, Ashburn, VA20147
| | - Nikki Jean
- Janelia Research Campus, HHMI, Ashburn, VA20147
| | - Margot Riggi
- Research Department Cell and Virus Structure, Max Planck Institute for Biochemistry, Martinsried/MunichD-82152, Germany
| | - Zhiheng Yu
- Janelia Research Campus, HHMI, Ashburn, VA20147
| | - Elizabeth Villa
- School of Biological Sciences, University of California, San Diego, La Jolla, CA92093
- HHMI, University of California, San Diego, La Jolla, CA92093
| | - Michael K. Rosen
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX75390
- HHMI, University of Texas Southwestern Medical Center, Dallas, TX75390
| |
Collapse
|
5
|
Khavnekar S, Wan W. An approach for coherent periodogram averaging of tilt-series data for improved contrast transfer function estimation. FEBS Open Bio 2025. [PMID: 40341839 DOI: 10.1002/2211-5463.70050] [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: 11/08/2024] [Revised: 03/21/2025] [Accepted: 04/23/2025] [Indexed: 05/11/2025] Open
Abstract
Cryo-electron microscopy (cryo-EM) has become an indispensable technique for determining three-dimensional structures of biological macromolecules. A critical aspect of achieving high-resolution cryo-EM reconstructions is accurately determining and correcting for the microscope's contrast transfer function (CTF). The CTF introduces defocus-dependent distortions during imaging; if not properly accounted for, the CTF can distort features in and limit the resolution of 3D reconstructions. For tilt-series data used in cryo-electron tomography (cryo-ET), CTF estimation becomes even more challenging due to the tilt of the specimen, which introduces a defocus gradient across the field of view, as well as the low dose and signal in individual tilt images. Here, we describe a simple algorithm to improve the accuracy of CTF estimation of tilted images by leveraging the tilt-series alignment parameters determined for tomographic reconstruction to explicitly account for the tilted specimen geometry. In brief, each tilt image is divided into patches, each of which are then stretched according to their defocus shift. These are then summed to provide a coherent power spectrum at the tilt axis, which can then be used in standard CTF estimation algorithms. This uses all the data in each image to enhance the visibility of Thon rings, thereby improving high-resolution CTF estimation and subsequent enhancements in the resolution of subtomogram averages.
Collapse
Affiliation(s)
| | - William Wan
- Department of Biochemistry and Center for Structural Biology, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
6
|
Kannan S, Altae-Tran H, Zhu S, Xu P, Strebinger D, Oshiro R, Faure G, Moeller L, Pham J, Mears KS, Ni HM, Macrae RK, Zhang F. Evolution-guided protein design of IscB for persistent epigenome editing in vivo. Nat Biotechnol 2025:10.1038/s41587-025-02655-3. [PMID: 40335752 DOI: 10.1038/s41587-025-02655-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 03/26/2025] [Indexed: 05/09/2025]
Abstract
Naturally existing enzymes have been adapted for a variety of molecular technologies, with enhancements or modifications to the enzymes introduced to improve the desired function; however, it is difficult to engineer variants with enhanced activity while maintaining specificity. Here we engineer the compact Obligate Mobile Element Guided Activity (OMEGA) RNA-guided endonuclease IscB and its guiding RNA (ωRNA) by combining ortholog screening, structure-guided protein domain design and RNA engineering, and deep learning-based structure prediction to generate an improved variant, NovaIscB. We show that the compact NovaIscB achieves up to 40% indel activity (~100-fold improvement over wild-type OgeuIscB) on the human genome with improved specificity relative to existing IscBs. We further show that NovaIscB can be fused with a methyltransferase to create a programmable transcriptional repressor, OMEGAoff, that is compact enough to be packaged in a single adeno-associated virus vector for persistent in vivo gene repression. This study highlights the power of combining natural diversity with protein engineering to design enhanced enzymes for molecular biology applications.
Collapse
Affiliation(s)
- Soumya Kannan
- Howard Hughes Medical Institute, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Yang Tan Collective, Cambridge, MA, USA
| | - Han Altae-Tran
- Howard Hughes Medical Institute, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Yang Tan Collective, Cambridge, MA, USA
| | - Shiyou Zhu
- Howard Hughes Medical Institute, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Yang Tan Collective, Cambridge, MA, USA
| | - Peiyu Xu
- Howard Hughes Medical Institute, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Yang Tan Collective, Cambridge, MA, USA
| | - Daniel Strebinger
- Howard Hughes Medical Institute, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Yang Tan Collective, Cambridge, MA, USA
| | - Rachel Oshiro
- Howard Hughes Medical Institute, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Yang Tan Collective, Cambridge, MA, USA
| | - Guilhem Faure
- Howard Hughes Medical Institute, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Yang Tan Collective, Cambridge, MA, USA
| | - Lukas Moeller
- Howard Hughes Medical Institute, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Yang Tan Collective, Cambridge, MA, USA
| | - Julie Pham
- Howard Hughes Medical Institute, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Yang Tan Collective, Cambridge, MA, USA
| | - Kepler S Mears
- Howard Hughes Medical Institute, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Yang Tan Collective, Cambridge, MA, USA
| | - Heyuan M Ni
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Rhiannon K Macrae
- Howard Hughes Medical Institute, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Yang Tan Collective, Cambridge, MA, USA
| | - Feng Zhang
- Howard Hughes Medical Institute, Cambridge, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Yang Tan Collective, Cambridge, MA, USA.
| |
Collapse
|
7
|
Soni K, Horvath A, Dybkov O, Schwan M, Trakansuebkul S, Flemming D, Wild K, Urlaub H, Fischer T, Sinning I. Structures of aberrant spliceosome intermediates on their way to disassembly. Nat Struct Mol Biol 2025; 32:914-925. [PMID: 39833470 PMCID: PMC12086092 DOI: 10.1038/s41594-024-01480-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 12/19/2024] [Indexed: 01/22/2025]
Abstract
Intron removal during pre-mRNA splicing is of extraordinary complexity and its disruption causes a vast number of genetic diseases in humans. While key steps of the canonical spliceosome cycle have been revealed by combined structure-function analyses, structural information on an aberrant spliceosome committed to premature disassembly is not available. Here, we report two cryo-electron microscopy structures of post-Bact spliceosome intermediates from Schizosaccharomyces pombe primed for disassembly. We identify the DEAH-box helicase-G-patch protein pair (Gih35-Gpl1, homologous to human DHX35-GPATCH1) and show how it maintains catalytic dormancy. In both structures, Gpl1 recognizes a remodeled active site introduced by an overstabilization of the U5 loop I interaction with the 5' exon leading to a single-nucleotide insertion at the 5' splice site. Remodeling is communicated to the spliceosome surface and the Ntr1 complex that mediates disassembly is recruited. Our data pave the way for a targeted analysis of splicing quality control.
Collapse
Affiliation(s)
- Komal Soni
- Heidelberg University Biochemistry Center (BZH), Heidelberg, Germany.
| | - Attila Horvath
- The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Olexandr Dybkov
- Bioanalytical Mass Spectrometry group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Merlin Schwan
- Heidelberg University Biochemistry Center (BZH), Heidelberg, Germany
| | - Sasanan Trakansuebkul
- The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Dirk Flemming
- Heidelberg University Biochemistry Center (BZH), Heidelberg, Germany
| | - Klemens Wild
- Heidelberg University Biochemistry Center (BZH), Heidelberg, Germany
| | - Henning Urlaub
- Bioanalytical Mass Spectrometry group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Bioanalytics, Institute for Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany
| | - Tamás Fischer
- The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia.
| | - Irmgard Sinning
- Heidelberg University Biochemistry Center (BZH), Heidelberg, Germany.
| |
Collapse
|
8
|
Gong R, Reynolds MJ, Carney KR, Hamilton K, Bidone TC, Alushin GM. Fascin structural plasticity mediates flexible actin bundle construction. Nat Struct Mol Biol 2025; 32:940-952. [PMID: 39833469 PMCID: PMC12086090 DOI: 10.1038/s41594-024-01477-2] [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: 03/27/2024] [Accepted: 12/17/2024] [Indexed: 01/22/2025]
Abstract
Fascin cross-links actin filaments (F-actin) into bundles that support tubular membrane protrusions including filopodia and stereocilia. Fascin dysregulation drives aberrant cell migration during metastasis, and fascin inhibitors are under development as cancer therapeutics. Here, we use cryo-EM, cryo-electron tomography coupled with custom denoising and computational modeling to probe human fascin-1's F-actin cross-linking mechanisms across spatial scales. Our fascin cross-bridge structure reveals an asymmetric F-actin binding conformation that is allosterically blocked by the inhibitor G2. Reconstructions of seven-filament hexagonal bundle elements, variability analysis and simulations show how structural plasticity enables fascin to bridge varied interfilament orientations, accommodating mismatches between F-actin's helical symmetry and bundle hexagonal packing. Tomography of many-filament bundles and modeling uncover geometric rules underlying emergent fascin binding patterns, as well as the accumulation of unfavorable cross-links that limit bundle size. Collectively, this work shows how fascin harnesses fine-tuned nanoscale structural dynamics to build and regulate micron-scale F-actin bundles.
Collapse
Affiliation(s)
- Rui Gong
- Laboratory of Structural Biophysics and Mechanobiology, The Rockefeller University, New York, NY, USA.
| | - Matthew J Reynolds
- Laboratory of Structural Biophysics and Mechanobiology, The Rockefeller University, New York, NY, USA
| | - Keith R Carney
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA
| | - Keith Hamilton
- Laboratory of Structural Biophysics and Mechanobiology, The Rockefeller University, New York, NY, USA
| | - Tamara C Bidone
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA
| | - Gregory M Alushin
- Laboratory of Structural Biophysics and Mechanobiology, The Rockefeller University, New York, NY, USA.
| |
Collapse
|
9
|
Ghanbarpour A, Telusma B, Powell BM, Zhang JJ, Bolstad I, Vargas C, Keller S, Baker TA, Sauer RT, Davis JH. An asymmetric nautilus-like HflK/C assembly controls FtsH proteolysis of membrane proteins. EMBO J 2025; 44:2501-2513. [PMID: 40082723 PMCID: PMC12048511 DOI: 10.1038/s44318-025-00408-1] [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: 08/28/2024] [Revised: 12/13/2024] [Accepted: 02/28/2025] [Indexed: 03/16/2025] Open
Abstract
The AAA protease FtsH associates with HflK/C subunits to form a megadalton-size complex that spans the inner membrane and extends into the periplasm of E. coli. How this bacterial complex and homologous assemblies in eukaryotic organelles recruit, extract, and degrade membrane-embedded substrates is unclear. Following the overproduction of protein components, recent cryo-EM structures showed symmetric HflK/C cages surrounding FtsH in a manner proposed to inhibit the degradation of membrane-embedded substrates. Here, we present structures of native protein complexes, in which HflK/C instead forms an asymmetric nautilus-shaped assembly with an entryway for membrane-embedded substrates to reach and be engaged by FtsH. Consistent with this nautilus-like structure, proteomic assays suggest that HflK/C enhances FtsH degradation of certain membrane-embedded substrates. Membrane curvature in our FtsH•HflK/C complexes is opposite that of surrounding membrane regions, a property that correlates with lipid scramblase activity and possibly with FtsH's function in the degradation of membrane-embedded proteins.
Collapse
Affiliation(s)
- Alireza Ghanbarpour
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02129, USA.
- Department of Biochemistry and Molecular Biophysics, Washington University in St. Louis, St. Louis, MO, 63110, USA.
| | - Bertina Telusma
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02129, USA
| | - Barrett M Powell
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02129, USA
| | - Jia Jia Zhang
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02129, USA
| | - Isabella Bolstad
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02129, USA
| | - Carolyn Vargas
- Biophysics, Institute of Molecular Biosciences (IMB), NAWI Graz, University of Graz, Humboldtstr. 50/III, Graz, 8010, Austria
- Field of Excellence BioHealth, University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| | - Sandro Keller
- Biophysics, Institute of Molecular Biosciences (IMB), NAWI Graz, University of Graz, Humboldtstr. 50/III, Graz, 8010, Austria
- Field of Excellence BioHealth, University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| | - Tania A Baker
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02129, USA
| | - Robert T Sauer
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02129, USA.
| | - Joseph H Davis
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02129, USA.
- Program in Computational and Systems Biology, Massachusetts Institute of Technology, Cambridge, MA, 02129, USA.
| |
Collapse
|
10
|
Shao Q, Khawaja A, Nguyen MD, Singh V, Zhang J, Liu Y, Nordin J, Adori M, Axel Innis C, Castro Dopico X, Rorbach J. T cell toxicity induced by tigecycline binding to the mitochondrial ribosome. Nat Commun 2025; 16:4080. [PMID: 40312422 PMCID: PMC12045974 DOI: 10.1038/s41467-025-59388-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 04/21/2025] [Indexed: 05/03/2025] Open
Abstract
Tetracyclines are essential bacterial protein synthesis inhibitors under continual development to combat antibiotic resistance yet suffer from unwanted side effects. Mitoribosomes - responsible for generating oxidative phosphorylation (OXPHOS) subunits - share structural similarities with bacterial machinery and may suffer from cross-reactivity. Since lymphocytes rely upon OXPHOS upregulation to establish immunity, we set out to assess the impact of ribosome-targeting antibiotics on human T cells. We find tigecycline, a third-generation tetracycline, to be the most cytotoxic compound tested. In vitro, 5-10 μM tigecycline inhibits mitochondrial but not cytosolic translation, mitochondrial complex I, III and IV expression, and curtails the activation and expansion of unique T cell subsets. By cryo-EM, we find tigecycline to occupy three sites on T cell mitoribosomes. In addition to the conserved A-site found in bacteria, tigecycline also attaches to the peptidyl transferase center of the large subunit. Furthermore, a third, distinct binding site on the large subunit, aligns with helices analogous to those in bacteria, albeit lacking methylation in humans. The data provide a mechanism to explain part of the anti-inflammatory effects of these drugs and inform antibiotic design.
Collapse
Affiliation(s)
- Qiuya Shao
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Anas Khawaja
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Minh Duc Nguyen
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
- Faculty of Pharmacy, Phenikaa University, Ha Dong, Hanoi, Vietnam
| | - Vivek Singh
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Jingdian Zhang
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Yong Liu
- Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Joel Nordin
- Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Monika Adori
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - C Axel Innis
- ARNA Laboratory, Univ. Bordeaux, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Bordeaux, France
| | - Xaquin Castro Dopico
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden.
- Department of Animal and Veterinary Sciences, Aarhus Universitet, Tjele, Denmark.
| | - Joanna Rorbach
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
11
|
Chen X, Wang L, Xie J, Nowak JS, Luo B, Zhang C, Jia G, Zou J, Huang D, Glatt S, Yang Y, Su Z. RNA sample optimization for cryo-EM analysis. Nat Protoc 2025; 20:1114-1157. [PMID: 39548288 DOI: 10.1038/s41596-024-01072-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 09/12/2024] [Indexed: 11/17/2024]
Abstract
RNAs play critical roles in most biological processes. Although the three-dimensional (3D) structures of RNAs primarily determine their functions, it remains challenging to experimentally determine these 3D structures due to their conformational heterogeneity and intrinsic dynamics. Cryogenic electron microscopy (cryo-EM) has recently played an emerging role in resolving dynamic conformational changes and understanding structure-function relationships of RNAs including ribozymes, riboswitches and bacterial and viral noncoding RNAs. A variety of methods and pipelines have been developed to facilitate cryo-EM structure determination of challenging RNA targets with small molecular weights at subnanometer to near-atomic resolutions. While a wide range of conditions have been used to prepare RNAs for cryo-EM analysis, correlations between the variables in these conditions and cryo-EM visualizations and reconstructions remain underexplored, which continue to hinder optimizations of RNA samples for high-resolution cryo-EM structure determination. Here we present a protocol that describes rigorous screenings and iterative optimizations of RNA preparation conditions that facilitate cryo-EM structure determination, supplemented by cryo-EM data processing pipelines that resolve RNA dynamics and conformational changes and RNA modeling algorithms that generate atomic coordinates based on moderate- to high-resolution cryo-EM density maps. The current protocol is designed for users with basic skills and experience in RNA biochemistry, cryo-EM and RNA modeling. The expected time to carry out this protocol may range from 3 days to more than 3 weeks, depending on the many variables described in the protocol. For particularly challenging RNA targets, this protocol could also serve as a starting point for further optimizations.
