1
|
Hsiao LC, Lee CH, Mazmanian K, Yoshida M, Ito G, Murata T, Utsunomiya-Tate N, Haino T, Tate SI, Hsu STD. Impacts of D-aspartate on the Aggregation Kinetics and Structural Polymorphism of Amyloid β Peptide 1-42. J Mol Biol 2025; 437:169092. [PMID: 40090459 DOI: 10.1016/j.jmb.2025.169092] [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: 11/12/2024] [Revised: 03/10/2025] [Accepted: 03/11/2025] [Indexed: 03/18/2025]
Abstract
Isomerization of L-Aspartate (L-Asp) into D-aspartate (D-Asp) occurs naturally in proteins at a rate that is much faster than that of other amino acid types. Accumulation of D-Asp is age-dependent, which could alter protein structures and, therefore, functions. Site-specific introduction of D-Asp can accelerate aggregation kinetics of a variety of proteins associated with misfolding diseases. Here, we showed by thioflavin T fluorescence that the isomerization of L-Asp at different positions of amyloid β peptide 1-42 (Aβ42) generates opposing effects on its aggregation kinetics. We further determined the atomic structures of Aβ42 amyloid fibrils harboring a single D-Asp at position 23 and two D-Asp at positions 7 and 23 by cryo-electron microscopy helical reconstruction - cross-validated by cryo-electron tomography and atomic force microscopy - to reveal how D-Asp7 contributes to the formation of a unique triple stranded amyloid fibril structure stabilized by two threads of well-ordered water molecules. These findings provide crucial insights into how the conversion from L- to D-Asp influences the aggregation propensity and amyloid polymorphism of Aβ42.
Collapse
Affiliation(s)
- Li-Ching Hsiao
- Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan; Institute of Biochemical Sciences, National Taiwan University, Taipei 10617, Taiwan
| | - Chih-Hsuan Lee
- Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan
| | - Karine Mazmanian
- Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan
| | - Masaya Yoshida
- Department of Chemistry, Graduate School of Advanced Science and Engineering, Hiroshima University, 1-3-1 Kagamiyama, Higashi-Hiroshima, Japan; International Institute for Sustainability with Knotted Chiral Meta Matter (WPI-SKCM(2)), Hiroshima University, 1-3-1 Kagamiyama, Higashi-Hiroshima, Hiroshima 739-8526, Japan
| | - Genta Ito
- Department of Biomolecular Chemistry, Faculty of Pharma-Sciences, Teikyo University, 2-11-1 Kaga, Itabashi-ku, Tokyo 173-8605, Japan
| | - Takuya Murata
- Department of Biomolecular Chemistry, Faculty of Pharma-Sciences, Teikyo University, 2-11-1 Kaga, Itabashi-ku, Tokyo 173-8605, Japan
| | - Naoko Utsunomiya-Tate
- Department of Biomolecular Chemistry, Faculty of Pharma-Sciences, Teikyo University, 2-11-1 Kaga, Itabashi-ku, Tokyo 173-8605, Japan
| | - Takeharu Haino
- Department of Chemistry, Graduate School of Advanced Science and Engineering, Hiroshima University, 1-3-1 Kagamiyama, Higashi-Hiroshima, Japan; International Institute for Sustainability with Knotted Chiral Meta Matter (WPI-SKCM(2)), Hiroshima University, 1-3-1 Kagamiyama, Higashi-Hiroshima, Hiroshima 739-8526, Japan
| | - Shih-Ichi Tate
- International Institute for Sustainability with Knotted Chiral Meta Matter (WPI-SKCM(2)), Hiroshima University, 1-3-1 Kagamiyama, Higashi-Hiroshima, Hiroshima 739-8526, Japan; Department of Mathematical and Life Sciences, Graduate School of Integrated Sciences for Life, Hiroshima University, 1-3-1 Kagamiyama, Higashi-Hiroshima, Hiroshima 739-8526, Japan; Research Center for the Mathematics on Chromatin Live Dynamics (RcMcD), Hiroshima University, 1-3-1 Kagamiyama, Higashi-Hiroshima, Hiroshima 739-8526, Japan; Meiji Institute for Advanced Study of Mathematical Sciences (MIMS), Organization for the Strategic Coordination of Research and Intellectual Properties, Meiji University, 4-21-1 Nakano, Nakano-ku, Tokyo 164-8525, Japan
| | - Shang-Te Danny Hsu
- Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan; Institute of Biochemical Sciences, National Taiwan University, Taipei 10617, Taiwan; International Institute for Sustainability with Knotted Chiral Meta Matter (WPI-SKCM(2)), Hiroshima University, 1-3-1 Kagamiyama, Higashi-Hiroshima, Hiroshima 739-8526, Japan.
| |
Collapse
|
2
|
Liu Y, Wang K, Wang W, Kashyap S, Jih J, Imani A, Hsiai T, Zhou ZH. Demonstration and structural basis of a therapeutic DNA aptamer for SARS-CoV-2 spike protein detection. Biosens Bioelectron 2025; 287:117691. [PMID: 40516427 DOI: 10.1016/j.bios.2025.117691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 05/23/2025] [Accepted: 06/11/2025] [Indexed: 06/16/2025]
Abstract
At the onset of the COVID-19 pandemic, the absence of rapid and precise diagnostic tools hindered early detection and response. To address this challenge, we developed a renewable electrochemical impedance biosensor (aptasensor) using a therapeutic DNA aptamer immobilized on a nanostructured gold nanoparticle/carbon nanotube (AuNP/CNT) electrode to detect the SARS-CoV-2 spike (S) protein receptor-binding domain (RBD). The aptasensor achieved a limit of detection of 0.19 pg mL-1 and a dynamic range from 1 to 105 pg mL-1. Following regeneration with a 60-s pH 2.0 rinse, the sensor retained over 90% of its original signal across five cycles and remained stable after two weeks of ambient storage. Dual-mode readouts, utilizing impedance spectroscopy and surface plasmon resonance (SPR), confirmed binding specificity and reproducibility. Cryogenic electron microscopy (cryoEM) resolved the aptamer-S protein complex in the open conformation, revealing a bridge-like interaction with conserved residues Y489, N487, F486, and S477. These contacts remained functional despite Omicron BA.2 mutations (S477N, N501Y) and aligned with previously reported mutational data. Specificity was further supported by negative controls and structural consistency with known hACE2 binding footprints. These results establish a robust, low-cost biosensor platform combining reuse, structural insight, and variant tolerance. The aptasensor's scalability and adaptability make it a strong candidate for future diagnostic applications targeting evolving viral threats.
Collapse
Affiliation(s)
- Yujun Liu
- California NanoSystems Institute, University of California at Los Angeles, Los Angeles, 90095, California, USA; Department of Microbiology, Immunology and Molecular Genetics, University of California at Los Angeles, Los Angeles, 90095, California, USA
| | - Kaidong Wang
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA; Department of Medicine, Greater Los Angeles VA Healthcare System, Los Angeles, CA, 90073, USA; Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Weiguang Wang
- California NanoSystems Institute, University of California at Los Angeles, Los Angeles, 90095, California, USA; Department of Microbiology, Immunology and Molecular Genetics, University of California at Los Angeles, Los Angeles, 90095, California, USA
| | - Saarang Kashyap
- California NanoSystems Institute, University of California at Los Angeles, Los Angeles, 90095, California, USA; Department of Microbiology, Immunology and Molecular Genetics, University of California at Los Angeles, Los Angeles, 90095, California, USA
| | - Jonathan Jih
- California NanoSystems Institute, University of California at Los Angeles, Los Angeles, 90095, California, USA; Department of Microbiology, Immunology and Molecular Genetics, University of California at Los Angeles, Los Angeles, 90095, California, USA; Molecular Biology Institute, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA
| | - Anthony Imani
- California NanoSystems Institute, University of California at Los Angeles, Los Angeles, 90095, California, USA; Department of Microbiology, Immunology and Molecular Genetics, University of California at Los Angeles, Los Angeles, 90095, California, USA
| | - Tzung Hsiai
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA; Department of Medicine, Greater Los Angeles VA Healthcare System, Los Angeles, CA, 90073, USA; Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California Los Angeles, Los Angeles, CA, 90095, USA.
| | - Z Hong Zhou
- California NanoSystems Institute, University of California at Los Angeles, Los Angeles, 90095, California, USA; Department of Microbiology, Immunology and Molecular Genetics, University of California at Los Angeles, Los Angeles, 90095, California, USA; Molecular Biology Institute, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA.