Collapse
Affiliation(s)
- Xingyu Chen
- The State Key Laboratory of Biotherapy, Department of Geriatrics and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Liu Wang
- The State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, Department of Cardiology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jiahao Xie
- The State Key Laboratory of Biotherapy, Department of Geriatrics and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Jakub S Nowak
- Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
| | - Bingnan Luo
- The State Key Laboratory of Biotherapy, Department of Geriatrics and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
| | - Chong Zhang
- The State Key Laboratory of Biotherapy, Department of Geriatrics and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Guowen Jia
- The State Key Laboratory of Biotherapy, Department of Geriatrics and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Jian Zou
- The State Key Laboratory of Biotherapy, Department of Geriatrics and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Dingming Huang
- The State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, Department of Cardiology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Sebastian Glatt
- Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
- Department for Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Yang Yang
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zhaoming Su
- The State Key Laboratory of Biotherapy, Department of Geriatrics and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
12
|
Bang I, Hattori T, Leloup N, Corrado A, Nyamaa A, Koide A, Geles K, Buck E, Koide S. Selective targeting of oncogenic hotspot mutations of the HER2 extracellular domain. Nat Chem Biol 2025; 21:706-715. [PMID: 39438724 DOI: 10.1038/s41589-024-01751-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 09/13/2024] [Indexed: 10/25/2024]
Abstract
Oncogenic mutations in the extracellular domain (ECD) of cell-surface receptors could serve as tumor-specific antigens that are accessible to antibody therapeutics. Such mutations have been identified in receptor tyrosine kinases including HER2. However, it is challenging to selectively target a point mutant, while sparing the wild-type protein. Here we developed antibodies selective to HER2 S310F and S310Y, the two most common oncogenic mutations in the HER2 ECD, via combinatorial library screening and structure-guided design. Cryogenic-electron microscopy structures of the HER2 S310F homodimer and an antibody bound to HER2 S310F revealed that these antibodies recognize the mutations in a manner that mimics the dimerization arm of HER2 and thus inhibit HER2 dimerization. These antibodies as T cell engagers selectively killed a HER2 S310F-driven cancer cell line in vitro, and in vivo as a xenograft. These results validate HER2 ECD mutations as actionable therapeutic targets and offer promising candidates toward clinical development.
Collapse
Affiliation(s)
- Injin Bang
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA
| | - Takamitsu Hattori
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, USA
| | - Nadia Leloup
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA
| | - Alexis Corrado
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA
| | - Atekana Nyamaa
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA
| | - Akiko Koide
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA
- Division of Hematology Oncology, Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Ken Geles
- Black Diamond Therapeutics, New York, NY, USA
| | | | - Shohei Koide
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA.
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
13
|
Wang J, Li Z, Lang H, Fu W, Gao Y, Yin S, Sun P, Li Z, Huang J, Liu S, Zhu Y, Sun F, Li D, Gao P, Ang Gao. Cyclic-dinucleotide-induced filamentous assembly of phospholipases governs broad CBASS immunity. Cell 2025:S0092-8674(25)00457-X. [PMID: 40345202 DOI: 10.1016/j.cell.2025.04.022] [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: 07/17/2024] [Revised: 01/10/2025] [Accepted: 04/16/2025] [Indexed: 05/11/2025]
Abstract
Cyclic-oligonucleotide-based antiphage signaling systems (CBASS), a widespread antiviral bacterial immune system homologous to the mammalian cGAS-STING pathway, synthesizes cyclic nucleotide signals and triggers effector proteins to induce cell death and prevent viral propagation. Among various CBASS effectors, phospholipase effectors are the first to be discovered and are one of the most widespread families that sense cyclic dinucleotides to degrade cell membrane phospholipids. Here, we report that CBASS phospholipases assemble from a dimeric inactive state into active higher-order filamentous oligomers upon sensing cyclic dinucleotides. Using a combined approach of cryo-electron microscopy and X-ray crystallography, we have determined the structures of CBASS phospholipase in the inactive dimeric state, the cyclic-dinucleotide-bound active higher-order state, and the substrate-analog-bound catalytic mimicry state, thereby visualizing the complete conformational reorganization process. Complemented by functional assays of intermolecular binding, phospholipase enzymatic activity, in vitro membrane disruption, and in vivo antiphage efficiency, our work elucidates the mechanisms of assembly and activation of CBASS phospholipases.
Collapse
Affiliation(s)
- Jingge Wang
- Department of Radiology, Zhuhai People's Hospital, The Affiliated Hospital of Beijing Institute of Technology, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Zhao Li
- Department of Radiology, Zhuhai People's Hospital, The Affiliated Hospital of Beijing Institute of Technology, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Hao Lang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wenfeng Fu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yina Gao
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Sen Yin
- Department of Radiology, Zhuhai People's Hospital, The Affiliated Hospital of Beijing Institute of Technology, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Panpan Sun
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhaolong Li
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiafeng Huang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Songqing Liu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Yun Zhu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Fei Sun
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong 510530, China
| | - Dong Li
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Pu Gao
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Ang Gao
- Department of Radiology, Zhuhai People's Hospital, The Affiliated Hospital of Beijing Institute of Technology, School of Life Science, Beijing Institute of Technology, Beijing 100081, China; Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250118, China; Center for Cell and Gene Therapy, The First Hospital of China Medical University, Shenyang 110001, China.
| |
Collapse
|
14
|
Huang QJ, Kim R, Song K, Grigorieff N, Munro JB, Schiffer CA, Somasundaran M. Virion-associated influenza hemagglutinin clusters upon sialic acid binding visualized by cryoelectron tomography. Proc Natl Acad Sci U S A 2025; 122:e2426427122. [PMID: 40244672 PMCID: PMC12037027 DOI: 10.1073/pnas.2426427122] [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: 12/17/2024] [Accepted: 03/12/2025] [Indexed: 04/18/2025] Open
Abstract
Influenza viruses are enveloped, negative-sense single-stranded RNA viruses covered in a dense layer of glycoproteins. Hemagglutinin (HA) accounts for 80 to 90% of influenza glycoprotein and plays a role in host cell binding and membrane fusion. While previous studies have characterized structures of purified receptor-free and receptor-bound HA, the effect of receptor binding on HA organization and structure on virions remains unknown. Here, we used cryoelectron tomography to visualize influenza virions bound to a sialic acid receptor mimic. Overall, receptor binding did not result in significant changes in viral morphology; however, we observed rearrangements of HA trimer organization and orientation. Compared to the even interglycoprotein spacing of unliganded HA trimers, receptor binding promotes HA trimer clustering and the formation of a triplet of trimers. Subtomogram averaging and refinement yielded 8 to 10 Å reconstructions that allowed us to visualize specific contacts between HAs from neighboring trimers and identify molecular features that mediate clustering. Taken together, we present structural evidence that receptor binding triggers clustering of HA trimers, revealing an additional layer of HA dynamics and plasticity.
Collapse
Affiliation(s)
- Qiuyu J. Huang
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA01605
| | - Ryan Kim
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA01605
| | - Kangkang Song
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA01605
| | - Nikolaus Grigorieff
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, MA01605
| | - James B. Munro
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA01605
- Department of Microbiology, University of Massachusetts Chan Medical School, Worcester, MA01605
| | - Celia A. Schiffer
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA01605
| | - Mohan Somasundaran
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA01605
| |
Collapse
|
15
|
Sichrovsky M, Lacabanne D, Ruprecht JJ, Rana JJ, Stanik K, Dionysopoulou M, Sowton AP, King MS, Jones SA, Cooper L, Hardwick SW, Paris G, Chirgadze DY, Ding S, Fearnley IM, Palmer SM, Pardon E, Steyaert J, Leone V, Forrest LR, Tavoulari S, Kunji ERS. Molecular basis of pyruvate transport and inhibition of the human mitochondrial pyruvate carrier. SCIENCE ADVANCES 2025; 11:eadw1489. [PMID: 40249800 PMCID: PMC12007569 DOI: 10.1126/sciadv.adw1489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 03/14/2025] [Indexed: 04/20/2025]
Abstract
The mitochondrial pyruvate carrier transports pyruvate, produced by glycolysis from sugar molecules, into the mitochondrial matrix, as a crucial transport step in eukaryotic energy metabolism. The carrier is a drug target for the treatment of cancers, diabetes mellitus, neurodegeneration, and metabolic dysfunction-associated steatotic liver disease. We have solved the structure of the human MPC1L/MPC2 heterodimer in the inward- and outward-open states by cryo-electron microscopy, revealing its alternating access rocker-switch mechanism. The carrier has a central binding site for pyruvate, which contains an essential lysine and histidine residue, important for its ΔpH-dependent transport mechanism. We have also determined the binding poses of three chemically distinct inhibitor classes, which exploit the same binding site in the outward-open state by mimicking pyruvate interactions and by using aromatic stacking interactions.
Collapse
Affiliation(s)
- Maximilian Sichrovsky
- MRC Mitochondrial Biology Unit, University of Cambridge, Keith Peters Building, Cambridge Biomedical Campus, Cambridge CB2 0XY, UK
| | - Denis Lacabanne
- MRC Mitochondrial Biology Unit, University of Cambridge, Keith Peters Building, Cambridge Biomedical Campus, Cambridge CB2 0XY, UK
| | - Jonathan J. Ruprecht
- MRC Mitochondrial Biology Unit, University of Cambridge, Keith Peters Building, Cambridge Biomedical Campus, Cambridge CB2 0XY, UK
| | - Jessica J. Rana
- Computational Structural Biology Section, National Institutes of Neurological Disorders and Stroke, NIH, Bethesda, MD 20892, USA
| | - Klaudia Stanik
- MRC Mitochondrial Biology Unit, University of Cambridge, Keith Peters Building, Cambridge Biomedical Campus, Cambridge CB2 0XY, UK
| | - Mariangela Dionysopoulou
- MRC Mitochondrial Biology Unit, University of Cambridge, Keith Peters Building, Cambridge Biomedical Campus, Cambridge CB2 0XY, UK
| | - Alice P. Sowton
- MRC Mitochondrial Biology Unit, University of Cambridge, Keith Peters Building, Cambridge Biomedical Campus, Cambridge CB2 0XY, UK
| | - Martin S. King
- MRC Mitochondrial Biology Unit, University of Cambridge, Keith Peters Building, Cambridge Biomedical Campus, Cambridge CB2 0XY, UK
| | - Scott A. Jones
- MRC Mitochondrial Biology Unit, University of Cambridge, Keith Peters Building, Cambridge Biomedical Campus, Cambridge CB2 0XY, UK
| | - Lee Cooper
- Department of Biochemistry, University of Cambridge, Sanger Building, Tennis Court Road, Cambridge CB2 1GA, UK
| | - Steven W. Hardwick
- Department of Biochemistry, University of Cambridge, Sanger Building, Tennis Court Road, Cambridge CB2 1GA, UK
| | - Giulia Paris
- Department of Biochemistry, University of Cambridge, Sanger Building, Tennis Court Road, Cambridge CB2 1GA, UK
| | - Dimitri Y. Chirgadze
- Department of Biochemistry, University of Cambridge, Sanger Building, Tennis Court Road, Cambridge CB2 1GA, UK
| | - Shujing Ding
- MRC Mitochondrial Biology Unit, University of Cambridge, Keith Peters Building, Cambridge Biomedical Campus, Cambridge CB2 0XY, UK
| | - Ian M. Fearnley
- MRC Mitochondrial Biology Unit, University of Cambridge, Keith Peters Building, Cambridge Biomedical Campus, Cambridge CB2 0XY, UK
| | - Shane M. Palmer
- MRC Mitochondrial Biology Unit, University of Cambridge, Keith Peters Building, Cambridge Biomedical Campus, Cambridge CB2 0XY, UK
| | - Els Pardon
- VIB-VUB Center for Structural Biology, VIB, Pleinlaan 2, B-1050 Brussels, Belgium
- Structural Biology Brussels, Vrije Universiteit Brussel, Pleinlaan 2, B-1050 Brussels, Belgium
| | - Jan Steyaert
- VIB-VUB Center for Structural Biology, VIB, Pleinlaan 2, B-1050 Brussels, Belgium
- Structural Biology Brussels, Vrije Universiteit Brussel, Pleinlaan 2, B-1050 Brussels, Belgium
| | - Vanessa Leone
- Computational Structural Biology Section, National Institutes of Neurological Disorders and Stroke, NIH, Bethesda, MD 20892, USA
- Department of Biophysics and Data Science Institute, Medical College of Wisconsin, Milwaukee, WI 53226-3548, USA
| | - Lucy R. Forrest
- Computational Structural Biology Section, National Institutes of Neurological Disorders and Stroke, NIH, Bethesda, MD 20892, USA
| | - Sotiria Tavoulari
- MRC Mitochondrial Biology Unit, University of Cambridge, Keith Peters Building, Cambridge Biomedical Campus, Cambridge CB2 0XY, UK
| | - Edmund R. S. Kunji
- MRC Mitochondrial Biology Unit, University of Cambridge, Keith Peters Building, Cambridge Biomedical Campus, Cambridge CB2 0XY, UK
| |
Collapse
|
16
|
Klumpe S, Senti KA, Beck F, Sachweh J, Hampoelz B, Ronchi P, Oorschot V, Brandstetter M, Yeroslaviz A, Briggs JAG, Brennecke J, Beck M, Plitzko JM. In-cell structure and snapshots of copia retrotransposons in intact tissue by cryo-ET. Cell 2025; 188:2094-2110.e18. [PMID: 40049165 DOI: 10.1016/j.cell.2025.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 11/19/2024] [Accepted: 02/05/2025] [Indexed: 03/10/2025]
Abstract
Long terminal repeat (LTR) retrotransposons belong to the transposable elements (TEs), autonomously replicating genetic elements that integrate into the host's genome. Among animals, Drosophila melanogaster serves as an important model organism for TE research and contains several LTR retrotransposons, including the Ty1-copia family, which is evolutionarily related to retroviruses and forms virus-like particles (VLPs). In this study, we use cryo-focused ion beam (FIB) milling and lift-out approaches to visualize copia VLPs in ovarian cells and intact egg chambers, resolving the in situ copia capsid structure to 7.7 Å resolution by cryoelectron tomography (cryo-ET). Although cytoplasmic copia VLPs vary in size, nuclear VLPs are homogeneous and form densely packed clusters, supporting a model in which nuclear import acts as a size selector. Analyzing flies deficient in the TE-suppressing PIWI-interacting RNA (piRNA) pathway, we observe copia's translocation into the nucleus during spermatogenesis. Our findings provide insights into the replication cycle and cellular structural biology of an active LTR retrotransposon.