| |
Collapse
|
3
|
Malewana RD, Stalls V, May A, Lu X, Martinez DR, Schäfer A, Li D, Barr M, Sutherland LL, Lee E, Parks R, Beck WE, Newman A, Bock KW, Minai M, Nagata BM, DeMarco CT, Denny TN, Oguin TH, Rountree W, Wang Y, Mansouri K, Edwards RJ, Smith L, Sempowski GD, Eaton A, Muramatsu H, Henderson R, Tam Y, Barbosa C, Tang J, Cain DW, Santra S, Moore IN, Andersen H, Lewis MG, Golding H, Seder R, Khurana S, Montefiori DC, Pardi N, Weissman D, Baric RS, Acharya P, Haynes BF, Saunders KO. Nonstabilized SARS-CoV-2 spike mRNA vaccination induces broadly neutralizing antibodies in nonhuman primates. Sci Transl Med 2025; 17:eadn5651. [PMID: 40498855 DOI: 10.1126/scitranslmed.adn5651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 04/06/2025] [Indexed: 06/18/2025]
Abstract
Immunization with messenger RNA (mRNA) or viral vectors encoding spike protein with diproline substitutions (S-2P) were shown to provide protective immunity, curbing the COVID-19 pandemic. However, in light of the emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants of concern (VOCs) that can cause COVID-19, it is essential that we understand how immunization with spike protein elicits neutralizing antibodies (nAbs). Here, we compared immunization of macaques with mRNA vaccines expressing ancestral spike protein with or without diproline substitutions, showing that the diproline substitutions were not required for protection against SARS-CoV-2 challenge or induction of broadly neutralizing B cell lineages. One group of nAbs elicited by the ancestral spike protein lacking diproline substitutions targeted the outer face of the receptor binding domain (RBD), neutralized all tested SARS-CoV-2 VOC pseudotyped viruses including Omicron XBB.1.5 in vitro, but lacked cross-sarbecovirus neutralization. Structural analysis showed that the macaque nAbs that could broadly neutralize VOCs bound to the same epitope as a human nAb, DH1193. In contrast, vaccine-induced antibodies that targeted the RBD inner face neutralized multiple sarbecoviruses, protected mice from bat CoV RsSHC014 challenge, but lacked Omicron variant neutralization. Thus, ancestral SARS-CoV-2 spike mRNA vaccines lacking proline substitutions can induce B cell lineages binding to distinct RBD sites that either broadly neutralize animal and human sarbecoviruses or neutralize recent Omicron VOCs. Thus, the use of a nonstabilized spike protein design in some COVID-19 vaccines does not preclude the elicitation of broad sarbecovirus and broad VOC nAbs.
Collapse
Affiliation(s)
- R Dilshan Malewana
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Victoria Stalls
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Aaron May
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Biochemistry, Duke University School of Medicine, Durham, NC 27710, USA
| | - Xiaozhi Lu
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - David R Martinez
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Immunobiology, Yale Center for Infection and Immunity, Yale School of Medicine, New Haven, CT 06510, USA
| | - Alexandra Schäfer
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Dapeng Li
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Maggie Barr
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Laura L Sutherland
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Esther Lee
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Robert Parks
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Whitney Edwards Beck
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Amanda Newman
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Kevin W Bock
- Infectious Disease Pathogenesis Section, Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20814, USA
| | - Mahnaz Minai
- Infectious Disease Pathogenesis Section, Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20814, USA
| | - Bianca M Nagata
- Infectious Disease Pathogenesis Section, Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20814, USA
| | - C Todd DeMarco
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Thomas N Denny
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Thomas H Oguin
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Wes Rountree
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Yunfei Wang
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Katayoun Mansouri
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Robert J Edwards
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Lena Smith
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Gregory D Sempowski
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Amanda Eaton
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Hiromi Muramatsu
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rory Henderson
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Ying Tam
- Acuitas Therapeutics LLC, Vancouver, BC V6T 1Z3, Canada
| | | | - Juanjie Tang
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), Food and Drug Administration, Silver Spring, MD 20871, USA
| | - Derek W Cain
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Sampa Santra
- Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Ian N Moore
- Infectious Disease Pathogenesis Section, Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20814, USA
| | | | | | - Hana Golding
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), Food and Drug Administration, Silver Spring, MD 20871, USA
| | - Robert Seder
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20814, USA
| | - Surender Khurana
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), Food and Drug Administration, Silver Spring, MD 20871, USA
| | - David C Montefiori
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA
| | - Norbert Pardi
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Drew Weissman
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ralph S Baric
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Priyamvada Acharya
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Biochemistry, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA
| | - Barton F Haynes
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Kevin O Saunders
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
4
|
Kizziah JL, Mukherjee A, Parker LK, Dokland T. Structure of the Staphylococcus aureus bacteriophage 80α neck shows details of the DNA, tail completion protein, and tape measure protein. Structure 2025; 33:1063-1073.e2. [PMID: 40174589 PMCID: PMC12145261 DOI: 10.1016/j.str.2025.03.007] [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: 01/14/2025] [Revised: 02/21/2025] [Accepted: 03/06/2025] [Indexed: 04/04/2025]
Abstract
The Staphylococcus aureus pathogenicity islands (SaPIs), including SaPI1, are a type of mobile genetic elements (MGEs) that are mobilized at high frequency by "helper" bacteriophages, such as 80α, leading to packaging of the SaPI genomes into virions made from helper-encoded structural proteins. 80α and SaPI1 virions consist of an icosahedral head connected via a portal vertex to a long, non-contractile tail. A connector or "neck" forms the interface between the tail and the head. Here, we have determined the high-resolution structure of the neck section of SaPI1 virions, including the dodecameric portal and head-tail-connector proteins, and the hexameric head-tail joining, tail terminator and major tail proteins. We also resolved the DNA, the tail completion protein (TCP), and the tape measure protein (TMP) inside the tail, features that have not previously been observed at high resolution. Our study provides insights into the assembly and infection process in this important group of MGEs.
Collapse
Affiliation(s)
- James L Kizziah
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Amarshi Mukherjee
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Laura K Parker
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Terje Dokland
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
5
|
Carr KD, Zambrano DED, Weidle C, Goodson A, Eisenach HE, Pyles H, Courbet A, King NP, Borst AJ. Protein identification using Cryo-EM and artificial intelligence guides improved sample purification. J Struct Biol X 2025; 11:100120. [PMID: 39958810 PMCID: PMC11830286 DOI: 10.1016/j.yjsbx.2025.100120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 01/14/2025] [Accepted: 01/17/2025] [Indexed: 02/18/2025] Open
Abstract
Protein purification is essential in protein biochemistry, structural biology, and protein design, enabling the determination of protein structures, the study of biological mechanisms, and the characterization of both natural and de novo designed proteins. However, standard purification strategies often encounter challenges, such as unintended co-purification of contaminants alongside the target protein. This issue is particularly problematic for self-assembling protein nanomaterials, where unexpected geometries may reflect novel assembly states, cross-contamination, or native proteins originating from the expression host. Here, we used an automated structure-to-sequence pipeline to first identify an unknown co-purifying protein found in several purified designed protein samples. By integrating cryo-electron microscopy (Cryo-EM), ModelAngelo's sequence-agnostic model-building, and Protein BLAST, we identified the contaminant as dihydrolipoamide succinyltransferase (DLST). This identification was validated through comparisons with DLST structures in the Protein Data Bank, AlphaFold 3 predictions based on the DLST sequence from our E. coli expression vector, and traditional biochemical methods. The identification informed subsequent modifications to our purification protocol, which successfully excluded DLST from future preparations. To explore the potential broader utility of this approach, we benchmarked four computational methods for DLST identification across varying resolution ranges. This study demonstrates the successful application of a structure-to-sequence protein identification workflow, integrating Cryo-EM, ModelAngelo, Protein BLAST, and AlphaFold 3 predictions, to identify and ultimately help guide the removal of DLST from sample purification efforts. It highlights the potential of combining Cryo-EM with AI-driven tools for accurate protein identification and addressing purification challenges across diverse contexts in protein science.