Collapse
Affiliation(s)
- Sven Klumpe
- Research Group CryoEM Technology, Max Planck Institute of Biochemistry, Martinsried, Germany.
| | - Kirsten A Senti
- Institute of Molecular Biotechnology Austria (IMBA), Vienna, Austria
| | - Florian Beck
- Research Group CryoEM Technology, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Jenny Sachweh
- Department Molecular Sociology, Max Planck Institute of Biophysics, Frankfurt, Germany
| | - Bernhard Hampoelz
- Department Molecular Sociology, Max Planck Institute of Biophysics, Frankfurt, Germany
| | - Paolo Ronchi
- EMBL EM Core Facility, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Viola Oorschot
- EMBL EM Core Facility, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | | | - Assa Yeroslaviz
- Computational Systems Biochemistry, Bioinformatics Core Facility, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - John A G Briggs
- Department of Cell and Virus Structure, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Julius Brennecke
- Institute of Molecular Biotechnology Austria (IMBA), Vienna, Austria.
| | - Martin Beck
- Department Molecular Sociology, Max Planck Institute of Biophysics, Frankfurt, Germany; Institute of Biochemistry, Goethe University Frankfurt, Frankfurt, Germany.
| | - Jürgen M Plitzko
- Research Group CryoEM Technology, Max Planck Institute of Biochemistry, Martinsried, Germany.
| |
Collapse
|
17
|
Nakamura K, Aoyama-Ishiwatari S, Nagao T, Paaran M, Obara CJ, Sakurai-Saito Y, Johnston J, Du Y, Suga S, Tsuboi M, Nakakido M, Tsumoto K, Kishi Y, Gotoh Y, Kwak C, Rhee HW, Seo JK, Kosako H, Potter C, Carragher B, Lippincott-Schwartz J, Polleux F, Hirabayashi Y. Mitochondrial complexity is regulated at ER-mitochondria contact sites via PDZD8-FKBP8 tethering. Nat Commun 2025; 16:3401. [PMID: 40246839 PMCID: PMC12006300 DOI: 10.1038/s41467-025-58538-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 03/24/2025] [Indexed: 04/19/2025] Open
Abstract
Mitochondria-ER membrane contact sites (MERCS) represent a fundamental ultrastructural feature underlying unique biochemistry and physiology in eukaryotic cells. The ER protein PDZD8 is required for the formation of MERCS in many cell types, however, its tethering partner on the outer mitochondrial membrane (OMM) is currently unknown. Here we identify the OMM protein FKBP8 as the tethering partner of PDZD8 using a combination of unbiased proximity proteomics, CRISPR-Cas9 endogenous protein tagging, Cryo-electron tomography, and correlative light-electron microscopy. Single molecule tracking reveals highly dynamic diffusion properties of PDZD8 along the ER membrane with significant pauses and captures at MERCS. Overexpression of FKBP8 is sufficient to narrow the ER-OMM distance, whereas independent versus combined deletions of these two proteins demonstrate their interdependence for MERCS formation. Furthermore, PDZD8 enhances mitochondrial complexity in a FKBP8-dependent manner. Our results identify a novel ER-mitochondria tethering complex that regulates mitochondrial morphology in mammalian cells.
Collapse
Affiliation(s)
- Koki Nakamura
- Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, Tokyo, 113-8656, Japan
| | - Saeko Aoyama-Ishiwatari
- Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, Tokyo, 113-8656, Japan
| | - Takahiro Nagao
- Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, Tokyo, 113-8656, Japan
| | - Mohammadreza Paaran
- Simons Electron Microscopy Center, New York Structural Biology Center, New York, NY, 10028, USA
- Chan Zuckerberg Imaging Institute, Redwood City, CA, USA
| | - Christopher J Obara
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, 20147, USA
| | - Yui Sakurai-Saito
- Department of Bioengineering, School of Engineering, The University of Tokyo, Tokyo, 113-8656, Japan
| | - Jake Johnston
- Simons Electron Microscopy Center, New York Structural Biology Center, New York, NY, 10028, USA
- Columbia University Medical Center, New York, NY, 10032, USA
| | - Yudan Du
- Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, Tokyo, 113-8656, Japan
| | - Shogo Suga
- Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, Tokyo, 113-8656, Japan
| | - Masafumi Tsuboi
- Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, Tokyo, 113-8656, Japan
| | - Makoto Nakakido
- Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, Tokyo, 113-8656, Japan
- Department of Bioengineering, School of Engineering, The University of Tokyo, Tokyo, 113-8656, Japan
| | - Kouhei Tsumoto
- Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, Tokyo, 113-8656, Japan
- Department of Bioengineering, School of Engineering, The University of Tokyo, Tokyo, 113-8656, Japan
- Medical Proteomics Laboratory, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan
| | - Yusuke Kishi
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, 113-0033, Japan
- Laboratory of Molecular Neurobiology, Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, 113-0032, Japan
| | - Yukiko Gotoh
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Chulhwan Kwak
- Department of Chemistry, Seoul National University, Seoul, 08826, Republic of Korea
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, 94304, USA
| | - Hyun-Woo Rhee
- School of Biological Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jeong Kon Seo
- Graduate School of Semiconductor Materials and Devices Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
- UNIST Central Research Facilities (UCRF), Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Korea
| | - Hidetaka Kosako
- Division of Cell Signaling, Fujii Memorial Institute of Medical Sciences, Institute of Advanced Medical Sciences, Tokushima University, Tokushima, 770-8503, Japan
| | - Clint Potter
- Simons Electron Microscopy Center, New York Structural Biology Center, New York, NY, 10028, USA
- Chan Zuckerberg Imaging Institute, Redwood City, CA, USA
| | - Bridget Carragher
- Simons Electron Microscopy Center, New York Structural Biology Center, New York, NY, 10028, USA
- Chan Zuckerberg Imaging Institute, Redwood City, CA, USA
| | | | - Franck Polleux
- Department of Neuroscience, Columbia University Medical Center, New York, NY, 10032, USA
- Mortimer B. Zuckerman Mind Brain Behavior Institute, New York, NY, 10027, USA
| | - Yusuke Hirabayashi
- Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, Tokyo, 113-8656, Japan.
- Department of Bioengineering, School of Engineering, The University of Tokyo, Tokyo, 113-8656, Japan.
| |
Collapse
|
18
|
Heinrichs M, Finke AF, Aibara S, Krempler A, Boshnakovska A, Rehling P, Hillen HS, Richter-Dennerlein R. Coupling of ribosome biogenesis and translation initiation in human mitochondria. Nat Commun 2025; 16:3641. [PMID: 40240327 PMCID: PMC12003892 DOI: 10.1038/s41467-025-58827-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 04/02/2025] [Indexed: 04/18/2025] Open
Abstract
Biogenesis of mitoribosomes requires dedicated chaperones, RNA-modifying enzymes, and GTPases, and defects in mitoribosome assembly lead to severe mitochondriopathies in humans. Here, we characterize late-step assembly states of the small mitoribosomal subunit (mtSSU) by combining genetic perturbation and mutagenesis analysis with biochemical and structural approaches. Isolation of native mtSSU biogenesis intermediates via a FLAG-tagged variant of the GTPase MTG3 reveals three distinct assembly states, which show how factors cooperate to mature the 12S rRNA. In addition, we observe four distinct primed initiation mtSSU states with an incompletely matured rRNA, suggesting that biogenesis and translation initiation are not mutually exclusive processes but can occur simultaneously. Together, these results provide insights into mtSSU biogenesis and suggest a functional coupling between ribosome biogenesis and translation initiation in human mitochondria.
Collapse
Affiliation(s)
- Marleen Heinrichs
- Department of Molecular Biology, 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
| | - Anna Franziska Finke
- Department of Cellular Biochemistry, University Medical Center Göttingen, Göttingen, Germany
- Research Group Structure and Function of Molecular Machines, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Shintaro Aibara
- Department of Molecular Biology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Angelique Krempler
- Department of Molecular Biology, University Medical Center Göttingen, Göttingen, Germany
| | - Angela Boshnakovska
- Department of Cellular Biochemistry, University Medical Center Göttingen, Göttingen, Germany
| | - Peter Rehling
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
- Department of Cellular Biochemistry, University Medical Center Göttingen, Göttingen, Germany
- Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Translational Neuroinflammation and Automated Microscopy, Göttingen, Germany
- Göttingen Center for Molecular Biosciences, University of Göttingen, Göttingen, Germany
| | - Hauke S Hillen
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany.
- Department of Cellular Biochemistry, University Medical Center Göttingen, Göttingen, Germany.
- Research Group Structure and Function of Molecular Machines, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany.
- Göttingen Center for Molecular Biosciences, University of Göttingen, Göttingen, Germany.
| | - Ricarda Richter-Dennerlein
- Department of Molecular Biology, 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.
- Göttingen Center for Molecular Biosciences, University of Göttingen, Göttingen, Germany.
| |
Collapse
|
19
|
Vishwakarma R, Marechal N, Morichaud Z, Blaise M, Margeat E, Brodolin K. Single-stranded DNA drives σ subunit loading onto mycobacterial RNA polymerase to unlock initiation-competent conformations. Nucleic Acids Res 2025; 53:gkaf272. [PMID: 40240004 PMCID: PMC12000874 DOI: 10.1093/nar/gkaf272] [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: 08/21/2024] [Revised: 03/20/2025] [Accepted: 03/26/2025] [Indexed: 04/18/2025] Open
Abstract
Initiation of transcription requires the formation of the "open" promoter complex (RPo). For this, the σ subunit of bacterial RNA polymerase (RNAP) binds to the nontemplate strand of the -10 element sequence of promoters and nucleates DNA unwinding. This is accompanied by a cascade of conformational changes on RNAP, the exact mechanics of which remains elusive. Here, using single-molecule Förster resonance energy transfer and cryo-electron microscopy, we explored the conformational landscape of RNAP from the human pathogen Mycobacterium tuberculosis upon binding to a single-stranded DNA (ssDNA) fragment that includes the -10 element sequence (-10 ssDNA). We found that like the transcription activator RNAP-binding protein A, -10 ssDNA induced σ subunit loading onto the DNA/RNA channels of RNAP. This triggered RNAP clamp closure and unswiveling that are required for RPo formation and RNA synthesis initiation. Our results reveal a mechanism of ssDNA-guided RNAP maturation and identify the σ subunit as a regulator of RNAP conformational dynamics.
Collapse
Affiliation(s)
- Rishi Kishore Vishwakarma
- Institut de Recherche en Infectiologie de Montpellier, Univ Montpellier, CNRS, Montpellier 34293, France
- Centre de Biologie Structurale, Univ Montpellier, CNRS, INSERM, Montpellier 34090, France
| | - Nils Marechal
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch 67404, France
| | - Zakia Morichaud
- Institut de Recherche en Infectiologie de Montpellier, Univ Montpellier, CNRS, Montpellier 34293, France
| | - Mickaël Blaise
- Institut de Recherche en Infectiologie de Montpellier, Univ Montpellier, CNRS, Montpellier 34293, France
| | - Emmanuel Margeat
- Centre de Biologie Structurale, Univ Montpellier, CNRS, INSERM, Montpellier 34090, France
| | - Konstantin Brodolin
- Institut de Recherche en Infectiologie de Montpellier, Univ Montpellier, CNRS, Montpellier 34293, France
- INSERM Occitanie Méditerranée, Montpellier 34394, France
| |
Collapse
|
20
|
Armbruster EG, Rani P, Lee J, Klusch N, Hutchings J, Hoffman LY, Buschkaemper H, Enustun E, Adler BA, Inlow K, VanderWal AR, Hoffman MY, Daksh D, Aindow A, Deep A, Rodriguez ZK, Morgan CJ, Ghassemian M, Laughlin TG, Charles E, Cress BF, Savage DF, Doudna JA, Pogliano K, Corbett KD, Villa E, Pogliano J. Sequential membrane- and protein-bound organelles compartmentalize genomes during phage infection. Cell Host Microbe 2025; 33:484-497.e6. [PMID: 40168997 DOI: 10.1016/j.chom.2025.03.005] [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: 01/27/2025] [Revised: 02/19/2025] [Accepted: 03/05/2025] [Indexed: 04/03/2025]
Abstract
Many eukaryotic viruses require membrane-bound compartments for replication, but no such organelles are known to be formed by prokaryotic viruses. Bacteriophages of the Chimalliviridae family sequester their genomes within a phage-generated organelle, the phage nucleus, which is enclosed by a lattice of the viral protein ChmA. We show that inhibiting phage nucleus formation arrests infections at an early stage in which the injected phage genome is enclosed within a membrane-bound early phage infection (EPI) vesicle. Early phage genes are expressed from the EPI vesicle, demonstrating its functionality as a prokaryotic, transcriptionally active, membrane-bound organelle. We also show that the phage nucleus is essential, with genome replication beginning after the injected DNA is transferred from the EPI vesicle to the phage nucleus. Our results show that Chimalliviridae require two sophisticated subcellular compartments of distinct compositions and functions that facilitate successive stages of the viral life cycle.
Collapse
Affiliation(s)
- Emily G Armbruster
- School of Biological Sciences, University of California, San Diego, La Jolla, San Diego, CA 92093, USA
| | - Phoolwanti Rani
- School of Biological Sciences, University of California, San Diego, La Jolla, San Diego, CA 92093, USA; Howard Hughes Medical Institute, University of California, San Diego, La Jolla, San Diego, CA 92093, USA
| | - Jina Lee
- School of Biological Sciences, University of California, San Diego, La Jolla, San Diego, CA 92093, USA
| | - Niklas Klusch
- School of Biological Sciences, University of California, San Diego, La Jolla, San Diego, CA 92093, USA; Howard Hughes Medical Institute, University of California, San Diego, La Jolla, San Diego, CA 92093, USA
| | - Joshua Hutchings
- School of Biological Sciences, University of California, San Diego, La Jolla, San Diego, CA 92093, USA; Howard Hughes Medical Institute, University of California, San Diego, La Jolla, San Diego, CA 92093, USA
| | - Lizbeth Y Hoffman
- School of Biological Sciences, University of California, San Diego, La Jolla, San Diego, CA 92093, USA; Howard Hughes Medical Institute, University of California, San Diego, La Jolla, San Diego, CA 92093, USA
| | - Hannah Buschkaemper
- School of Biological Sciences, University of California, San Diego, La Jolla, San Diego, CA 92093, USA; Gene Center and Department of Biochemistry, Ludwig Maximilian University of Munich, 80539 Munich, Germany
| | - Eray Enustun
- School of Biological Sciences, University of California, San Diego, La Jolla, San Diego, CA 92093, USA
| | - Benjamin A Adler
- California Institute for Quantitative Biosciences (QB3), University of California, Berkeley, Berkeley, CA 94720, USA; Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Koe Inlow
- School of Biological Sciences, University of California, San Diego, La Jolla, San Diego, CA 92093, USA
| | - Arica R VanderWal
- School of Biological Sciences, University of California, San Diego, La Jolla, San Diego, CA 92093, USA; Howard Hughes Medical Institute, University of California, San Diego, La Jolla, San Diego, CA 92093, USA
| | - Madelynn Y Hoffman
- School of Biological Sciences, University of California, San Diego, La Jolla, San Diego, CA 92093, USA
| | - Daksh Daksh
- National Institute of Science, Education and Research (NISER), Bhubaneshwar 752050, Orissa, India
| | - Ann Aindow
- School of Biological Sciences, University of California, San Diego, La Jolla, San Diego, CA 92093, USA
| | - Amar Deep
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, San Diego, CA 92093, USA
| | - Zaida K Rodriguez
- School of Biological Sciences, University of California, San Diego, La Jolla, San Diego, CA 92093, USA
| | - Chase J Morgan
- School of Biological Sciences, University of California, San Diego, La Jolla, San Diego, CA 92093, USA
| | - Majid Ghassemian
- Biomolecular and Proteomics Mass Spectrometry Facility, University of California, San Diego, La Jolla, San Diego, CA 92093, USA
| | - Thomas G Laughlin
- School of Biological Sciences, University of California, San Diego, La Jolla, San Diego, CA 92093, USA
| | - Emeric Charles
- Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Brady F Cress
- Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - David F Savage
- Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Jennifer A Doudna
- California Institute for Quantitative Biosciences (QB3), University of California, Berkeley, Berkeley, CA 94720, USA; Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Chemistry, University of California, Berkeley, Berkeley, CA 94720, USA; Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA; MBIB Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Kit Pogliano
- School of Biological Sciences, University of California, San Diego, La Jolla, San Diego, CA 92093, USA
| | - Kevin D Corbett
- School of Biological Sciences, University of California, San Diego, La Jolla, San Diego, CA 92093, USA; Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, San Diego, CA 92093, USA
| | - Elizabeth Villa
- School of Biological Sciences, University of California, San Diego, La Jolla, San Diego, CA 92093, USA; Howard Hughes Medical Institute, University of California, San Diego, La Jolla, San Diego, CA 92093, USA.
| | - Joe Pogliano
- School of Biological Sciences, University of California, San Diego, La Jolla, San Diego, CA 92093, USA.
| |
Collapse
|
21
|
Chang YT, Barad BA, Hamid J, Rahmani H, Zid BM, Grotjahn DA. Cytoplasmic ribosomes on mitochondria alter the local membrane environment for protein import. J Cell Biol 2025; 224:e202407110. [PMID: 40047641 PMCID: PMC11893167 DOI: 10.1083/jcb.202407110] [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: 07/17/2024] [Revised: 12/04/2024] [Accepted: 01/29/2025] [Indexed: 03/12/2025] Open
Abstract
Most of the mitochondria proteome is nuclear-encoded, synthesized by cytoplasmic ribosomes, and targeted to the mitochondria posttranslationally. However, a subset of mitochondrial-targeted proteins is imported co-translationally, although the molecular mechanisms governing this process remain unclear. We employ cellular cryo-electron tomography to visualize interactions between cytoplasmic ribosomes and mitochondria in Saccharomyces cerevisiae. We use surface morphometrics tools to identify a subset of ribosomes optimally oriented on mitochondrial membranes for protein import. This allows us to establish the first subtomogram average structure of a cytoplasmic ribosome at the mitochondrial surface in the native cellular context, which showed three distinct connections with the outer mitochondrial membrane surrounding the peptide exit tunnel. Further, this analysis demonstrated that cytoplasmic ribosomes primed for mitochondrial protein import cluster on the outer mitochondrial membrane at sites of local constrictions of the outer and inner mitochondrial membranes. Overall, our study reveals the architecture and the spatial organization of cytoplasmic ribosomes at the mitochondrial surface, providing a native cellular context to define the mechanisms that mediate efficient mitochondrial co-translational protein import.
Collapse
Affiliation(s)
- Ya-Ting Chang
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Benjamin A. Barad
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
- Department of Chemical Physiology and Biochemistry, School of Medicine, Oregon Health and Science University, Portland, OR, USA
| | - Juliette Hamid
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA, USA
| | - Hamidreza Rahmani
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Brian M. Zid
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA, USA
| | - Danielle A. Grotjahn
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| |
Collapse
|
22
|
Steinruecke F, Markert JW, Farnung L. Structural basis of human replisome progression into a nucleosome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.04.647053. [PMID: 40236192 PMCID: PMC11996536 DOI: 10.1101/2025.04.04.647053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Epigenetic inheritance requires the transfer of parental histones to newly synthesized DNA during eukaryotic chromosome replication, yet the structural mechanisms underlying replisome engagement with nucleosomes remain unclear. Here we establish an in vitro chromatin replication system and report four cryo-EM structures of the human replisome in complex with a parental nucleosome. The structures capture distinct states of nucleosomal DNA unwrapping and nucleosome integrity during nucleosome disassembly by the encroaching replisome.
Collapse
|
23
|
Kim J, Rhee J, Kang S, Jung M, Kim J, Jeon M, Park J, Ham J, Kim BH, Lee WC, Roh SH, Park J. Self-supervised machine learning framework for high-throughput electron microscopy. SCIENCE ADVANCES 2025; 11:eads5552. [PMID: 40173219 PMCID: PMC11963987 DOI: 10.1126/sciadv.ads5552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 02/26/2025] [Indexed: 04/04/2025]
Abstract
Transmission electron microscopy (TEM) is a crucial analysis method in materials science and structural biology, as it offers a high spatiotemporal resolution for structural characterization and reveals structure-property relationships and structural dynamics at atomic and molecular levels. Despite technical advancements in EM, the nature of the electron beam makes the EM imaging inherently detrimental to materials even in low-dose applications. We introduce SHINE, the Self-supervised High-throughput Image denoising Neural network for Electron microscopy, accelerating minimally invasive low-dose EM of diverse material systems. SHINE uses only a single raw image dataset with intrinsic noise, which makes it suitable for limited-size datasets and eliminates the need for expensive ground-truth training datasets. We quantitatively demonstrate that SHINE overcomes the information limit in the current high-resolution TEM, in situ liquid phase TEM, time-series scanning TEM, and cryo-TEM, facilitating unambiguous high-throughput structure analysis across a broad spectrum of materials.
Collapse
Affiliation(s)
- Joodeok Kim
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea
| | - Jinho Rhee
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea
| | - Sungsu Kang
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea
| | - Mingyu Jung
- School of Biological Sciences, Institute of Molecular Biology and Genetics, Seoul National University, Seoul 08826, Republic of Korea
| | - Jihoon Kim
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea
| | - Miji Jeon
- School of Biological Sciences, Institute of Molecular Biology and Genetics, Seoul National University, Seoul 08826, Republic of Korea
| | - Junsun Park
- School of Biological Sciences, Institute of Molecular Biology and Genetics, Seoul National University, Seoul 08826, Republic of Korea
| | - Jimin Ham
- Department of Mechanical Engineering, BK21FOUR ERICA-ACE Center, Hanyang University, Ansan, Gyeonggi 15588, Republic of Korea
| | - Byung Hyo Kim
- Department of Materials Science and Engineering, Soongsil University, Seoul 06978, Republic of Korea
| | - Won Chul Lee
- Department of Mechanical Engineering, BK21FOUR ERICA-ACE Center, Hanyang University, Ansan, Gyeonggi 15588, Republic of Korea
| | - Soung-Hun Roh
- School of Biological Sciences, Institute of Molecular Biology and Genetics, Seoul National University, Seoul 08826, Republic of Korea
| | - Jungwon Park
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea
- Institute of Engineering Research, College of Engineering, Seoul National University, Seoul 08826, Republic of Korea
- Advanced Institute of Convergence Technology, Seoul National University, Suwon 16229, Republic of Korea
- Hyundai Motor Group-Seoul National University (HMG-SNU) Joint Battery Research Center (JBRC), Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
24
|
Kidane AK, Rosenfeld JR, Johnston JD, Dubbeldam C, Paraan M, Obermeyer AC. Cationic amino acid identity and net charge influence condensate properties in E. coli. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.29.646054. [PMID: 40236055 PMCID: PMC11996340 DOI: 10.1101/2025.03.29.646054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Understanding the formation of biomolecular condensates (BMC) in biological systems has proven to be a paradigm shift in our understanding of the subcellular organization of biomacromolecules. From RNA metabolism, stress response mechanisms, and amyloidogenic pathologies, condensates have been implicated to play a role in a myriad of cellular phenomena. Despite their near ubiquity, we still do no wholly understand how the primary sequence of biomolecules influences their biophysical and rheological properties. Here, we aim to understand the impact of primary cationic amino acid composition on the properties of condensates. Using engineered recombinant proteins, we show that the formation and phase boundaries of coacervates formed between proteins and RNA is dependent on the cationic amino acid identity, as well as the net charge of the protein involved in condensation. Despite the equivalent charge between arginine and lysine at physiological pH, arginine has been shown to promote increased encapsulation efficiency and salt stability, as well as reduced protein mobility within condensates. We show that arginine-tagged globular proteins also have a higher salt resistance in vitro when compared to similar lysine-tagged globular proteins. This translates to a cellular context in which arginine tagged proteins promote increased condensate formation in model E. coli cells. We were also able to observe a reduction in the total fluorescent recovery and protein mobility within arginine-based condensates via FRAP. Together, these results suggest that in addition to electrostatic interactions and disorder as the main driving forces of phase separation in biological contexts, the primary sequence and side chain composition of proteins plays a significant role in dictating dynamics of coacervates.
Collapse
|
25
|
Tlučková K, Kaczmarek B, Salmazo A, Bernecky C. Mechanism of mammalian transcriptional repression by noncoding RNA. Nat Struct Mol Biol 2025; 32:607-612. [PMID: 39762629 PMCID: PMC11996674 DOI: 10.1038/s41594-024-01448-7] [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: 09/05/2023] [Accepted: 11/08/2024] [Indexed: 04/16/2025]
Abstract
Transcription by RNA polymerase II (Pol II) can be repressed by noncoding RNA, including the human RNA Alu. However, the mechanism by which endogenous RNAs repress transcription remains unclear. Here we present cryogenic-electron microscopy structures of Pol II bound to Alu RNA, which reveal that Alu RNA mimics how DNA and RNA bind to Pol II during transcription elongation. Further, we show how distinct domains of the general transcription factor TFIIF control repressive activity. Together, we reveal how a noncoding RNA can regulate mammalian gene expression.
Collapse
Affiliation(s)
- Katarína Tlučková
- Institute of Science and Technology Austria (ISTA), Klosterneuburg, Austria
| | - Beata Kaczmarek
- Institute of Science and Technology Austria (ISTA), Klosterneuburg, Austria
| | - Anita Salmazo
- Institute of Science and Technology Austria (ISTA), Klosterneuburg, Austria
| | - Carrie Bernecky
- Institute of Science and Technology Austria (ISTA), Klosterneuburg, Austria.
| |
Collapse
|
26
|
Bhatta A, Kuhle B, Yu RD, Spanaus L, Ditter K, Bohnsack KE, Hillen HS. Molecular basis of human nuclear and mitochondrial tRNA 3' processing. Nat Struct Mol Biol 2025; 32:613-624. [PMID: 39747487 PMCID: PMC11996679 DOI: 10.1038/s41594-024-01445-w] [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: 04/04/2024] [Accepted: 11/06/2024] [Indexed: 01/04/2025]
Abstract
Eukaryotic transfer RNA (tRNA) precursors undergo sequential processing steps to become mature tRNAs. In humans, ELAC2 carries out 3' end processing of both nucleus-encoded (nu-tRNAs) and mitochondria-encoded (mt-tRNAs) tRNAs. ELAC2 is self-sufficient for processing of nu-tRNAs but requires TRMT10C and SDR5C1 to process most mt-tRNAs. Here we show that TRMT10C and SDR5C1 specifically facilitate processing of structurally degenerate mt-tRNAs lacking the canonical elbow. Structures of ELAC2 in complex with TRMT10C, SDR5C1 and two divergent mt-tRNA substrates reveal two distinct mechanisms of pre-tRNA recognition. While canonical nu-tRNAs and mt-tRNAs are recognized by direct ELAC2-RNA interactions, processing of noncanonical mt-tRNAs depends on protein-protein interactions between ELAC2 and TRMT10C. These results provide the molecular basis for tRNA 3' processing in both the nucleus and the mitochondria and explain the organelle-specific requirement for additional factors. Moreover, they suggest that TRMT10C-SDR5C1 evolved as a mitochondrial tRNA maturation platform to compensate for the structural erosion of mt-tRNAs in bilaterian animals.
Collapse
Affiliation(s)
- Arjun Bhatta
- Department of Cellular Biochemistry, University Medical Center Göttingen, Göttingen, Germany
- Research Group Structure and Function of Molecular Machines, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- International Max Planck Research School for Molecular Biology, University of Göttingen, Göttingen, Germany
| | - Bernhard Kuhle
- Department of Cellular Biochemistry, University Medical Center Göttingen, Göttingen, Germany
- Research Group Structure and Function of Molecular Machines, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Ryan D Yu
- Department of Cellular Biochemistry, University Medical Center Göttingen, Göttingen, Germany
- Research Group Structure and Function of Molecular Machines, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- International Max Planck Research School for Molecular Biology, University of Göttingen, Göttingen, Germany
| | - Lucas Spanaus
- Department of Cellular Biochemistry, University Medical Center Göttingen, Göttingen, Germany
- Research Group Structure and Function of Molecular Machines, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Katja Ditter
- Department of Cellular Biochemistry, University Medical Center Göttingen, Göttingen, Germany
- Research Group Structure and Function of Molecular Machines, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Katherine E Bohnsack
- Department of Molecular Biology, University Medical Center Göttingen, Göttingen, Germany
| | - Hauke S Hillen
- Department of Cellular Biochemistry, University Medical Center Göttingen, Göttingen, Germany.
- Research Group Structure and Function of Molecular Machines, 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.
- Research Group Structure and Function of Molecular Machines, Göttingen Center for Molecular Biosciences (GZMB), University of Göttingen, Göttingen, Germany.
| |
Collapse
|
27
|
Lange F, Ratz M, Dohrke JN, Le Vasseur M, Wenzel D, Ilgen P, Riedel D, Jakobs S. In situ architecture of the human prohibitin complex. Nat Cell Biol 2025; 27:633-640. [PMID: 40119201 PMCID: PMC11991916 DOI: 10.1038/s41556-025-01620-1] [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: 02/23/2024] [Accepted: 01/14/2025] [Indexed: 03/24/2025]
Abstract
Prohibitins are a highly conserved family of proteins that have been implicated in a variety of functions including mitochondrial stress signalling and housekeeping, cell cycle progression, apoptosis, lifespan regulation and many others. The human prohibitins prohibitin 1 and prohibitin 2 have been proposed to act as scaffolds within the mitochondrial inner membrane, but their molecular organization has remained elusive. Here we determined the molecular organization of the human prohibitin complex within the mitochondrial inner membrane using an integrative structural biology approach combining quantitative western blotting, cryo-electron tomography, subtomogram averaging and molecular modelling. The proposed bell-shaped structure consists of 11 alternating prohibitin 1 and prohibitin 2 molecules. This study reveals an average of about 43 prohibitin complexes per crista, covering 1-3% of the crista membrane area. These findings provide a structural basis for understanding the functional contributions of prohibitins to the integrity and spatial organization of the mitochondrial inner membrane.
Collapse
Affiliation(s)
- Felix Lange
- Department of NanoBiophotonics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Clinic of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Michael Ratz
- Department of NanoBiophotonics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Jan-Niklas Dohrke
- Department of NanoBiophotonics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Clinic of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | | | - Dirk Wenzel
- Laboratory of Electron Microscopy, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Peter Ilgen
- Department of NanoBiophotonics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Clinic of Neurology, University Medical Center Göttingen, Göttingen, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Translational Neuroinflammation and Automated Microscopy, Göttingen, Germany
| | - Dietmar Riedel
- Laboratory of Electron Microscopy, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Stefan Jakobs
- Department of NanoBiophotonics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany.
- Clinic of Neurology, University Medical Center Göttingen, Göttingen, Germany.