Collapse
Affiliation(s)
- Kenneth D. Carr
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Dane Evan D. Zambrano
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Connor Weidle
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Alex Goodson
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Helen E. Eisenach
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Harley Pyles
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Alexis Courbet
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Neil P. King
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Andrew J. Borst
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
6
|
Zerio CJ, Bai Y, Sosa-Alvarado BA, Guzi T, Lander GC. Human polymerase θ helicase positions DNA microhomologies for double-strand break repair. Nat Struct Mol Biol 2025; 32:1061-1068. [PMID: 40021738 DOI: 10.1038/s41594-025-01514-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 02/10/2025] [Indexed: 03/03/2025]
Abstract
DNA double-strand breaks occur daily in all human cells and must be repaired with high fidelity to minimize genomic instability. Deficiencies in high-fidelity DNA repair by homologous recombination lead to dependence on DNA polymerase θ, which identifies DNA microhomologies in 3' single-stranded DNA overhangs and anneals them to initiate error-prone double-strand break repair. The resulting genomic instability is associated with numerous cancers, thereby making this polymerase an attractive therapeutic target. However, despite the biomedical importance of polymerase θ, the molecular details of how it initiates DNA break repair remain unclear. Here, we present cryo-electron microscopy structures of the polymerase θ helicase domain bound to microhomology-containing DNA, revealing DNA-induced rearrangements of the helicase that enable DNA repair. Our structures show that DNA-bound helicase dimers facilitate a microhomology search that positions 3' single-stranded DNA ends in proximity to align complementary bases and anneal DNA microhomology. We characterize the molecular determinants that enable the helicase domain of polymerase θ to identify and pair DNA microhomologies to initiate mutagenic DNA repair, thereby providing insight into potentially targetable interactions for therapeutic interventions.
Collapse
Affiliation(s)
- Christopher J Zerio
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, CA, USA
| | | | | | | | - Gabriel C Lander
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, CA, USA.
| |
Collapse
|
7
|
Roth P, Fotiadis D. Cryo-EM structure of a phosphotransferase system glucose transporter stalled in an intermediate conformation. J Struct Biol X 2025; 11:100124. [PMID: 40124667 PMCID: PMC11930441 DOI: 10.1016/j.yjsbx.2025.100124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 02/22/2025] [Accepted: 03/03/2025] [Indexed: 03/25/2025] Open
Abstract
The phosphotransferase system glucose-specific transporter IICBGlc serves as a central nutrient uptake system in bacteria. It transports glucose across the plasma membrane via the IICGlc domain and phosphorylates the substrate within the cell to produce the glycolytic intermediate, glucose-6-phosphate, through the IIBGlc domain. Furthermore, IICGlc consists of a transport (TD) and a scaffold domain, with the latter being involved in dimer formation. Transport is mediated by an elevator-type mechanism within the IICGlc domain, where the substrate binds to the mobile TD. This domain undergoes a large-scale rigid-body movement relative to the static scaffold domain, translocating glucose across the membrane. Structures of elevator-type transporters are typically captured in either inward- or outward-facing conformations. Intermediate states remain elusive, awaiting structural determination and mechanistic interpretation. Here, we present a single-particle cryo-EM structure of purified, n-dodecyl-β-D-maltopyranoside-solubilized IICBGlc from Escherichia coli. While the IIBGlc protein domain is flexible remaining unresolved, the dimeric IICGlc transporter is found trapped in a hitherto unobserved intermediate conformational state. Specifically, the TD is located halfway between inward- and outward-facing states. Structural analysis revealed a specific n-dodecyl-β-D-maltopyranoside molecule bound to the glucose binding site. The sliding of the TD is potentially impeded halfway due to the bulky nature of the ligand and a shift of the thin gate, thereby stalling the transporter. In conclusion, this study presents a novel conformational state of IICGlc, and provides new structural and mechanistic insights into a potential stalling mechanism, paving the way for the rational design of transport inhibitors targeting this critical bacterial metabolic process.
Collapse
Affiliation(s)
- Patrick Roth
- Institute of Biochemistry and Molecular Medicine, Medical Faculty, University of Bern, Bern, Switzerland
| | - Dimitrios Fotiadis
- Institute of Biochemistry and Molecular Medicine, Medical Faculty, University of Bern, Bern, Switzerland
| |
Collapse
|
8
|
Introini B, Hahn A, Kühlbrandt W. Cryo-EM structure of the NDH-PSI-LHCI supercomplex from Spinacia oleracea. Nat Struct Mol Biol 2025; 32:968-978. [PMID: 39856350 DOI: 10.1038/s41594-024-01478-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 12/17/2024] [Indexed: 01/27/2025]
Abstract
The nicotinamide adenine dinucleotide phosphate (NADPH) dehydrogenase (NDH) complex is crucial for photosynthetic cyclic electron flow and respiration, transferring electrons from ferredoxin to plastoquinone while transporting H+ across the chloroplast membrane. This process boosts adenosine triphosphate production, regardless of NADPH levels. In flowering plants, NDH forms a supercomplex with photosystem I, enhancing its stability under high light. We report the cryo-electron microscopy structure of the NDH supercomplex in Spinacia oleracea at a resolution of 3.0-3.3 Å. The supercomplex consists of 41 protein subunits, 154 chlorophylls and 38 carotenoids. Subunit interactions are reinforced by 46 distinct lipids. The structure of NDH resembles that of mitochondrial complex I closely, including the quinol-binding site and an extensive internal aqueous passage for proton translocation. A well-resolved catalytic plastoquinone (PQ) occupies the PQ channel. The pronounced structural similarity to complex I sheds light on electron transfer and proton translocation within the NDH supercomplex.
Collapse
Affiliation(s)
- Bianca Introini
- Department of Structural Biology, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
| | - Alexander Hahn
- Department of Structural Biology, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
- MVZ am Helios Klinikum, Emil von Behring GmbH, Institut für Gewebediagnostik/Pathologie, Berlin, Germany
| | - Werner Kühlbrandt
- Department of Structural Biology, Max Planck Institute of Biophysics, Frankfurt am Main, Germany.
| |
Collapse
|
9
|
Klebl DP, McMillan SN, Risi C, Forgacs E, Virok B, Atherton JL, Harris SA, Stofella M, Winkelmann DA, Sobott F, Galkin VE, Knight PJ, Muench SP, Scarff CA, White HD. Swinging lever mechanism of myosin directly shown by time-resolved cryo-EM. Nature 2025; 642:519-526. [PMID: 40205053 PMCID: PMC12158783 DOI: 10.1038/s41586-025-08876-5] [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: 01/05/2024] [Accepted: 03/07/2025] [Indexed: 04/11/2025]
Abstract
Myosins produce force and movement in cells through interactions with F-actin1. Generation of movement is thought to arise through actin-catalysed conversion of myosin from an ATP-generated primed (pre-powerstroke) state to a post-powerstroke state, accompanied by myosin lever swing2,3. However, the initial, primed actomyosin state has never been observed, and the mechanism by which actin catalyses myosin ATPase activity is unclear. Here, to address these issues, we performed time-resolved cryogenic electron microscopy (cryo-EM)4 of a myosin-5 mutant having slow hydrolysis product release5,6. Primed actomyosin was predominantly captured 10 ms after mixing primed myosin with F-actin, whereas post-powerstroke actomyosin predominated at 120 ms, with no abundant intermediate states detected. For detailed interpretation, cryo-EM maps were fitted with pseudo-atomic models. Small but critical changes accompany the primed motor binding to actin through its lower 50-kDa subdomain, with the actin-binding cleft open and phosphate release prohibited. Amino-terminal actin interactions with myosin promote rotation of the upper 50-kDa subdomain, closing the actin-binding cleft, and enabling phosphate release. The formation of interactions between the upper 50-kDa subdomain and actin creates the strong-binding interface needed for effective force production. The myosin-5 lever swings through 93°, predominantly along the actin axis, with little twisting. The magnitude of lever swing matches the typical step length of myosin-5 along actin7. These time-resolved structures demonstrate the swinging lever mechanism, elucidate structural transitions of the power stroke, and resolve decades of conjecture on how myosins generate movement.
Collapse
Affiliation(s)
- David P Klebl
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, UK
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK
- Department of Cell and Virus Structure, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Sean N McMillan
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Cristina Risi
- Department of Biomedical and Translational Sciences, Macon & Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, VA, USA
| | - Eva Forgacs
- Department of Biomedical and Translational Sciences, Macon & Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, VA, USA
| | - Betty Virok
- Department of Biomedical and Translational Sciences, Macon & Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, VA, USA
| | - Jennifer L Atherton
- Department of Biomedical and Translational Sciences, Macon & Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, VA, USA
| | - Sarah A Harris
- School of Mathematical and Physical Sciences, University of Sheffield, Sheffield, UK
| | - Michele Stofella
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Donald A Winkelmann
- Department of Pathology & Laboratory Medicine, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, USA
| | - Frank Sobott
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Vitold E Galkin
- Department of Biomedical and Translational Sciences, Macon & Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, VA, USA
| | - Peter J Knight
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Stephen P Muench
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, UK.