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Translational Neuroinflammation and Automated Microscopy, Göttingen, Germany.
| |
Collapse
|
28
|
Readshaw JJ, Doyle LA, Puiu M, Kelly A, Nelson A, Kaiser AJ, McGuire S, Peralta-Acosta J, Smith DL, Stoddard BL, Kaiser BK, Blower TR. PglZ from Type I BREX phage defence systems is a metal-dependent nuclease that forms a sub-complex with BrxB. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.26.645558. [PMID: 40196517 PMCID: PMC11974810 DOI: 10.1101/2025.03.26.645558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
BREX (Bacteriophage Exclusion) systems, identified through shared identity with Pgl (Phage Growth Limitation) systems, are a widespread, highly diverse group of phage defence systems found throughout bacteria and archaea. The varied BREX Types harbour multiple protein subunits (between four and eight) and all encode a conserved putative phosphatase (PglZ aka BrxZ) and an equally conserved, putative ATPase (BrxC). Almost all BREX systems also contain a site-specific methyltransferase (PglX aka BrxX). Despite having determined the structure and fundamental biophysical and biochemical behaviours for the PglX methyltransferase, the BrxL effector, the BrxA DNA-binding protein and the BrxR transcriptional regulator, the mechanism by which BREX impedes phage replication remains largely undetermined. In this study, we identify a stable BREX sub-complex of PglZ:BrxB, validate the structure and dynamic behaviour of that sub-complex, and assess the biochemical activity of PglZ, revealing it to be a metal-dependent nuclease. PglZ can cleave cyclic oligonucleotides, linear oligonucleotides, plasmid DNA and both non-modified and modified linear phage genomes. PglZ nuclease activity has no obvious role in BREX-dependent methylation, but does contribute to BREX phage defence. BrxB binding does not impact PglZ nuclease activity. These data contribute to our growing understanding of the BREX phage defence mechanism.
Collapse
Affiliation(s)
- Jennifer J. Readshaw
- Department of Biosciences, Durham University, Stockton Road, Durham, DH1 3LE, UK
| | - Lindsey A. Doyle
- Division of Basic Sciences, Fred Hutchinson Cancer Center, 1100 Fairview Ave. N. Seattle WA 98019, USA
| | - Maria Puiu
- Department of Biosciences, Durham University, Stockton Road, Durham, DH1 3LE, UK
| | - Abigail Kelly
- Department of Biosciences, Durham University, Stockton Road, Durham, DH1 3LE, UK
| | - Andrew Nelson
- Department of Applied Sciences, University of Northumbria, Newcastle Upon Tyne NE1 8ST, UK
| | - Alex J. Kaiser
- Division of Basic Sciences, Fred Hutchinson Cancer Center, 1100 Fairview Ave. N. Seattle WA 98019, USA
| | - Sydney McGuire
- Department of Biology, Seattle University, 901 12 Ave. Seattle WA 98122, USA
| | | | - Darren L. Smith
- Department of Applied Sciences, University of Northumbria, Newcastle Upon Tyne NE1 8ST, UK
| | - Barry L. Stoddard
- Division of Basic Sciences, Fred Hutchinson Cancer Center, 1100 Fairview Ave. N. Seattle WA 98019, USA
| | - Brett K. Kaiser
- Department of Biology, Seattle University, 901 12 Ave. Seattle WA 98122, USA
| | - Tim R. Blower
- Department of Biosciences, Durham University, Stockton Road, Durham, DH1 3LE, UK
- New England Biolabs, 240 County Road, Ipswich, MA 01938, USA
| |
Collapse
|
29
|
Tollervey F, Rios MU, Zagoriy E, Woodruff JB, Mahamid J. Molecular architectures of centrosomes in C. elegans embryos visualized by cryo-electron tomography. Dev Cell 2025; 60:885-900.e5. [PMID: 39721584 PMCID: PMC11948214 DOI: 10.1016/j.devcel.2024.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 09/29/2024] [Accepted: 12/02/2024] [Indexed: 12/28/2024]
Abstract
Centrosomes organize microtubules that are essential for mitotic divisions in animal cells. They consist of centrioles surrounded by pericentriolar material (PCM). Questions related to mechanisms of centriole assembly, PCM organization, and spindle microtubule formation remain unanswered, partly due to limited availability of molecular-resolution structural data inside cells. Here, we use cryo-electron tomography to visualize centrosomes across the cell cycle in cells isolated from C. elegans embryos. We describe a pseudo-timeline of centriole assembly and identify distinct structural features in both mother and daughter centrioles. We find that centrioles and PCM microtubules differ in protofilament number (13 versus 11), which could be explained by atypical γ-tubulin ring complexes with 11-fold symmetry identified at the minus ends of short PCM microtubule segments. We further characterize a porous and disordered network that forms the interconnected PCM. Thus, our work builds a three-dimensional structural atlas that helps explain how centrosomes assemble, grow, and achieve function.
Collapse
Affiliation(s)
- Fergus Tollervey
- Structural and Computational Biology Unit, European Molecular Biology Laboratory (EMBL), 69117 Heidelberg, Germany; Collaboration for Joint PhD Degree between EMBL and Heidelberg University, Faculty of Biosciences, Heidelberg, Germany
| | - Manolo U Rios
- Department of Cell Biology and Biophysics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Evgenia Zagoriy
- Structural and Computational Biology Unit, European Molecular Biology Laboratory (EMBL), 69117 Heidelberg, Germany
| | - Jeffrey B Woodruff
- Department of Cell Biology and Biophysics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Julia Mahamid
- Structural and Computational Biology Unit, European Molecular Biology Laboratory (EMBL), 69117 Heidelberg, Germany; Cell Biology and Biophysics Unit, EMBL, 69117 Heidelberg, Germany.
| |
Collapse
|
30
|
Park YJ, Liu C, Lee J, Brown JT, Ma CB, Liu P, Gen R, Xiong Q, Zepeda SK, Stewart C, Addetia A, Craig CJ, Tortorici MA, Alshukairi AN, Starr TN, Yan H, Veesler D. Molecular basis of convergent evolution of ACE2 receptor utilization among HKU5 coronaviruses. Cell 2025; 188:1711-1728.e21. [PMID: 39922192 DOI: 10.1016/j.cell.2024.12.032] [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: 08/16/2024] [Revised: 10/25/2024] [Accepted: 12/20/2024] [Indexed: 02/10/2025]
Abstract
DPP4 was considered a canonical receptor for merbecoviruses until the recent discovery of African bat-borne MERS-related coronaviruses using ACE2. The extent and diversity of ACE2 utilization among merbecoviruses and their receptor species tropism remain unknown. Here, we reveal that HKU5 enters host cells utilizing Pipistrellus abramus (P.abr) and several non-bat mammalian ACE2s through a binding mode distinct from that of any other known ACE2-using coronaviruses. We defined the molecular determinants of receptor species tropism and identified a single amino acid mutation enabling HKU5 to utilize human ACE2, providing proof of principle for machine-learning-assisted outbreak preparedness. We show that MERS-CoV and HKU5 have markedly distinct antigenicity and identified several HKU5 inhibitors, including two clinical compounds. Our findings profoundly alter our understanding of coronavirus evolution, as several merbecovirus clades independently evolved ACE2 utilization, and pave the way for developing countermeasures against viruses poised for human emergence.
Collapse
Affiliation(s)
- Young-Jun Park
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA
| | - Chen Liu
- State Key Laboratory of Virology and Biosafety, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Jimin Lee
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Jack T Brown
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Cheng-Bao Ma
- State Key Laboratory of Virology and Biosafety, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Peng Liu
- State Key Laboratory of Virology and Biosafety, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Risako Gen
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Qing Xiong
- State Key Laboratory of Virology and Biosafety, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Samantha K Zepeda
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Cameron Stewart
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Amin Addetia
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Caroline J Craig
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | | | - Abeer N Alshukairi
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia; Department of Medicine, King Faisal Specialist Hospital and Research Center, Jeddah, Saudi Arabia
| | - Tyler N Starr
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Huan Yan
- State Key Laboratory of Virology and Biosafety, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei 430072, China.
| | - David Veesler
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
31
|
Schmidt J, Brandenburg V, Elders H, Shahzad S, Schäkermann S, Fiedler R, Knoke L, Pfänder Y, Dietze P, Bille H, Gärtner B, Albin L, Leichert L, Bandow J, Hofmann E, Narberhaus F. Two redox-responsive LysR-type transcription factors control the oxidative stress response of Agrobacterium tumefaciens. Nucleic Acids Res 2025; 53:gkaf267. [PMID: 40193708 PMCID: PMC11975290 DOI: 10.1093/nar/gkaf267] [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: 12/12/2024] [Revised: 03/10/2025] [Accepted: 03/24/2025] [Indexed: 04/09/2025] Open
Abstract
Pathogenic bacteria often encounter fluctuating reactive oxygen species (ROS) levels, particularly during host infection, necessitating robust redox-sensing mechanisms for survival. The LysR-type transcriptional regulator (LTTR) OxyR is a widely conserved bacterial thiol-based redox sensor. However, members of the Rhizobiales also encode LsrB, a second LTTR with potential redox-sensing function. This study explores the roles of OxyR and LsrB in the plant-pathogen Agrobacterium tumefaciens. Through single and combined deletions, we observed increased H2O2 sensitivity, underscoring their function in oxidative defense. Genome-wide transcriptome profiling under H2O2 exposure revealed that OxyR and LsrB co-regulate key antioxidant genes, including katG, encoding a bifunctional catalase/peroxidase. Agrobacterium tumefaciens LsrB possesses four cysteine residues potentially involved in redox sensing. To elucidate the structural basis for redox-sensing, we applied single-particle cryo-EM (cryogenic electron microscopy) to experimentally confirm an AlphaFold model of LsrB, identifying two proximal cysteine pairs. In vitro thiol-trapping coupled with mass spectrometry confirmed reversible thiol modifications of all four residues, suggesting a functional role in redox regulation. Collectively, these findings reveal that A. tumefaciens employs two cysteine-based redox sensing transcription factors, OxyR and LsrB, to withstand oxidative stress encountered in host and soil environments.
Collapse
Affiliation(s)
- Janka J Schmidt
- Microbial Biology, Ruhr University Bochum, 44801 Bochum, Germany
| | | | - Hannah Elders
- Protein Crystallography, Ruhr University Bochum, 44801 Bochum, Germany
| | - Saba Shahzad
- Ernst Ruska-Centre for Microscopy and Spectroscopy with Electrons (ER-C-3): Structural Biology, Institute of Biological Information Processing (IBI-6): Structural Cell Biology, Forschungszentrum Jülich, 52428 Jülich, Germany
| | - Sina Schäkermann
- Applied Microbiology, Ruhr University Bochum, 44801 Bochum, Germany
- Center for System-based Antibiotic Research, Ruhr University Bochum, 44801 Bochum, Germany
| | - Ronja Fiedler
- Microbial Biology, Ruhr University Bochum, 44801 Bochum, Germany
| | - Lisa R Knoke
- Microbial Biochemistry, Ruhr University Bochum, 44801 Bochum, Germany
| | - Yvonne Pfänder
- Microbial Biology, Ruhr University Bochum, 44801 Bochum, Germany
| | - Pascal Dietze
- Applied Microbiology, Ruhr University Bochum, 44801 Bochum, Germany
| | - Hannah Bille
- Microbial Biology, Ruhr University Bochum, 44801 Bochum, Germany
| | - Bela Gärtner
- Microbial Biology, Ruhr University Bochum, 44801 Bochum, Germany
| | - Lennart J Albin
- Microbial Biology, Ruhr University Bochum, 44801 Bochum, Germany
| | - Lars I Leichert
- Microbial Biochemistry, Ruhr University Bochum, 44801 Bochum, Germany
| | - Julia E Bandow
- Applied Microbiology, Ruhr University Bochum, 44801 Bochum, Germany
- Center for System-based Antibiotic Research, Ruhr University Bochum, 44801 Bochum, Germany
| | - Eckhard Hofmann
- Protein Crystallography, Ruhr University Bochum, 44801 Bochum, Germany
| | - Franz Narberhaus
- Microbial Biology, Ruhr University Bochum, 44801 Bochum, Germany
| |
Collapse
|
32
|
Helena-Bueno K, Kopetschke S, Filbeck S, Chan LI, Birsan S, Baslé A, Hudson M, Pfeffer S, Hill CH, Melnikov SV. Structurally heterogeneous ribosomes cooperate in protein synthesis in bacterial cells. Nat Commun 2025; 16:2751. [PMID: 40113756 PMCID: PMC11926189 DOI: 10.1038/s41467-025-57955-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 03/05/2025] [Indexed: 03/22/2025] Open
Abstract
Ribosome heterogeneity is a paradigm in biology, pertaining to the existence of structurally distinct populations of ribosomes within a single organism or cell. This concept suggests that structurally distinct pools of ribosomes have different functional properties and may be used to translate specific mRNAs. However, it is unknown to what extent structural heterogeneity reflects genuine functional specialization rather than stochastic variations in ribosome assembly. Here, we address this question by combining cryo-electron microscopy and tomography to observe individual structurally heterogeneous ribosomes in bacterial cells. We show that 70% of ribosomes in Psychrobacter urativorans contain a second copy of the ribosomal protein bS20 at a previously unknown binding site on the large ribosomal subunit. We then determine that this second bS20 copy appears to be functionally neutral. This demonstrates that ribosome heterogeneity does not necessarily lead to functional specialization, even when it involves significant variations such as the presence or absence of a ribosomal protein. Instead, we show that heterogeneous ribosomes can cooperate in general protein synthesis rather than specialize in translating discrete populations of mRNA.
Collapse
Affiliation(s)
| | - Sophie Kopetschke
- Centre for Molecular Biology, Heidelberg University, Heidelberg, Germany
| | - Sebastian Filbeck
- Centre for Molecular Biology, Heidelberg University, Heidelberg, Germany
| | - Lewis I Chan
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Sonia Birsan
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Arnaud Baslé
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Maisie Hudson
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Stefan Pfeffer
- Centre for Molecular Biology, Heidelberg University, Heidelberg, Germany.
| | - Chris H Hill
- York Structural Biology Laboratory, University of York, York, UK.
- York Biomedical Research Institute, University of York, York, UK.
- Department of Biology, University of York, York, UK.
| | - Sergey V Melnikov
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK.
| |
Collapse
|
33
|
Chiarugi S, Margheriti F, De Lorenzi V, Martino E, Margheritis EG, Moscardini A, Marotta R, Chaves-Sanjuan A, Del Seppia C, Federighi G, Lapi D, Bandiera T, Rapposelli S, Scuri R, Bolognesi M, Garau G. NAPE-PLD is target of thiazide diuretics. Cell Chem Biol 2025; 32:449-462.e5. [PMID: 39999832 DOI: 10.1016/j.chembiol.2025.01.008] [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: 10/21/2023] [Revised: 01/01/2025] [Accepted: 01/31/2025] [Indexed: 02/27/2025]
Abstract
Thiazide and thiazide-like diuretics are among the most efficacious and used drugs for the treatment of hypertension, edema, and major cardiovascular outcomes. Despite more then than six decades of clinical use, the molecular target and mechanism of action by which these drugs cure hypertension after long-term use have remained mysterious. Here we report the discovery and validation of a previously unknown renal and extrarenal target of these antihypertensives, the membrane-associated phospholipase N-acylphosphatidylethanolamine-specific phospholipase D (NAPE-PLD) of the endocannabinoid system. Structural and functional insights, together with preclinical studies in hypertensive rats, disclose the molecular and physiological basis by which thiazides cause acute diuresis and, at the same time, the distinctive chronic reduction of vascular resistance. Our results shed light on the mechanism of treatment of hypertension and will be useful for developing more efficacious medications for the management of vascular risk factors, as well as associated leukoencephalopathies and myelin disorders.