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK.
| | - Charlotte A Scarff
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK.
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK.
| | - Howard D White
- Department of Biomedical and Translational Sciences, Macon & Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, VA, USA.
| |
Collapse
|
10
|
Karlsson E, Andén O, Fan C, Fourati Z, Haouz A, Zhuang Y, Howard RJ, Delarue M, Lindahl E. Vestibular modulation by stimulant derivatives in a pentameric ligand-gated ion channel. Br J Pharmacol 2025; 182:2790-2802. [PMID: 40065647 DOI: 10.1111/bph.70011] [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: 05/24/2024] [Revised: 10/23/2024] [Accepted: 11/20/2024] [Indexed: 05/21/2025] Open
Abstract
BACKGROUND AND PURPOSE Allosteric modulation of pentameric ligand-gated ion channels (pLGICs) are critical for the action of neurotransmitters and many psychoactive drugs. However, details of their modulatory mechanisms remain unclear, especially beyond the orthosteric neurotransmitter-binding sites. The recently reported prokaryotic symbiont of Tevnia jerichonana ligand-gated ion channel (sTeLIC), a pH-gated homologue of eukaryotic receptors in the pLGIC family, is thought to be modulated by aromatic compounds via a relatively uncharacterised modulatory site in the extracellular vestibule. EXPERIMENTAL APPROACH We have characterised the effects of psychostimulant derivatives on sTeLIC using two-electrode voltage-clamp electrophysiology in the presence and absence of engineered mutations, and determined X-ray and cryo-EM structures of the channel in both closed and open states. KEY RESULTS We have shown that sTeLIC is sensitive to potentiation by several amphiphilic compounds, which preferentially bind to a vestibular pocket in the contracted open-state extracellular domain. CONCLUSIONS AND IMPLICATIONS This work provides a detailed structure-function mechanism for allosteric potentiation via a noncanonical ligand site, with potential conservation of the eukaryotic pentameric ligand-gated ion channels.
Collapse
Affiliation(s)
- Emelia Karlsson
- Department of Biochemistry and Biophysics, Science for Life Laboratory, Stockholm University, Solna, Sweden
| | - Olivia Andén
- Department of Biochemistry and Biophysics, Science for Life Laboratory, Stockholm University, Solna, Sweden
| | - Chen Fan
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Solna, Sweden
| | - Zaineb Fourati
- Cibles Thérapeutiques et Conception de Médicaments (CiTCoM), CNRS UMR 8038, Université Paris-Cité, Paris, France
| | - Ahmed Haouz
- Plateforme de Cristallographie et de Cristallogenèse, Institut Pasteur, CNRS UMR 3528, Université Paris-Cité, Paris, France
| | - Yuxuan Zhuang
- Department of Biochemistry and Biophysics, Science for Life Laboratory, Stockholm University, Solna, Sweden
| | - Rebecca J Howard
- Department of Biochemistry and Biophysics, Science for Life Laboratory, Stockholm University, Solna, Sweden
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Solna, Sweden
| | - Marc Delarue
- Architecture and Dynamics of Biological Macromolecules, Institut Pasteur, CNRS UMR 3528, Université Paris-Cité, Paris, France
| | - Erik Lindahl
- Department of Biochemistry and Biophysics, Science for Life Laboratory, Stockholm University, Solna, Sweden
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Solna, Sweden
| |
Collapse
|
11
|
Rocereta JA, Sturhahn T, Pumroy RA, Fricke TC, Herzog C, Leffler A, Moiseenkova-Bell V. Structural insights into TRPV2 modulation by probenecid. Nat Struct Mol Biol 2025; 32:1019-1029. [PMID: 39972168 DOI: 10.1038/s41594-025-01494-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 01/22/2025] [Indexed: 02/21/2025]
Abstract
The transient receptor potential vanilloid 2 (TRPV2) cation channel is a key player in cardiovascular physiology and pathophysiology. Probenecid (PBC), an FDA-approved uricosuric agent thought to activate TRPV2, has shown promise in enhancing cardiovascular function in both preclinical and clinical studies. Here our electrophysiological data reveal that PBC significantly potentiates rat TRPV2 to known stimuli, and cryo electron microscopy structures show that PBC directly interacts with rat TRPV2 in a previously unidentified intracellular binding pocket. PBC binding at a conserved TRPV2-specific histidine prevents the channel from taking on the inactivated carboxyl-terminal conformation. This effect extends to TRPV1 and TRPV3 channels when glutamine is substituted with histidine at the corresponding position, increasing their sensitivity to PBC. While PBC alone does not induce TRPV2 opening, its combination with 2-aminoethoxydiphenyl borate enables the channel to adopt an intermediate, potentiated state. Our results offer insights into potential therapeutic advancements for TRPV2 through this pocket.
Collapse
Affiliation(s)
- Julia A Rocereta
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Structural Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Toni Sturhahn
- Department of Anesthesiology and Intensive Care Medicine, Hannover Medical School, Hannover, Germany
| | - Ruth A Pumroy
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Structural Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Tabea C Fricke
- Department of Anesthesiology and Intensive Care Medicine, Hannover Medical School, Hannover, Germany
| | - Christine Herzog
- Department of Anesthesiology and Intensive Care Medicine, Hannover Medical School, Hannover, Germany
| | - Andreas Leffler
- Department of Anesthesiology and Intensive Care Medicine, Hannover Medical School, Hannover, Germany.
| | - Vera Moiseenkova-Bell
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Institute of Structural Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
12
|
Wilson JS, Fortier LC, Fagan RP, Bullough PA. Molecular mechanism of bacteriophage contraction structure of an S-layer-penetrating bacteriophage. Life Sci Alliance 2025; 8:e202403088. [PMID: 40139691 PMCID: PMC11948020 DOI: 10.26508/lsa.202403088] [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: 10/11/2024] [Revised: 03/03/2025] [Accepted: 03/04/2025] [Indexed: 03/29/2025] Open
Abstract
The molecular details of phage tail contraction and bacterial cell envelope penetration remain poorly understood and are completely unknown for phages infecting bacteria enveloped by proteinaceous S-layers. Here, we reveal the extended and contracted atomic structures of an intact contractile-tailed phage (φCD508) that binds to and penetrates the protective S-layer of the Gram-positive human pathogen Clostridioides difficile The tail is unusually long (225 nm), and it is also notable that the tail contracts less than those studied in related contractile injection systems such as the model phage T4 (∼20% compared with ∼50%). Surprisingly, we find no evidence of auxiliary enzymatic domains that other phages exploit in cell wall penetration, suggesting that sufficient energy is released upon tail contraction to penetrate the S-layer and the thick cell wall without enzymatic activity. Instead, the unusually long tail length, which becomes more flexible upon contraction, likely contributes toward the required free energy release for envelope penetration.
Collapse
Affiliation(s)
- Jason S Wilson
- Molecular Microbiology, School of Biosciences, University of Sheffield, Sheffield, UK
| | - Louis-Charles Fortier
- Department of Microbiology and Infectious Diseases, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Canada
| | - Robert P Fagan
- Molecular Microbiology, School of Biosciences, University of Sheffield, Sheffield, UK
- The Florey Institute, University of Sheffield, Sheffield, UK
| | - Per A Bullough
- Molecular Microbiology, School of Biosciences, University of Sheffield, Sheffield, UK
- The Florey Institute, University of Sheffield, Sheffield, UK
| |
Collapse
|
13
|
Raju S, Palakurty S, Sariol A, Wagoner N, Adams LJ, Hui S, Klimstra WB, Fremont DH, Diamond MS. Structural basis for plasticity in receptor engagement by an encephalitic alphavirus. Cell 2025; 188:2943-2956.e24. [PMID: 40187344 DOI: 10.1016/j.cell.2025.02.036] [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: 11/18/2024] [Revised: 01/15/2025] [Accepted: 02/28/2025] [Indexed: 04/07/2025]
Abstract
The structural basis for shifts in receptor usage remains poorly understood despite the implications for virus adaptation and emergence. Western equine encephalitis virus (WEEV) strains exhibit different patterns of engagement for two of their entry receptors: very-low-density lipoprotein receptor (VLDLR) and protocadherin 10 (PCDH10). Using structural and functional studies, we show that while all WEEV strains have a lipoprotein class A (LA) domain binding site near the E1 fusion loop, VLDLR engagement requires a second binding site in E2 that can vary with single nucleotide substitutions. We also resolve a structure of PCDH10 bound to WEEV, which reveals interactions near the E1 fusion loop with residues that also mediate LA domain binding. Evolutionary analysis enabled the generation of a PCDH10 decoy that protects in vivo against all WEEV strains tested. Our experiments demonstrate how viruses can engage multiple receptors using shared determinants, which likely impacts cellular tropism and virulence.
Collapse
Affiliation(s)
- Saravanan Raju
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Sathvik Palakurty
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Alan Sariol
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ngan Wagoner
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Lucas J Adams
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Sean Hui
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - William B Klimstra
- The Center for Vaccine Research and Department of Immunology, The University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Daved H Fremont
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA; Department of Biochemistry & Molecular Biophysics, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | - Michael S Diamond
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO 63110, USA; Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
14
|
Freeman KG, Mondal S, Macale LS, Podgorski J, White SJ, Silva BH, Ortiz V, Huet A, Perez RJ, Narsico JT, Ho MC, Jacobs-Sera D, Lowary TL, Conway JF, Park D, Hatfull GF. Structure and infection dynamics of mycobacteriophage Bxb1. Cell 2025; 188:2925-2942.e17. [PMID: 40239650 PMCID: PMC12124961 DOI: 10.1016/j.cell.2025.03.027] [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: 08/21/2024] [Revised: 11/27/2024] [Accepted: 03/14/2025] [Indexed: 04/18/2025]
Abstract
Mycobacteriophage Bxb1 is a well-characterized virus of Mycobacterium smegmatis with double-stranded DNA and a long, flexible tail. Mycobacteriophages show considerable potential as therapies for Mycobacterium infections, but little is known about the structural details of these phages or how they bind to and traverse the complex Mycobacterium cell wall. Here, we report the complete structure and atomic model of phage Bxb1, including the arrangement of immunodominant domains of both the capsid and tail tube subunits, as well as the assembly of the protein subunits in the tail-tip complex. The structure contains protein assemblies with 3-, 5-, 6-, and 12-fold symmetries, which interact to satisfy several symmetry mismatches. Cryoelectron tomography of phage particles bound to M. smegmatis reveals the structural transitions that occur for free phage particles to bind to the cell surface and navigate through the cell wall to enable DNA transfer into the cytoplasm.
Collapse
Affiliation(s)
- Krista G Freeman
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sudipta Mondal
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, CA, USA
| | - Lourriel S Macale
- Institute of Biological Chemistry, Academia Sinica, Taipei 115, Taiwan
| | - Jennifer Podgorski
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, CT, USA
| | - Simon J White
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, CT, USA
| | - Benjamin H Silva
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, CA, USA
| | - Valery Ortiz
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, CA, USA
| | - Alexis Huet
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Ronelito J Perez
- Institute of Biological Chemistry, Academia Sinica, Taipei 115, Taiwan
| | - Joemark T Narsico
- Institute of Biological Chemistry, Academia Sinica, Taipei 115, Taiwan
| | - Meng-Chiao Ho
- Institute of Biological Chemistry, Academia Sinica, Taipei 115, Taiwan; Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan
| | - Deborah Jacobs-Sera
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Todd L Lowary
- Institute of Biological Chemistry, Academia Sinica, Taipei 115, Taiwan; Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan
| | - James F Conway
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Donghyun Park
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, CA, USA.
| | - Graham F Hatfull
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
15
|
Kujirai T, Kato J, Yamamoto K, Hirai S, Fujii T, Maehara K, Harada A, Negishi L, Ogasawara M, Yamaguchi Y, Ohkawa Y, Takizawa Y, Kurumizaka H. Multiple structures of RNA polymerase II isolated from human nuclei by ChIP-CryoEM analysis. Nat Commun 2025; 16:4724. [PMID: 40436841 PMCID: PMC12119854 DOI: 10.1038/s41467-025-59580-x] [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: 08/02/2024] [Accepted: 04/28/2025] [Indexed: 06/01/2025] Open
Abstract
RNA polymerase II (RNAPII) is a central transcription enzyme that exists as multiple forms with or without accessory factors, and transcribes the genomic DNA packaged in chromatin. To understand how RNAPII functions in the human genome, we isolate transcribing RNAPII complexes from human nuclei by chromatin immunopurification, and determine the cryo-electron microscopy structures of RNAPII elongation complexes (ECs) associated with genomic DNA in distinct forms, without or with the elongation factors SPT4/5, ELOF1, and SPT6. This ChIP-cryoEM method also reveals the two EC-nucleosome complexes corresponding nucleosome disassembly/reassembly processes. In the structure of EC-downstream nucleosome, EC paused at superhelical location (SHL) -5 in the nucleosome, suggesting that SHL(-5) pausing occurs in a sequence-independent manner during nucleosome disassembly. In the structure of the EC-upstream nucleosome, EC directly contacts the nucleosome through the nucleosomal DNA-RPB4/7 stalk and the H2A-H2B dimer-RPB2 wall interactions, suggesting that EC may be paused during nucleosome reassembly. These representative EC structures transcribing the human genome provide mechanistic insights into understanding RNAPII transcription on chromatin.
Collapse
Affiliation(s)
- Tomoya Kujirai
- Laboratory of Chromatin Structure and Function, Institute for Quantitative Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, Japan
- Laboratory for Transcription Structural Biology, RIKEN Center for Biosystems Dynamics Research, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Japan
| | - Junko Kato
- Laboratory of Chromatin Structure and Function, Institute for Quantitative Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, Japan
| | - Kyoka Yamamoto
- Laboratory of Chromatin Structure and Function, Institute for Quantitative Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, Japan
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, Japan
| | - Seiya Hirai
- Laboratory of Chromatin Structure and Function, Institute for Quantitative Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, Japan
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, Japan
| | - Takeru Fujii
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi, Fukuoka, Japan
| | - Kazumitsu Maehara
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi, Fukuoka, Japan
- Department of Multi-Omics, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, Japan
| | - Akihito Harada
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi, Fukuoka, Japan
- Department of Multi-Omics, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, Japan
| | - Lumi Negishi
- Laboratory of Chromatin Structure and Function, Institute for Quantitative Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, Japan
| | - Mitsuo Ogasawara
- Laboratory of Chromatin Structure and Function, Institute for Quantitative Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, Japan
| | - Yuki Yamaguchi
- School of Life Science and Technology, Institute of Science Tokyo, 4259 Nagatsuta, Yokohama, Japan
| | - Yasuyuki Ohkawa
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi, Fukuoka, Japan
| | - Yoshimasa Takizawa
- Laboratory of Chromatin Structure and Function, Institute for Quantitative Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, Japan
| | - Hitoshi Kurumizaka
- Laboratory of Chromatin Structure and Function, Institute for Quantitative Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, Japan.
- Laboratory for Transcription Structural Biology, RIKEN Center for Biosystems Dynamics Research, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Japan.
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, Japan.
| |
Collapse
|
16
|
Williams SM, Raffl S, Kienesberger S, Ilangovan A, Zechner EL, Waksman G. Cryo-EM Structure of the relaxosome, a complex essential for bacterial mating and the spread of antibiotic resistance genes. Nat Commun 2025; 16:4906. [PMID: 40425557 PMCID: PMC12117103 DOI: 10.1038/s41467-025-60116-6] [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: 12/19/2024] [Accepted: 05/13/2025] [Indexed: 05/29/2025] Open
Abstract
Bacterial mating, or conjugation, was discovered nearly 80 years ago as a process transferring genes from one bacterial cell (the donor) to another (the recipient). It requires three key multiprotein complexes in the donor cell: a DNA-processing machinery called the relaxosome, a double-membrane spanning type 4 secretion system (T4SS), and an extracellular appendage termed pilus. While the near-atomic resolution structures of the T4SS and pilus are already known, that of the relaxosome has not been reported to date. Here, we describe the cryo-EM structure of the fully assembled relaxosome encoded by the paradigm F plasmid in two different states corresponding to distinct functional steps along the DNA processing reaction. By varying the structures of model DNAs we delineate conformational changes required to initiate conjugation. Mutational studies of the various protein-protein and protein-DNA interaction hubs suggest a complex sensitive to trigger signals, that could arise from cell-to-cell contacts with recipient cells.