Collapse
Affiliation(s)
- Sara Chiarugi
- BioStructures Lab, Istituto Italiano di Tecnologia (IIT@NEST), Piazza San Silvestro 12, 56127 Pisa, Italy; Laboratorio NEST, Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy
| | - Francesco Margheriti
- BioStructures Lab, Istituto Italiano di Tecnologia (IIT@NEST), Piazza San Silvestro 12, 56127 Pisa, Italy; Laboratorio NEST, Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy
| | - Valentina De Lorenzi
- BioStructures Lab, Istituto Italiano di Tecnologia (IIT@NEST), Piazza San Silvestro 12, 56127 Pisa, Italy
| | - Elisa Martino
- BioStructures Lab, Istituto Italiano di Tecnologia (IIT@NEST), Piazza San Silvestro 12, 56127 Pisa, Italy; Laboratorio NEST, Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy
| | | | - Aldo Moscardini
- Laboratorio NEST, Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy
| | - Roberto Marotta
- Electron Microscopy Facility, Istituto Italiano di Tecnologia (IIT) Via Morego 30, 16163 Genova, Italy
| | - Antonio Chaves-Sanjuan
- Department of BioSciences, University of Milano, Via Celoria 26, 20133 Milano, Italy; Pediatric Research Center Fondazione R.E. Invernizzi and NOLIMITS Center, Università degli Studi di Milano, Milan, Italy
| | | | - Giuseppe Federighi
- Department of Translational Research on New Technologies in Medicine and Surgery, University of Pisa, Via San Zeno 31, 56126 Pisa, Italy
| | - Dominga Lapi
- Department of Biology, University of Pisa, Via Alessandro Volta 4, 56127 Pisa, Italy
| | - Tiziano Bandiera
- D3 PharmaChemistry, Istituto Italiano di Tecnologia (IIT), Via Morego 30, 16163 Genova, Italy
| | - Simona Rapposelli
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Rossana Scuri
- Department of Translational Research on New Technologies in Medicine and Surgery, University of Pisa, Via San Zeno 31, 56126 Pisa, Italy
| | - Martino Bolognesi
- Department of BioSciences, University of Milano, Via Celoria 26, 20133 Milano, Italy; Pediatric Research Center Fondazione R.E. Invernizzi and NOLIMITS Center, Università degli Studi di Milano, Milan, Italy
| | - Gianpiero Garau
- BioStructures Lab, Istituto Italiano di Tecnologia (IIT@NEST), Piazza San Silvestro 12, 56127 Pisa, Italy; Laboratorio NEST, Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy; Laboratori Aliveda, Viale Karol Wojtyla 19, 56042 Crespina Lorenzana, Pisa, Italy.
| |
Collapse
|
34
|
Ma CB, Liu C, Park YJ, Tang J, Chen J, Xiong Q, Lee J, Stewart C, Asarnow D, Brown J, Tortorici MA, Yang X, Sun YH, Chen YM, Yu X, Si JY, Liu P, Tong F, Huang ML, Li J, Shi ZL, Deng Z, Veesler D, Yan H. Multiple independent acquisitions of ACE2 usage in MERS-related coronaviruses. Cell 2025; 188:1693-1710.e18. [PMID: 39922191 DOI: 10.1016/j.cell.2024.12.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 09/20/2024] [Accepted: 12/20/2024] [Indexed: 02/10/2025]
Abstract
The angiotensin-converting enzyme 2 (ACE2) receptor is shared by various coronaviruses with distinct receptor-binding domain (RBD) architectures, yet our understanding of these convergent acquisition events remains elusive. Here, we report that two bat MERS-related coronaviruses (MERSr-CoVs) infecting Pipistrellus nathusii (P.nat)-MOW15-22 and PnNL2018B-use ACE2 as their receptor, with narrow ortholog specificity. Cryoelectron microscopy structures of the MOW15-22/PnNL2018B RBD-ACE2 complexes unveil an unexpected and entirely distinct binding mode, mapping >45 Å away from that of any other known ACE2-using coronaviruses. Functional profiling of ACE2 orthologs from 105 mammalian species led to the identification of host tropism determinants, including an ACE2 N432-glycosylation restricting viral recognition, and the design of a soluble P.nat ACE2 mutant with potent viral neutralizing activity. Our findings reveal convergent acquisition of ACE2 usage for merbecoviruses found in European bats, underscoring the extraordinary diversity of ACE2 recognition modes among coronaviruses and the promiscuity of this receptor.
Collapse
Affiliation(s)
- Cheng-Bao Ma
- State Key Laboratory of Virology and Biosafety, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430072, Hubei, China
| | - Chen Liu
- State Key Laboratory of Virology and Biosafety, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430072, Hubei, China
| | - Young-Jun Park
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA
| | - Jingjing Tang
- State Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Jing Chen
- State Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Qing Xiong
- State Key Laboratory of Virology and Biosafety, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430072, Hubei, China
| | - Jimin Lee
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Cameron Stewart
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Daniel Asarnow
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Jack Brown
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | | | - Xiao Yang
- State Key Laboratory of Virology and Biosafety, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430072, Hubei, China
| | - Ye-Hui Sun
- State Key Laboratory of Virology and Biosafety, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430072, Hubei, China
| | - Yuan-Mei Chen
- State Key Laboratory of Virology and Biosafety, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430072, Hubei, China
| | - Xiao Yu
- State Key Laboratory of Virology and Biosafety, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430072, Hubei, China
| | - Jun-Yu Si
- State Key Laboratory of Virology and Biosafety, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430072, Hubei, China
| | - Peng Liu
- State Key Laboratory of Virology and Biosafety, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430072, Hubei, China
| | - Fei Tong
- State Key Laboratory of Virology and Biosafety, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430072, Hubei, China
| | - Mei-Ling Huang
- State Key Laboratory of Virology and Biosafety, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430072, Hubei, China
| | - Jing Li
- State Key Laboratory of Virology and Biosafety, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430072, Hubei, China
| | - Zheng-Li Shi
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou 510005, China.
| | - Zengqin Deng
- State Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China; Hubei Jiangxia Laboratory, Wuhan 430207, China.
| | - David Veesler
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA.
| | - Huan Yan
- State Key Laboratory of Virology and Biosafety, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430072, Hubei, China.
| |
Collapse
|
35
|
Wang J, Ma Y, Li Z, Yuan H, Liu B, Li Z, Su M, Habib G, Liu Y, Fu L, Wang P, Li M, He J, Chen J, Zhou P, Shi Z, Chen X, Xiong X. SARS-related coronavirus S-protein structures reveal synergistic RBM interactions underpinning high-affinity human ACE2 binding. SCIENCE ADVANCES 2025; 11:eadr8772. [PMID: 40085715 PMCID: PMC11908486 DOI: 10.1126/sciadv.adr8772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 02/07/2025] [Indexed: 03/16/2025]
Abstract
High-affinity and specific binding toward the human angiotensin-converting enzyme 2 (hACE2) receptor by severe acute respiratory syndrome coronavirus (SARS)-related coronaviruses (SARSr-CoVs) remains incompletely understood. We report cryo-electron microscopy structures of eight different S-proteins from SARSr-CoVs found across Asia, Europe, and Africa. These S-proteins all adopt tightly packed, locked, prefusion conformations. These structures enable the classification of SARSr-CoV S-proteins into three types, based on their receptor-binding motif (RBM) structures and ACE2 binding characteristics. Type-2 S-proteins often preferentially bind bat ACE2 (bACE2) over hACE2. We report a structure of a type-2 BtKY72-RBD in complex with bACE2 to understand ACE2 specificity. Structure-guided mutagenesis of BtKY72-RBD reveals that multiple synergistic mutations in four different regions of RBM are required to achieve high-affinity hACE2 binding. Similar RBM changes can also confer hACE2 binding to another type-2 BM48-31 S-protein, which is primarily non-ACE2 binding. These results provide an understanding of how high-affinity hACE2 binding may be acquired by SARSr-CoV S-proteins.
Collapse
Affiliation(s)
- Jingjing Wang
- State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Research Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Yong Ma
- State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Research Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Zimu Li
- State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Research Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Guangzhou National Laboratory, Guangzhou, Guangdong, China
- Graduate School of Guangzhou Medical University, Guangzhou, China
| | - Hang Yuan
- State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Research Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Banghui Liu
- State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Research Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Zexuan Li
- State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Research Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Mengzhen Su
- State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Research Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Science and Technology of China, Hefei, China
| | - Gul Habib
- State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Research Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Yutong Liu
- State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Research Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Lutang Fu
- Cryo-electron Microscopy Center, Southern University of Science and Technology, Shenzhen, China
| | - Peiyi Wang
- Cryo-electron Microscopy Center, Southern University of Science and Technology, Shenzhen, China
| | - Mei Li
- Guangzhou National Laboratory, Guangzhou, Guangdong, China
| | - Jun He
- State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Research Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Jing Chen
- Guangzhou National Laboratory, Guangzhou, Guangdong, China
| | - Peng Zhou
- Guangzhou National Laboratory, Guangzhou, Guangdong, China
| | - Zhengli Shi
- Guangzhou National Laboratory, Guangzhou, Guangdong, China
| | - Xinwen Chen
- Guangzhou National Laboratory, Guangzhou, Guangdong, China
| | - Xiaoli Xiong
- State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Research Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| |
Collapse
|
36
|
Liu C, Park YJ, Ma CB, Stuart C, Gen R, Sun YC, Yang X, Lin MY, Xiong Q, Si JY, Liu P, Veesler D, Yan H. ACE2 utilization of HKU25 clade MERS-related coronaviruses with broad geographic distribution. RESEARCH SQUARE 2025:rs.3.rs-6097445. [PMID: 40162213 PMCID: PMC11952669 DOI: 10.21203/rs.3.rs-6097445/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Dipeptidyl peptidase-4 (DPP4) is a well-established receptor for several MERS-related coronaviruses (MERSr-CoVs) isolated from humans, camels, pangolins, and bats 1-6. However, the receptor usage of many genetically diverse bat MERSr-CoVs with broad geographical distributions remains poorly understood. Recent studies have identified angiotensin-converting enzyme 2 (ACE2) as an entry receptor for multiple merbecovirus clades. Here, using viral antigen and pseudovirus-based functional assays, we demonstrate that several bat merbecoviruses from the HKU25 clade previously thought to utilize DPP4 7, employ ACE2 as their functional receptor. Cryo-electron microscopy analysis revealed that HsItaly2011 and VsCoV-a7 recognize ACE2 with a binding mode sharing similarity with that of HKU5 but involving remodeled interfaces and distinct ortholog selectivity, suggesting a common evolutionary origin of ACE2 utilization for these two clades of viruses. EjCoV-3, a strain closely related to the DPP4-using MERSr-CoV BtCoV-422, exhibited relatively broad ACE2 ortholog tropism and could utilize human ACE2 albeit suboptimally. Despite differences in entry mechanisms and spike proteolytic activation compared to MERS-CoV, these viruses remain sensitive to several broadly neutralizing antibodies and entry inhibitors. These findings redefine our understanding of the evolution of receptor usage among MERSr-CoVs and highlight the versatility of ACE2 as a functional receptor for diverse coronaviruses.
Collapse
Affiliation(s)
- Chen Liu
- State Key Laboratory of Virology and Biosafety, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University; Wuhan, Hubei, 430072, China
| | - Young-Jun Park
- Department of Biochemistry, University of Washington; Seattle, WA 98195, USA
- Howard Hughes Medical Institute, University of Washington; Seattle, WA 98195, USA
| | - Cheng-Bao Ma
- State Key Laboratory of Virology and Biosafety, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University; Wuhan, Hubei, 430072, China
| | - Cameron Stuart
- Department of Biochemistry, University of Washington; Seattle, WA 98195, USA
| | - Risako Gen
- Department of Biochemistry, University of Washington; Seattle, WA 98195, USA
| | - Yu-Cheng Sun
- State Key Laboratory of Virology and Biosafety, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University; Wuhan, Hubei, 430072, China
| | - Xiao Yang
- State Key Laboratory of Virology and Biosafety, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University; Wuhan, Hubei, 430072, China
| | - Mei-Yi Lin
- State Key Laboratory of Virology and Biosafety, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University; Wuhan, Hubei, 430072, China
| | - Qing Xiong
- State Key Laboratory of Virology and Biosafety, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University; Wuhan, Hubei, 430072, China
| | - Jun-Yu Si
- State Key Laboratory of Virology and Biosafety, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University; Wuhan, Hubei, 430072, China
| | - Peng Liu
- State Key Laboratory of Virology and Biosafety, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University; Wuhan, Hubei, 430072, China
| | - David Veesler
- Department of Biochemistry, University of Washington; Seattle, WA 98195, USA
- Howard Hughes Medical Institute, University of Washington; Seattle, WA 98195, USA
| | - Huan Yan
- State Key Laboratory of Virology and Biosafety, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University; Wuhan, Hubei, 430072, China
| |
Collapse
|
37
|
Gao Q, Hofer FW, Filbeck S, Vermeulen BJA, Würtz M, Neuner A, Kaplan C, Zezlina M, Sala C, Shin H, Gruss OJ, Schiebel E, Pfeffer S. Structural mechanisms for centrosomal recruitment and organization of the microtubule nucleator γ-TuRC. Nat Commun 2025; 16:2453. [PMID: 40074789 PMCID: PMC11903878 DOI: 10.1038/s41467-025-57729-2] [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: 06/22/2024] [Accepted: 02/27/2025] [Indexed: 03/14/2025] Open
Abstract
The γ-tubulin ring complex (γ-TuRC) acts as a structural template for microtubule formation at centrosomes, associating with two main compartments: the pericentriolar material and the centriole lumen. In the pericentriolar material, the γ-TuRC is involved in microtubule organization, while the function of the centriole lumenal pool remains unclear. The conformational landscape of the γ-TuRC, which is crucial for its activity, and its centrosomal anchoring mechanisms, which determine γ-TuRC activity and turnover, are not understood. Using cryo-electron tomography, we analyze γ-TuRCs in human cells and purified centrosomes. Pericentriolar γ-TuRCs simultaneously associate with the essential adapter NEDD1 and the microcephaly protein CDK5RAP2. NEDD1 forms a tetrameric structure at the γ-TuRC base through interactions with four GCP3/MZT1 modules and GCP5/6-specific extensions, while multiple copies of CDK5RAP2 engage the γ-TuRC in two distinct binding patterns to promote γ-TuRC closure and activation. In the centriole lumen, the microtubule branching factor Augmin tethers a condensed cluster of γ-TuRCs to the centriole wall with defined directional orientation. Centriole-lumenal γ-TuRC-Augmin is protected from degradation during interphase and released in mitosis to aid chromosome alignment. This study provides a unique view on γ-TuRC structure and molecular organization at centrosomes and identifies an important cellular function of centriole-lumenal γ-TuRCs.
Collapse
Affiliation(s)
- Qi Gao
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), Heidelberg, Germany
| | - Florian W Hofer
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), Heidelberg, Germany
| | - Sebastian Filbeck
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), Heidelberg, Germany
| | - Bram J A Vermeulen
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), Heidelberg, Germany
| | - Martin Würtz
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), Heidelberg, Germany
- European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Annett Neuner
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), Heidelberg, Germany
| | | | - Maja Zezlina
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), Heidelberg, Germany
| | - Cornelia Sala
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), Heidelberg, Germany
| | - Hyesu Shin
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), Heidelberg, Germany
| | | | - Elmar Schiebel
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), Heidelberg, Germany.
| | - Stefan Pfeffer
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), Heidelberg, Germany.
| |
Collapse
|
38
|
Kabinger F, Doze V, Schmitzová J, Lidschreiber M, Dienemann C, Cramer P. Structural basis of SARS-CoV-2 polymerase inhibition by nonnucleoside inhibitor HeE1-2Tyr. Proc Natl Acad Sci U S A 2025; 122:e2419854122. [PMID: 40035759 PMCID: PMC11912441 DOI: 10.1073/pnas.2419854122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 01/18/2025] [Indexed: 03/06/2025] Open
Abstract
Targeting the RNA-dependent RNA polymerase (RdRp) of SARS-CoV-2 with small molecules is a promising therapeutic strategy against COVID-19, but potent and safe inhibitors are lacking. HeE1-2Tyr, a nonnucleoside inhibitor of Dengue virus RdRp, was also shown to inhibit SARS-CoV-2 RdRp in vitro and to have antiviral activity in cells, but the underlying mechanism remains unclear. Here, we elucidate the molecular mechanism of HeE1-2Tyr-mediated SARS-CoV-2 RdRp inhibition. Biochemical assays confirm that HeE1-2Tyr inhibits RdRp with an IC50 of 5 µM and show that it competes with RNA binding to RdRp in vitro. Structural analysis using cryo-EM reveals that a stack of three HeE1-2Tyr molecules binds to the RNA binding site of RdRp. The identification of the conserved HeE1-2Tyr binding site and its intriguing inhibition mechanism of three stacked molecules that outcompete RNA may facilitate further development of pan-corona nonnucleoside inhibitors.
Collapse
Affiliation(s)
- Florian Kabinger
- Department of Molecular Biology, Max Planck Institute for Multidisciplinary Sciences, Göttingen37077, Germany
| | - Valerie Doze
- Department of Molecular Biology, Max Planck Institute for Multidisciplinary Sciences, Göttingen37077, Germany
| | - Jana Schmitzová
- Department of Molecular Biology, Max Planck Institute for Multidisciplinary Sciences, Göttingen37077, Germany
| | - Michael Lidschreiber
- Department of Molecular Biology, Max Planck Institute for Multidisciplinary Sciences, Göttingen37077, Germany
| | - Christian Dienemann
- Department of Molecular Biology, Max Planck Institute for Multidisciplinary Sciences, Göttingen37077, Germany
| | - Patrick Cramer
- Department of Molecular Biology, Max Planck Institute for Multidisciplinary Sciences, Göttingen37077, Germany
| |
Collapse
|
39
|
Ning J, Glausier JR, Warshamanage R, Gunther-Cummins L, Burnley T, Palmer CM, Gonzalez-Burgos G, Miyamae T, Wang J, Carlisle D, Hsieh C, Schmelzer T, Buck SA, Franks J, Hampton CM, Stauffer WR, Lewis DA, Friedlander RM, Macaluso FP, Winn M, Marko M, Freyberg Z. Uncovering synaptic and cellular nanoarchitecture of brain tissue via seamless in situ trimming and milling for cryo-electron tomography. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.09.642162. [PMID: 40161621 PMCID: PMC11952431 DOI: 10.1101/2025.03.09.642162] [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: 04/02/2025]
Abstract
Cell-cell communication underlies all emergent properties of the brain, including cognition, learning and memory. The physical basis for these communications is the synapse, a multi-component structure requiring coordinated interactions between diverse cell types. However, many aspects of three-dimensional (3D) synaptic organization remain poorly understood. Here, we developed an approach, seamless in situ trimming and milling (SISTM), to reliably fabricate sufficiently thin lamellae for mapping of the 3D nanoarchitecture of synapses in mouse, monkey and human brain tissue under near-native conditions via cryo-electron tomography (cryo-ET). We validated SISTM in a mouse model of Huntington's disease, demonstrating distinct 3D alterations to synaptic vesicles and mitochondria. By successfully applying SISTM to macaque brain, we described the 3D architecture of a tripartite synapse within the cortex. Subtomogram averaging (STA) enabled spatial mapping of astrocyte-neuron contacts within the tripartite synapse, revealing neurexin-neuroligin complexes as potential constituents that tether the two cell types. Finally, we showed that the defining features of synaptic nanoarchitecture were conserved across species and evident in human brain tissue obtained postmortem. Combining SISTM with cryo-ET and STA is a starting point for a new understanding of brain organization, disease-induced structural alterations and the development of rational, structure-guided therapeutics.
Collapse
Affiliation(s)
- Jiying Ning
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Jill R. Glausier
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Rangana Warshamanage
- Scientific Computing Department, Science and Technology Facilities Council, Research Complex at Harwell, Didcot OX11 0FA, UK
| | | | - Tom Burnley
- Scientific Computing Department, Science and Technology Facilities Council, Research Complex at Harwell, Didcot OX11 0FA, UK
| | - Colin M. Palmer
- Scientific Computing Department, Science and Technology Facilities Council, Research Complex at Harwell, Didcot OX11 0FA, UK
| | | | - Takeaki Miyamae
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Jing Wang
- Thermo Fisher Scientific, 5350 NE Dawson Creek Drive, Hillsboro, OR, 97124, USA
| | - Diane Carlisle
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Chyongere Hsieh
- Wadsworth Center, New York State Department of Health, Empire State Plaza, Albany, NY 12201, USA
| | | | - Silas A. Buck
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Jonathan Franks
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Center for Biological Imaging, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Cheri M. Hampton
- Materials and Manufacturing Directorate, Air Force Research Laboratory, Wright-Patterson Air Force Base, Dayton, OH, 45433, USA
- BlueHalo, Dayton, OH 45432, USA
| | - William R. Stauffer
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - David A. Lewis
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Robert M. Friedlander
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Frank P. Macaluso
- Analytical Imaging Facility, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Martyn Winn
- Scientific Computing Department, Science and Technology Facilities Council, Research Complex at Harwell, Didcot OX11 0FA, UK
| | - Michael Marko
- Wadsworth Center, New York State Department of Health, Empire State Plaza, Albany, NY 12201, USA
| | - Zachary Freyberg
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| |
Collapse
|
40
|
Berger C, Watson H, Naismith JH, Dumoux M, Grange M. Xenon plasma focused ion beam lamella fabrication on high-pressure frozen specimens for structural cell biology. Nat Commun 2025; 16:2286. [PMID: 40055361 PMCID: PMC11889171 DOI: 10.1038/s41467-025-57493-3] [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: 06/18/2024] [Accepted: 02/24/2025] [Indexed: 05/13/2025] Open
Abstract
Cryo focused ion beam lamella preparation is a potent tool for in situ structural biology, enabling the study of macromolecules in their native cellular environments. However, throughput is currently limited, especially for thicker, more biologically complex samples. We describe how xenon plasma focused ion beam milling can be used for routine bulk milling of thicker, high-pressure frozen samples. We demonstrate lamellae preparation with a high success rate on these samples and determine a 4.0 Å structure of the Escherichia coli ribosome on these lamellae using sub volume averaging. We determine the effects on sample integrity of increased ion currents up to 60 nA during bulk milling of thicker planar samples, showing no measurable damage to macromolecules beyond an amorphous layer on the backside of the lamellae. The use of xenon results in substantial structural damage to particles up to approximately 30 nm in depth from the milled surfaces, and the effects of damage become negligibly small by 45 nm. Our results outline how the use of high currents using xenon plasma focused ion beam milling may be integrated into FIB milling regimes for preparing thin lamellae for high-resolution in situ structural biology.
Collapse
Affiliation(s)
- Casper Berger
- Structural Biology, The Rosalind Franklin Institute, Harwell Science & Innovation Campus, Didcot, OX11 0QS, UK
| | - Helena Watson
- Structural Biology, The Rosalind Franklin Institute, Harwell Science & Innovation Campus, Didcot, OX11 0QS, UK
- School of Molecular Biosciences, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - James H Naismith
- Structural Biology, The Rosalind Franklin Institute, Harwell Science & Innovation Campus, Didcot, OX11 0QS, UK
- Mathematical, Physical and Life Sciences Division, University of Oxford, Oxford, UK
| | - Maud Dumoux
- Structural Biology, The Rosalind Franklin Institute, Harwell Science & Innovation Campus, Didcot, OX11 0QS, UK
| | - Michael Grange
- Structural Biology, The Rosalind Franklin Institute, Harwell Science & Innovation Campus, Didcot, OX11 0QS, UK.
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, OX3 7BN, Oxford, United Kingdom.
| |
Collapse
|
41
|
Naganuma M, Kujirai T, Ehara H, Uejima T, Ito T, Goto M, Aoki M, Henmi M, Miyamoto-Kohno S, Shirouzu M, Kurumizaka H, Sekine SI. Structural insights into promoter-proximal pausing of RNA polymerase II at +1 nucleosome. SCIENCE ADVANCES 2025; 11:eadu0577. [PMID: 40043114 PMCID: PMC11881899 DOI: 10.1126/sciadv.adu0577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 01/29/2025] [Indexed: 05/13/2025]
Abstract
The metazoan transcription elongation complex (EC) of RNA polymerase II (RNAPII) generally stalls between the transcription start site and the first (+1) nucleosome. This promoter-proximal pausing involves negative elongation factor (NELF), 5,6-dichloro-1-β-d-ribobenzimidazole sensitivity-inducing factor (DSIF), and transcription elongation factor IIS (TFIIS) and is critical for subsequent productive transcription elongation. However, the detailed pausing mechanism and the involvement of the +1 nucleosome remain enigmatic. Here, we report cryo-electron microscopy structures of ECs stalled on nucleosomal DNA. In the absence of TFIIS, the EC is backtracked/arrested due to conflicts between NELF and the nucleosome. We identified two alternative binding modes of NELF, one of which reveals a critical contact with the downstream DNA through the conserved NELF-E basic helix. Upon binding with TFIIS, the EC progressed to the nucleosome to establish a paused EC with a partially unwrapped nucleosome. This paused EC strongly restricts EC progression further downstream. These structures illuminate the mechanism of RNAPII pausing/stalling at the +1 nucleosome.
Collapse
Affiliation(s)
- Masahiro Naganuma
- RIKEN Center for Biosystems Dynamics Research, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan
| | - Tomoya Kujirai
- RIKEN Center for Biosystems Dynamics Research, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan
- Laboratory of Chromatin Structure and Function, Institute for Quantitative Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-0032, Japan
| | - Haruhiko Ehara
- RIKEN Center for Biosystems Dynamics Research, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan
| | - Tamami Uejima
- RIKEN Center for Biosystems Dynamics Research, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan
| | - Tomoko Ito
- Laboratory of Chromatin Structure and Function, Institute for Quantitative Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-0032, Japan
| | - Mie Goto
- RIKEN Center for Biosystems Dynamics Research, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan
| | - Mari Aoki
- RIKEN Center for Biosystems Dynamics Research, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan
| | - Masami Henmi
- RIKEN Center for Biosystems Dynamics Research, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan
| | - Sayako Miyamoto-Kohno
- RIKEN Center for Biosystems Dynamics Research, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan
| | - Mikako Shirouzu
- RIKEN Center for Biosystems Dynamics Research, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan
| | - Hitoshi Kurumizaka
- RIKEN Center for Biosystems Dynamics Research, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan
- Laboratory of Chromatin Structure and Function, Institute for Quantitative Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-0032, Japan
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-0032, Japan
| | - Shun-ichi Sekine
- RIKEN Center for Biosystems Dynamics Research, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan
| |
Collapse
|
42
|
Sala FA, Ditter K, Dybkov O, Urlaub H, Hillen HS. Structural basis of Nipah virus RNA synthesis. Nat Commun 2025; 16:2261. [PMID: 40050611 PMCID: PMC11885841 DOI: 10.1038/s41467-025-57219-5] [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: 10/12/2024] [Accepted: 02/14/2025] [Indexed: 03/09/2025] Open
Abstract
Nipah virus (NiV) is a non-segmented negative-strand RNA virus (nsNSV) with high pandemic potential, as it frequently causes zoonotic outbreaks and can be transmitted from human to human. Its RNA-dependent RNA polymerase (RdRp) complex, consisting of the L and P proteins, carries out viral genome replication and transcription and is therefore an attractive drug target. Here, we report cryo-EM structures of the NiV polymerase complex in the apo and in an early elongation state with RNA and incoming substrate bound. The structure of the apo enzyme reveals the architecture of the NiV L-P complex, which shows a high degree of similarity to other nsNSV polymerase complexes. The structure of the RNA-bound NiV L-P complex shows how the enzyme interacts with template and product RNA during early RNA synthesis and how nucleoside triphosphates are bound in the active site. Comparisons show that RNA binding leads to rearrangements of key elements in the RdRp core and to ordering of the flexible C-terminal domains of NiV L required for RNA capping. Taken together, these results reveal the first structural snapshots of an actively elongating nsNSV L-P complex and provide insights into the mechanisms of genome replication and transcription by NiV and related viruses.
Collapse
Affiliation(s)
- Fernanda A Sala
- Department of Cellular Biochemistry, University Medical Center Göttingen, Göttingen, Germany
- Research Group Structure and Function of Molecular Machines, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Katja Ditter
- Department of Cellular Biochemistry, University Medical Center Göttingen, Göttingen, Germany
- Research Group Structure and Function of Molecular Machines, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Olexandr Dybkov
- Bioanalytical Mass Spectrometry Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Henning Urlaub
- Bioanalytical Mass Spectrometry Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Bioanalytics Group, Institute for Clinical Chemistry, 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
| | - Hauke S Hillen
- Department of Cellular Biochemistry, University Medical Center Göttingen, Göttingen, Germany.
- Research Group Structure and Function of Molecular Machines, 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.
- Göttingen Center for Molecular Biosciences (GZMB), Research Group Structure and Function of Molecular Machines, University of Göttingen, Göttingen, Germany.
| |
Collapse
|
43
|
Ruma YN, Nannenga BL, Gonen T. Unraveling atomic complexity from frozen samples. STRUCTURAL DYNAMICS (MELVILLE, N.Y.) 2025; 12:020901. [PMID: 40255534 PMCID: PMC12009148 DOI: 10.1063/4.0000303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Accepted: 03/26/2025] [Indexed: 04/22/2025]
Abstract
Cryo-electron microscopy (cryo-EM) is a significant driver of recent advances in structural biology. Cryo-EM is comprised of several distinct and complementary methods, which include single particle analysis, cryo-electron tomography, and microcrystal electron diffraction. In this Perspective, we will briefly discuss the different branches of cryo-EM in structural biology and the current challenges in these areas.
Collapse
Affiliation(s)
| | | | - Tamir Gonen
- Author to whom correspondence should be addressed:
| |
Collapse
|
44
|
Li Y, Song J, Mikusevic V, Marden JJ, Becerril A, Kuang H, Wang B, Rice WJ, Mindell JA, Wang DN. Substrate translocation and inhibition in human dicarboxylate transporter NaDC3. Nat Struct Mol Biol 2025; 32:502-512. [PMID: 39622972 DOI: 10.1038/s41594-024-01433-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 10/28/2024] [Indexed: 03/20/2025]
Abstract
The human high-affinity sodium-dicarboxylate cotransporter (NaDC3) imports various substrates into the cell as tricarboxylate acid cycle intermediates, lipid biosynthesis precursors and signaling molecules. Understanding the cellular signaling process and developing inhibitors require knowledge of the structural basis of the dicarboxylate specificity and inhibition mechanism of NaDC3. To this end, we determined the cryo-electron microscopy structures of NaDC3 in various dimers, revealing the protomer in three conformations: outward-open Co, outward-occluded Coo and inward-open Ci. A dicarboxylate is first bound and recognized in Co and how the substrate interacts with NaDC3 in Coo likely helps to further determine the substrate specificity. A phenylalanine from the scaffold domain interacts with the bound dicarboxylate in the Coo state and modulates the kinetic barrier to the transport domain movement. Structural comparison of an inhibitor-bound structure of NaDC3 to that of the sodium-dependent citrate transporter suggests ways for making an inhibitor that is specific for NaDC3.