Collapse
Affiliation(s)
- Sunanda M Williams
- Institute of Structural and Molecular Biology, School of Natural Sciences, Birkbeck College, Malet Street, London, WC1E 7HX, UK.
| | - Sandra Raffl
- Institute of Molecular Biosciences, University of Graz, BioTechMed-Graz, Humboldtstrasse 50, 8010, Graz, Austria
| | - Sabine Kienesberger
- Institute of Molecular Biosciences, University of Graz, BioTechMed-Graz, Humboldtstrasse 50, 8010, Graz, Austria
| | - Aravindan Ilangovan
- Institute of Structural and Molecular Biology, School of Natural Sciences, Birkbeck College, Malet Street, London, WC1E 7HX, UK
- Centre for Molecular Cell Biology, School of Biological and Behavioural Sciences, Queen Mary University of London, Newark Street, London, E1 2AT, UK
| | - Ellen L Zechner
- Institute of Molecular Biosciences, University of Graz, BioTechMed-Graz, Humboldtstrasse 50, 8010, Graz, Austria
| | - Gabriel Waksman
- Institute of Structural and Molecular Biology, School of Natural Sciences, Birkbeck College, Malet Street, London, WC1E 7HX, UK.
- Institute of Structural and Molecular Biology, Division of Biosciences, Gower Street, University College London, London, WC1E 6BT, UK.
| |
Collapse
|
17
|
Chen M. Building molecular model series from heterogeneous CryoEM structures using Gaussian mixture models and deep neural networks. Commun Biol 2025; 8:798. [PMID: 40415012 DOI: 10.1038/s42003-025-08202-9] [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: 11/04/2024] [Accepted: 05/09/2025] [Indexed: 05/27/2025] Open
Abstract
Cryogenic electron microscopy (CryoEM) produces structures of macromolecules at near-atomic resolution. However, building molecular models with good stereochemical geometry from those structures can be challenging and time-consuming, especially when many structures are obtained from datasets with conformational heterogeneity. Here we present a model refinement protocol that automatically generates series of molecular models from CryoEM datasets, which describe the dynamics of the macromolecular system and have near-perfect geometry scores. This method makes it easier to interpret the movement of the protein complex from heterogeneity analysis and to compare the structural dynamics observed from CryoEM data with results from other experimental and simulation techniques.
Collapse
Affiliation(s)
- Muyuan Chen
- Division of CryoEM and Bioimaging, SSRL, SLAC National Accelerator Laboratory, Stanford University, Menlo Park, CA, USA.
| |
Collapse
|
18
|
Rosales-Hernandez C, Thoms M, Berninghausen O, Becker T, Beckmann R. Mechanistic insights into Bcs1-mediated mitochondrial membrane translocation of the folded Rieske protein. EMBO J 2025:10.1038/s44318-025-00459-4. [PMID: 40410623 DOI: 10.1038/s44318-025-00459-4] [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: 12/13/2024] [Revised: 04/25/2025] [Accepted: 04/25/2025] [Indexed: 05/25/2025] Open
Abstract
A functional mitochondrial respiratory chain requires coordinated and tightly regulated assembly of mitochondrial- and nuclear-encoded subunits. For bc1 complex (complex III) assembly, the iron-sulfur protein Rip1 must first be imported into the mitochondrial matrix to fold and acquire its 2Fe-2S cluster, then translocated and inserted into the inner mitochondrial membrane (IM). This translocation of folded Rip1 is accomplished by Bcs1, an unusual heptameric AAA ATPase that couples ATP hydrolysis to translocation. However, the molecular and mechanistic details of Bcs1-mediated Rip1 translocation have remained elusive. Here, we provide structural and biochemical evidence on how Bcs1 alternates between conformational states to translocate Rip1 across the IM. Using cryo-electron microscopy (cryo-EM), we identified substrate-bound pre-translocation and pre-release states, revealing how electrostatic interactions promote Rip1 binding to Bcs1. An ATP-induced conformational switch of the Bcs1 heptamer facilitates Rip1 translocation between two distinct aqueous vestibules-one exposed to the matrix, the other to the intermembrane space-in an airlock-like mechanism. This would minimize disruption of the IM permeability barrier, which could otherwise lead to proton leakage and compromised mitochondrial energy conversion.
Collapse
Affiliation(s)
- Cristian Rosales-Hernandez
- Department of Biochemistry, Gene Center, University of Munich, Feodor-Lynen-Str. 25, 81377, Munich, Germany
| | - Matthias Thoms
- Department of Biochemistry, Gene Center, University of Munich, Feodor-Lynen-Str. 25, 81377, Munich, Germany
| | - Otto Berninghausen
- Department of Biochemistry, Gene Center, University of Munich, Feodor-Lynen-Str. 25, 81377, Munich, Germany
| | - Thomas Becker
- Department of Biochemistry, Gene Center, University of Munich, Feodor-Lynen-Str. 25, 81377, Munich, Germany
| | - Roland Beckmann
- Department of Biochemistry, Gene Center, University of Munich, Feodor-Lynen-Str. 25, 81377, Munich, Germany.
| |
Collapse
|
19
|
Corrà S, Zuppardo A, Valenzuela S, Jenninger L, Cerutti R, Sillamaa S, Hoberg E, Johansson KAS, Rovsnik U, Volta S, Silva-Pinheiro P, Davis H, Trifunovic A, Minczuk M, Gustafsson CM, Suomalainen A, Zeviani M, Macao B, Zhu X, Falkenberg M, Viscomi C. Modelling POLG mutations in mice unravels a critical role of POLγΒ in regulating phenotypic severity. Nat Commun 2025; 16:4782. [PMID: 40404629 PMCID: PMC12098916 DOI: 10.1038/s41467-025-60059-y] [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: 08/26/2024] [Accepted: 05/14/2025] [Indexed: 05/24/2025] Open
Abstract
DNA polymerase γ (POLγ), responsible for mitochondrial DNA replication, consists of a catalytic POLγA subunit and two accessory POLγB subunits. Mutations in POLG, which encodes POLγA, lead to various mitochondrial diseases. We investigated the most common POLG mutations (A467T, W748S, G848S, Y955C) by characterizing human and mouse POLγ variants. Our data reveal that these mutations significantly impair POLγ activities, with mouse variants exhibiting milder defects. Cryogenic electron microscopy highlighted structural differences between human and mouse POLγ, particularly in the POLγB subunit, which may explain the higher activity of mouse POLγ and the reduced severity of mutations in mice. We further generated a panel of mouse models mirroring common human POLG mutations, providing crucial insights into the pathogenesis of POLG-related disorders and establishing robust models for therapeutic development. Our findings emphasize the importance of POLγB in modulating the severity of POLG mutations.
Collapse
Affiliation(s)
- Samantha Corrà
- Veneto Institute of Molecular Medicine (VIMM), Via Orus, 2-35129, Padova, Italy
| | - Alessandro Zuppardo
- Veneto Institute of Molecular Medicine (VIMM), Via Orus, 2-35129, Padova, Italy
- Department of Biomedical Sciences, University of Padova, Via Ugo Bassi, 58/B-35131, Padova, Italy
| | - Sebastian Valenzuela
- Department of Medical Biochemistry and Cell Biology, University of Gothenburg, Medicinaregatan 9A, P.O. Box 440, 41390, Gothenburg, Sweden
| | - Louise Jenninger
- Department of Medical Biochemistry and Cell Biology, University of Gothenburg, Medicinaregatan 9A, P.O. Box 440, 41390, Gothenburg, Sweden
| | - Raffaele Cerutti
- Veneto Institute of Molecular Medicine (VIMM), Via Orus, 2-35129, Padova, Italy
- Department of Biomedical Sciences, University of Padova, Via Ugo Bassi, 58/B-35131, Padova, Italy
| | - Sirelin Sillamaa
- Department of Medical Biochemistry and Cell Biology, University of Gothenburg, Medicinaregatan 9A, P.O. Box 440, 41390, Gothenburg, Sweden
| | - Emily Hoberg
- Department of Medical Biochemistry and Cell Biology, University of Gothenburg, Medicinaregatan 9A, P.O. Box 440, 41390, Gothenburg, Sweden
| | - Katarina A S Johansson
- Department of Medical Biochemistry and Cell Biology, University of Gothenburg, Medicinaregatan 9A, P.O. Box 440, 41390, Gothenburg, Sweden
| | - Urska Rovsnik
- Department of Medical Biochemistry and Cell Biology, University of Gothenburg, Medicinaregatan 9A, P.O. Box 440, 41390, Gothenburg, Sweden
| | - Sara Volta
- Veneto Institute of Molecular Medicine (VIMM), Via Orus, 2-35129, Padova, Italy
- Department of Biomedical Sciences, University of Padova, Via Ugo Bassi, 58/B-35131, Padova, Italy
| | - Pedro Silva-Pinheiro
- MRC Mitochondrial Biology Unit, University of Cambridge, Hills Road, Cambridge, CB2 0XY, UK
| | - Hannah Davis
- The Mary Lyon Centre, MRC Harwell, Becquerel Ave, Didcot, Oxfordshire, OX11 0RD, UK
| | - Aleksandra Trifunovic
- Institute for Mitochondrial Diseases and Aging, Faculty of Medicine, CECAD Research Center, 50931, Cologne, Germany
| | - Michal Minczuk
- MRC Mitochondrial Biology Unit, University of Cambridge, Hills Road, Cambridge, CB2 0XY, UK
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, CB2 2PY, UK
| | - Claes M Gustafsson
- Department of Medical Biochemistry and Cell Biology, University of Gothenburg, Medicinaregatan 9A, P.O. Box 440, 41390, Gothenburg, Sweden
| | - Anu Suomalainen
- Research Programs Unit, Stem Cells and Metabolism, Biomedicum-Helsinki, Haartmaninkatu 8, University of Helsinki, 00290, Helsinki, Finland
- HUSlab, Helsinki University Hospital, University of Helsinki, 00290, Helsinki, Finland
| | - Massimo Zeviani
- Department of Neurosciences, University of Padova, Via Belzoni 160, 35121, Padova, Italy
- Institute for Maternal and Child Health, IRCCS "Burlo Garofolo", Via Istria 61, 34137, Trieste, Italy
| | - Bertil Macao
- Department of Medical Biochemistry and Cell Biology, University of Gothenburg, Medicinaregatan 9A, P.O. Box 440, 41390, Gothenburg, Sweden
| | - Xuefeng Zhu
- Department of Medical Biochemistry and Cell Biology, University of Gothenburg, Medicinaregatan 9A, P.O. Box 440, 41390, Gothenburg, Sweden.
- School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, 063210, China.
- National Key Laboratory for Development and Utilization of Forest Food Resources, Zhejiang A&F University, Hangzhou, 311300, China.
| | - Maria Falkenberg
- Department of Medical Biochemistry and Cell Biology, University of Gothenburg, Medicinaregatan 9A, P.O. Box 440, 41390, Gothenburg, Sweden.
| | - Carlo Viscomi
- Veneto Institute of Molecular Medicine (VIMM), Via Orus, 2-35129, Padova, Italy.
- Department of Biomedical Sciences, University of Padova, Via Ugo Bassi, 58/B-35131, Padova, Italy.
| |
Collapse
|
20
|
Mandala VS, MacKinnon R. Electric field-induced pore constriction in the human K v2.1 channel. Proc Natl Acad Sci U S A 2025; 122:e2426744122. [PMID: 40366685 DOI: 10.1073/pnas.2426744122] [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: 12/20/2024] [Accepted: 04/10/2025] [Indexed: 05/15/2025] Open
Abstract
Gating in voltage-dependent ion channels is regulated by the transmembrane voltage. This form of regulation is enabled by voltage-sensing domains (VSDs) that respond to transmembrane voltage differences by changing their conformation and exerting force on the pore to open or close it. Here, we use cryogenic electron microscopy to study the neuronal Kv2.1 channel in lipid vesicles with and without a voltage difference across the membrane. Hyperpolarizing voltage differences displace the positively charged S4 helix in the voltage sensor by one helical turn (~5 Å). When this displacement occurs, the S4 helix changes its contact with the pore at two different interfaces. When these changes are observed in fewer than four voltage sensors, the pore remains open, but when they are observed in all four voltage sensors, the pore constricts. The constriction occurs because the S4 helix, as it displaces inward, squeezes the right-handed helical bundle of pore-lining S6 helices. A similar conformational change occurs upon hyperpolarization of the EAG1 channel but with two helical turns displaced instead of one. Therefore, while Kv2.1 and EAG1 are from distinct architectural classes of voltage-dependent ion channels, called domain-swapped and non-domain-swapped, the way the voltage sensors gate their pores is very similar.
Collapse
Affiliation(s)
- Venkata Shiva Mandala
- Laboratory of Molecular Neurobiology and Biophysics, HHMI, The Rockefeller University, New York, NY 10065
| | - Roderick MacKinnon
- Laboratory of Molecular Neurobiology and Biophysics, HHMI, The Rockefeller University, New York, NY 10065
| |
Collapse
|
21
|
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
|
22
|
Meng X, Li Y, Xu J, Wu K, Hu W, Wu C, Xu HE, Xu Y. Structural insights into the activation of the human prostaglandin E 2 receptor EP1 subtype by prostaglandin E 2. Proc Natl Acad Sci U S A 2025; 122:e2423840122. [PMID: 40366695 DOI: 10.1073/pnas.2423840122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 04/04/2025] [Indexed: 05/15/2025] Open
Abstract
Prostaglandin E2 (PGE2) mediates diverse physiological processes through four G protein-coupled receptor subtypes (EP1-EP4). While structures of EP2, EP3, and EP4 have been determined, the structural basis for PGE2 recognition and activation of the EP1 receptor subtype has remained elusive due to its inherent instability. Here, we present the cryoelectron microscopy structure of the human EP1 receptor in complex with PGE2 and heterotrimeric Gq protein at 2.55 Å resolution, completing the structural characterization of the EP receptor family. Our structure reveals a unique binding mode of PGE2 within EP1, involving key interactions with residues in the orthosteric pocket. Notably, we observe a less pronounced outward displacement of transmembrane helix 6 compared to other EP receptor subtypes, suggesting a distinct activation mechanism for EP1. Through extensive mutational analyses, we identify critical residues involved in PGE2 recognition, EP1 activation, and Gq protein coupling. By overcoming the challenges associated with the instability of EP1, our findings provide valuable insights into the subtype-specific activation mechanisms of EP receptors and lay the foundation for the development of more selective EP1-targeted therapeutics.
Collapse
Affiliation(s)
- Xue Meng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yang Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Jiuyin Xu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Kai Wu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Wen Hu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Canrong Wu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Research Center for Medicinal Structural Biology, National Research Center for Translational Medicine at Shanghai, State Key Laboratory of Medical Genomics, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - H Eric Xu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- School of Pharmacy, Fudan University, Shanghai 201203, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
- Research Center for Medicinal Structural Biology, National Research Center for Translational Medicine at Shanghai, State Key Laboratory of Medical Genomics, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Youwei Xu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the National Medical Products Administration and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, Guangdong, China
| |
Collapse
|
23
|
El Mammeri N, Duan P, Hong M. Structures of ΔD421 Truncated Tau Fibrils. J Mol Biol 2025; 437:169051. [PMID: 40021051 DOI: 10.1016/j.jmb.2025.169051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 02/23/2025] [Accepted: 02/24/2025] [Indexed: 03/03/2025]
Abstract
The microtubule-associated protein tau aggregates into pathological β-sheet amyloid fibrils in Alzheimer's disease (AD) and other neurodegenerative diseases. In these aggregates, tau is chemically modified, including abnormal hyperphosphorylation and truncation. Truncation after D421 in the C-terminal domain occurs at early stages of AD. Here we investigate the structures of ΔD421-truncated 0N4R tau fibrils assembled in vitro in the absence of anionic cofactors. Using solid-state NMR spectroscopy and cryoelectron microscopy, we show that ΔD421-truncated 0N4R tau forms homogeneous fibrils whose rigid core adopts a three-layered β-sheet structure that spans R2, R3 and R4 repeats. This structure is essentially identical to that of full-length tau containing phospho-mimetic mutations at the PHF1 epitope in the C-terminal domain. In comparison, a ΔD421-truncated tau that additionally contains three phospho-mimetic mutations at the AT8 epitope in the proline-rich region forms a fibril core that includes the first half of the C-terminal domain, which is excluded from all known pathological tau fibril cores. These results indicate that the posttranslational modification code of tau contains redundancy: both charge modification and truncation of the C-terminal domain promote a three-layered β-sheet structure, which resembles pathological four-repeat tau structures in several tauopathies. In comparison, reducing the positive charges at the AT8 epitope in ΔD421-truncated tau promotes a fibril core that includes an immobilized C-terminal domain. The absence of this structure in tauopathy brains implies that ΔD421 truncation does not occur in conjunction with AT8 phosphorylation in diseased brains.