Collapse
Affiliation(s)
- Yan Li
- Department of Cell Biology, New York University School of Medicine, New York, NY, USA
| | - Jinmei Song
- Department of Cell Biology, New York University School of Medicine, New York, NY, USA
| | - Vedrana Mikusevic
- Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Jennifer J Marden
- Department of Cell Biology, New York University School of Medicine, New York, NY, USA
| | - Alissa Becerril
- Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Huihui Kuang
- Cryo-Electron Microscopy Core, New York University School of Medicine, New York, NY, USA
| | - Bing Wang
- Cryo-Electron Microscopy Core, New York University School of Medicine, New York, NY, USA
| | - William J Rice
- Department of Cell Biology, New York University School of Medicine, New York, NY, USA
- Cryo-Electron Microscopy Core, New York University School of Medicine, New York, NY, USA
| | - Joseph A Mindell
- Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA.
| | - Da-Neng Wang
- Department of Cell Biology, New York University School of Medicine, New York, NY, USA.
| |
Collapse
|
45
|
Junglas B, Kartte D, Kutzner M, Hellmann N, Ritter I, Schneider D, Sachse C. Structural basis for Vipp1 membrane binding: from loose coats and carpets to ring and rod assemblies. Nat Struct Mol Biol 2025; 32:555-570. [PMID: 39379528 PMCID: PMC11919686 DOI: 10.1038/s41594-024-01399-z] [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: 11/14/2023] [Accepted: 09/05/2024] [Indexed: 10/10/2024]
Abstract
Vesicle-inducing protein in plastids 1 (Vipp1) is critical for thylakoid membrane biogenesis and maintenance. Although Vipp1 has recently been identified as a member of the endosomal sorting complexes required for transport III superfamily, it is still unknown how Vipp1 remodels membranes. Here, we present cryo-electron microscopy structures of Synechocystis Vipp1 interacting with membranes: seven structures of helical and stacked-ring assemblies at 5-7-Å resolution engulfing membranes and three carpet structures covering lipid vesicles at ~20-Å resolution using subtomogram averaging. By analyzing ten structures of N-terminally truncated Vipp1, we show that helix α0 is essential for membrane tubulation and forms the membrane-anchoring domain of Vipp1. Lastly, using a conformation-restrained Vipp1 mutant, we reduced the structural plasticity of Vipp1 and determined two structures of Vipp1 at 3.0-Å resolution, resolving the molecular details of membrane-anchoring and intersubunit contacts of helix α0. Our data reveal membrane curvature-dependent structural transitions from carpets to rings and rods, some of which are capable of inducing and/or stabilizing high local membrane curvature triggering membrane fusion.
Collapse
Affiliation(s)
- Benedikt Junglas
- Ernst-Ruska Centre for Microscopy and Spectroscopy with Electrons, ER-C-3/Structural Biology, Forschungszentrum Jülich, Jülich, Germany
| | - David Kartte
- Ernst-Ruska Centre for Microscopy and Spectroscopy with Electrons, ER-C-3/Structural Biology, Forschungszentrum Jülich, Jülich, Germany
- Department of Biology, Heinrich Heine University, Düsseldorf, Germany
| | - Mirka Kutzner
- Department of Chemistry, Biochemistry, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Nadja Hellmann
- Department of Chemistry, Biochemistry, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Ilona Ritter
- Ernst-Ruska Centre for Microscopy and Spectroscopy with Electrons, ER-C-3/Structural Biology, Forschungszentrum Jülich, Jülich, Germany
| | - Dirk Schneider
- Department of Chemistry, Biochemistry, Johannes Gutenberg University Mainz, Mainz, Germany
- Institute of Molecular Physiology, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Carsten Sachse
- Ernst-Ruska Centre for Microscopy and Spectroscopy with Electrons, ER-C-3/Structural Biology, Forschungszentrum Jülich, Jülich, Germany.
- Department of Biology, Heinrich Heine University, Düsseldorf, Germany.
| |
Collapse
|
46
|
Gao Y, Wang L, Doeswijk T, Winblad B, Schedin‐Weiss S, Tjernberg LO. Intraneuronal Aβ accumulation causes tau hyperphosphorylation via endolysosomal leakage. Alzheimers Dement 2025; 21:e70091. [PMID: 40145397 PMCID: PMC11947758 DOI: 10.1002/alz.70091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 02/14/2025] [Accepted: 02/16/2025] [Indexed: 03/28/2025]
Abstract
INTRODUCTION Alzheimer's disease (AD) is characterized by amyloid beta (Aβ) peptide plaques and intracellular neurofibrillary tangles formed by hyperphosphorylated tau. Many attempts have been made to clarify the link between Aβ and tau in the pathogenesis, but conclusive data describing a pathway for this connection are still lacking. METHODS We developed a neuronal model of Aβ-induced toxicity and studied downstream effects of intraneuronal Aβ42 accumulation on tau hyperphosphorylation using confocal microscopy and live cell imaging. RESULTS Aβ42 added to the medium was endocytosed into neurons, inducing the formation of endolysosomal protofibrils and endolysosomal leakage, which in turn promoted tau hyperphosphorylation. Asparaginyl endopeptidase (AEP) was released from the disrupted lysosomes, and inhibition of this peptidase activity reduced tau hyperphosphorylation. DISCUSSION The data suggest a mechanism of AD in which Aβ42 accumulates and aggregates gradually in neurons over time, leading to endolysosomal leakage and release of AEP, which subsequently triggers tau hyperphosphorylation. HIGHLIGHTS Aβ42 endocytosis leads to its endolysosomal accumulation in neurons over time. Aβ42 polymerizes into protofibrils and causes endolysosomal leakage. Tau hyperphosphorylation is induced by endolysosomal asparagine endopeptidase leakage. Tau hyperphosphorylation is inhibited by an asparagine endopeptidase inhibitor.
Collapse
Affiliation(s)
- Yang Gao
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and SocietyKarolinska InstitutetSolnaSweden
| | - Lisha Wang
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and SocietyKarolinska InstitutetSolnaSweden
| | - Tosca Doeswijk
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and SocietyKarolinska InstitutetSolnaSweden
| | - Bengt Winblad
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and SocietyKarolinska InstitutetSolnaSweden
- Theme Inflammation and AgingKarolinska University HospitalHuddingeSweden
| | - Sophia Schedin‐Weiss
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and SocietyKarolinska InstitutetSolnaSweden
| | - Lars O. Tjernberg
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and SocietyKarolinska InstitutetSolnaSweden
| |
Collapse
|
47
|
Woldeyes RA, Nishiga M, Roest ASV, Engel L, Giri P, Montenegro GC, Dunn AR, Spudich JA, Bernstein D, Schmid MF, Wu JC, Chiu W. Structure of the Thin Filament in Human iPSC-derived Cardiomyocytes and its Response to Heart Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.10.26.564098. [PMID: 37961228 PMCID: PMC10634850 DOI: 10.1101/2023.10.26.564098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Cardiovascular diseases are a leading cause of death worldwide, but our understanding of the underlying mechanisms is limited, in part because of the complexity of the cellular machinery that controls the heart muscle contraction cycle. Cryogenic electron tomography (cryo-ET) provides a way to visualize diverse cellular machinery while preserving contextual information like subcellular localization and transient complex formation, but this approach has not been widely applied to the study of heart muscle cells (cardiomyocytes). Here, we deploy an optimized cryo-ET platform that enables cellular-structural biology in human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). Using this platform, we reconstructed sub-nanometer resolution structures of the human cardiac muscle thin filament, a central component of the contractile machinery. Reconstructing the troponin complex, a regulatory component of the thin filament, from within cells, we identified previously unobserved conformations that highlight the structural flexibility of this regulatory complex. We next measured the impact of chemical and genetic perturbations associated with cardiovascular disease on the structure of troponin. In both cases, we found changes in troponin structure that are consistent with known disease phenotypes-highlighting the value of our approach for dissecting complex disease mechanisms in the cellular context.
Collapse
Affiliation(s)
- Rahel A. Woldeyes
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Masataka Nishiga
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Alison S. Vander Roest
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Engineering, University of Michigan, MI, USA
| | - Leeya Engel
- Faculty of Mechanical Engineering, Technion - Israel Institute of Technology, Haifa, Israel
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
| | - Prerna Giri
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Alexander R. Dunn
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
| | - James A. Spudich
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
| | - Daniel Bernstein
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael F. Schmid
- Department of Bioengineering, Stanford University, Stanford, CA, USA
- Division of Cryo-EM and Bioimaging, SSRL, SLAC National Accelerator Laboratory, Stanford University, Menlo Park, CA, USA
| | - Joseph C. Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Wah Chiu
- Department of Bioengineering, Stanford University, Stanford, CA, USA
- Division of Cryo-EM and Bioimaging, SSRL, SLAC National Accelerator Laboratory, Stanford University, Menlo Park, CA, USA
| |
Collapse
|
48
|
Sheokand PK, James AM, Jenkins B, K. Lysyganicz P, Lacabanne D, King MS, Kunji ERS, Siniossoglou S, Koulman A, Murphy MP, Petkevicius K. TRAM-LAG1-CLN8 family proteins are acyltransferases regulating phospholipid composition. SCIENCE ADVANCES 2025; 11:eadr3723. [PMID: 39970228 PMCID: PMC11838012 DOI: 10.1126/sciadv.adr3723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 01/15/2025] [Indexed: 02/21/2025]
Abstract
The diversity of cellular phospholipids, crucial for membrane homeostasis and function, arises from enzymatic remodeling of their fatty acyl chains. In this work, we reveal that poorly understood TRAM-LAG1-CLN8 domain (TLCD)-containing proteins are phospholipid remodeling enzymes. We demonstrate that TLCD1 is an evolutionarily conserved lysophosphatidylethanolamine acyltransferase, which regulates cellular phospholipid composition and generates previously undescribed fatty acid and thiamine (vitamin B1) esters as its secondary products. Furthermore, we establish that human TLCD protein CLN8, mutations of which cause fatal neurodegenerative Batten disease, is a lysophosphatidylglycerol acyltransferase. We show that CLN8 catalyzes the essential step in the biosynthesis of bis(monoacylglycero)phosphate, a phospholipid critical for lysosome function. Our study unveils a family of acyltransferases integral to cellular membrane phospholipid homeostasis and human disease.
Collapse
Affiliation(s)
- Pradeep K. Sheokand
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Andrew M. James
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Benjamin Jenkins
- Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Pawel K. Lysyganicz
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Denis Lacabanne
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Martin S. King
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Edmund R. S. Kunji
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Symeon Siniossoglou
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Albert Koulman
- Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Michael P. Murphy
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Kasparas Petkevicius
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| |
Collapse
|
49
|
Liu C, Park YJ, Ma CB, Stuart C, Gen R, Sun YC, Yang X, Lin MY, Xiong Q, Si JY, Liu P, Veesler D, Yan H. ACE2 utilization of HKU25 clade MERS-related coronaviruses with broad geographic distribution. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.19.639017. [PMID: 40027745 PMCID: PMC11870458 DOI: 10.1101/2025.02.19.639017] [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: 03/05/2025]
Abstract
Dipeptidyl peptidase-4 (DPP4) is a well-established receptor for several MERS-related coronaviruses (MERSr-CoVs) isolated from humans, camels, pangolins, and bats (1-6). However, the receptor usage of many genetically diverse bat MERSr-CoVs with broad geographical distributions remains poorly understood. Recent studies have identified angiotensin-converting enzyme 2 (ACE2) as an entry receptor for multiple merbecovirus clades. Here, using viral antigen and pseudovirus-based functional assays, we demonstrate that several bat merbecoviruses from the HKU25 clade previously thought to utilize DPP4 (7), employ ACE2 as their functional receptor. Cryo-electron microscopy analysis revealed that HsItaly2011 and VsCoV-a7 recognize ACE2 with a binding mode sharing similarity with that of HKU5 but involving remodeled interfaces and distinct ortholog selectivity, suggesting a common evolutionary origin of ACE2 utilization for these two clades of viruses. EjCoV-3, a strain closely related to the DPP4-using MERSr-CoV BtCoV-422, exhibited relatively broad ACE2 ortholog tropism and could utilize human ACE2 albeit suboptimally. Despite differences in entry mechanisms and spike proteolytic activation compared to MERS-CoV, these viruses remain sensitive to several broadly neutralizing antibodies and entry inhibitors. These findings redefine our understanding of the evolution of receptor usage among MERSr-CoVs and highlight the versatility of ACE2 as a functional receptor for diverse coronaviruses.
Collapse
Affiliation(s)
- Chen Liu
- State Key Laboratory of Virology and Biosafety, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University; Wuhan, Hubei, 430072, China
| | - Young-Jun Park
- Department of Biochemistry, University of Washington; Seattle, WA 98195, USA
- Howard Hughes Medical Institute, University of Washington; Seattle, WA 98195, USA
| | - Cheng-Bao Ma
- State Key Laboratory of Virology and Biosafety, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University; Wuhan, Hubei, 430072, China
| | - Cameron Stuart
- Department of Biochemistry, University of Washington; Seattle, WA 98195, USA
| | - Risako Gen
- Department of Biochemistry, University of Washington; Seattle, WA 98195, USA
| | - Yu-Cheng Sun
- State Key Laboratory of Virology and Biosafety, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University; Wuhan, Hubei, 430072, China
| | - Xiao Yang
- State Key Laboratory of Virology and Biosafety, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University; Wuhan, Hubei, 430072, China
| | - Mei-Yi Lin
- State Key Laboratory of Virology and Biosafety, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University; Wuhan, Hubei, 430072, China
| | - Qing Xiong
- State Key Laboratory of Virology and Biosafety, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University; Wuhan, Hubei, 430072, China
| | - Jun-Yu Si
- State Key Laboratory of Virology and Biosafety, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University; Wuhan, Hubei, 430072, China
| | - Peng Liu
- State Key Laboratory of Virology and Biosafety, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University; Wuhan, Hubei, 430072, China
| | - David Veesler
- Department of Biochemistry, University of Washington; Seattle, WA 98195, USA
- Howard Hughes Medical Institute, University of Washington; Seattle, WA 98195, USA
| | - Huan Yan
- State Key Laboratory of Virology and Biosafety, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University; Wuhan, Hubei, 430072, China
| |
Collapse
|
50
|
Li Q, Zhang L, Xu Q, Zhang P, Zhu S. SLICK: A Sandwich-LIke Culturing Kit for in situ Cryo-ET Sample Preparation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.14.638381. [PMID: 39990359 PMCID: PMC11844457 DOI: 10.1101/2025.02.14.638381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
In situ cryo-electron tomography (cryo-ET) has recently been widely used in observing subcellular structures and macromolecules in their native states at high resolution. One of the reasons that it has not been more widely adopted by cell biologists and structural biologists is the difficulties in sample preparation. Here we present the Sandwich-LIke Culturing Kit (SLICK), simplifying the procedure and increasing the throughput for sample preparation for in situ cryo-ET (69 words).
Collapse
|