Collapse
Affiliation(s)
- Nadia El Mammeri
- Department of Chemistry, Massachusetts Institute of Technology, 170 Albany Street, Cambridge, MA 02139, United States
| | - Pu Duan
- Department of Chemistry, Massachusetts Institute of Technology, 170 Albany Street, Cambridge, MA 02139, United States
| | - Mei Hong
- Department of Chemistry, Massachusetts Institute of Technology, 170 Albany Street, Cambridge, MA 02139, United States.
| |
Collapse
|
24
|
James AM, Farnung L. Structural basis of human CHD1 nucleosome recruitment and pausing. Mol Cell 2025; 85:1938-1951.e6. [PMID: 40334658 PMCID: PMC12126155 DOI: 10.1016/j.molcel.2025.04.020] [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: 11/14/2024] [Revised: 02/26/2025] [Accepted: 04/16/2025] [Indexed: 05/09/2025]
Abstract
Chromatin remodelers regulate gene expression and genome maintenance by controlling nucleosome positioning, but the structural basis for their regulated and directional activity remains poorly understood. Here, we present three cryoelectron microscopy (cryo-EM) structures of human chromodomain helicase DNA-binding protein 1 (CHD1) bound to nucleosomes that reveal previously unobserved recruitment and regulatory states. We identify a structural element, termed the "anchor element," that connects the CHD1 ATPase motor to the nucleosome entry-side acidic patch. The anchor element coordinates with other regulatory modules, including the gating element, which undergoes a conformational switch critical for remodeling. Our structures demonstrate how the DNA-binding region of CHD1 binds entry- and exit-side DNA during remodeling to achieve directional sliding. The observed structural elements are conserved across chromatin remodelers, suggesting a unified mechanism for nucleosome recognition and remodeling. Our findings show how chromatin remodelers couple nucleosome recruitment to regulated DNA translocation, providing a framework for understanding chromatin remodeler mechanisms beyond DNA translocation.
Collapse
Affiliation(s)
- Allison M James
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Lucas Farnung
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
25
|
Zhou Q, Sagmeister T, Hutten S, Bourgeois B, Pavkov-Keller T, Dormann D, Madl T. Structural basis of phosphorylation-independent nuclear import of CIRBP by TNPO3. Nat Commun 2025; 16:4456. [PMID: 40360518 PMCID: PMC12075686 DOI: 10.1038/s41467-025-59802-2] [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: 08/22/2024] [Accepted: 05/06/2025] [Indexed: 05/15/2025] Open
Abstract
Transportin 3 (TNPO3) is a nuclear import receptor known for its broad substrate specificity, often recognizing arginine-serine (SR/RS) repeat-rich nuclear localization signals (NLS) in SRSF proteins. While serine phosphorylation or glutamate presence has been associated with these NLSs, recent proteomic studies identified TNPO3 cargoes lacking SR/RS repeats. One such example is the cold-inducible RNA-binding protein (CIRBP), which contains a non-classical RSY-NLS. Using X-ray crystallography, here we investigate the TNPO3-CIRBP interaction and find that tyrosines within the RSY-NLS play a key role in binding, independent of phosphorylation. Surprisingly, serine and tyrosine phosphorylation in CIRBP's NLS inhibits TNPO3 binding, suggesting a regulatory mechanism for nuclear import. Our study reveals a non-conventional nuclear import mechanism mediated by TNPO3, which may extend to other known or yet undiscovered TNPO3 cargoes.
Collapse
Affiliation(s)
- Qishun Zhou
- Research Unit Integrative Structural Biology, Medicinal Chemistry, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
- Institut Pasteur, Université Paris Cité, CNRS UMR3528, Bacterial Transmembrane Systems Unit, Paris, France
| | - Theo Sagmeister
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Saskia Hutten
- Johannes Gutenberg Universität Mainz, Institute of Molecular Physiology, Mainz, Germany
| | - Benjamin Bourgeois
- Research Unit Integrative Structural Biology, Medicinal Chemistry, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Tea Pavkov-Keller
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
- Field of Excellence BioHealth, University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| | - Dorothee Dormann
- Johannes Gutenberg Universität Mainz, Institute of Molecular Physiology, Mainz, Germany
- Institute of Molecular Biology (IMB) Mainz, Mainz, Germany
| | - Tobias Madl
- Research Unit Integrative Structural Biology, Medicinal Chemistry, Otto Loewi Research Center, Medical University of Graz, Graz, Austria.
- BioTechMed-Graz, Graz, Austria.
| |
Collapse
|
26
|
Tassinari M, Tanzi G, Maggiore F, Groeneweg S, van Geest FS, Freund MET, Stavast CJ, Boniardi I, Pasqualato S, Visser WE, Coscia F. Molecular mechanism of thyroxine transport by monocarboxylate transporters. Nat Commun 2025; 16:4493. [PMID: 40368961 PMCID: PMC12078798 DOI: 10.1038/s41467-025-59751-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 05/02/2025] [Indexed: 05/16/2025] Open
Abstract
Thyroid hormones (the common name for prohormone thyroxine and the bioactive form triiodothyronine) control major developmental and metabolic processes. Release of thyroid hormones from the thyroid gland into the bloodstream and their transport into target cells is facilitated by plasma membrane transporters, including monocarboxylate transporter (MCT)8 and the highly homologous MCT10. However, the molecular mechanism underlying thyroid hormone transport is unknown. The relevance of such transporters is illustrated in patients with MCT8 deficiency, a severe neurodevelopmental and metabolic disorder. Using cryogenic-sample electron microscopy (cryo-EM), we determined the ligand-free and thyroxine-bound human MCT8 structures in the outward-facing state and the thyroxine-bound human MCT10 in the inward-facing state. Our structural analysis revealed a network of conserved gate residues involved in conformational changes upon thyroxine binding, triggering ligand release in the opposite compartment. We then determined the structure of a folded but inactive patient-derived MCT8 mutant, indicating a subtle conformational change which explains its reduced transport activity. Finally, we report a structure of MCT8 bound to its inhibitor silychristin, locked in the outward-facing state, revealing the molecular basis of its action and specificity. Taken together, this study advances mechanistic understanding of normal and disordered thyroid hormone transport.
Collapse
Affiliation(s)
| | | | | | - Stefan Groeneweg
- Department of Internal Medicine, Academic Centre for Thyroid Diseases, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Ferdy S van Geest
- Department of Internal Medicine, Academic Centre for Thyroid Diseases, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Matthijs E T Freund
- Department of Internal Medicine, Academic Centre for Thyroid Diseases, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Christiaan J Stavast
- Department of Internal Medicine, Academic Centre for Thyroid Diseases, Erasmus Medical Centre, Rotterdam, The Netherlands
| | | | | | - W Edward Visser
- Department of Internal Medicine, Academic Centre for Thyroid Diseases, Erasmus Medical Centre, Rotterdam, The Netherlands.
| | | |
Collapse
|
27
|
Gen R, Addetia A, Asarnow D, Park YJ, Quispe J, Chan MC, Brown JT, Lee J, Campbell MG, Lapointe CP, Veesler D. SARS-CoV-2 nsp1 mediates broad inhibition of translation in mammals. Cell Rep 2025; 44:115696. [PMID: 40359110 DOI: 10.1016/j.celrep.2025.115696] [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: 01/04/2025] [Revised: 03/13/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) non-structural protein 1 (nsp1) promotes innate immune evasion by inhibiting host translation in human cells. However, the role of nsp1 in other host species remains elusive, especially in bats-natural reservoirs of sarbecoviruses with a markedly different innate immune system than humans. We reveal that nsp1 potently inhibits translation in Rhinolophus lepidus bat cells, which belong to the same genus as known sarbecovirus reservoir hosts. We determined a cryoelectron microscopy structure of nsp1 bound to the R. lepidus 40S ribosomal subunit, showing that it blocks the mRNA entry channel by targeting a highly conserved site among mammals. Accordingly, we found that nsp1 blocked protein translation in mammalian cells from several species, underscoring its broadly inhibitory activity and conserved role in numerous SARS-CoV-2 hosts. Our findings illuminate the arms race between coronaviruses and mammalian host immunity, providing a foundation for understanding the determinants of viral maintenance in bat hosts and spillover.
Collapse
Affiliation(s)
- Risako Gen
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Amin Addetia
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Daniel Asarnow
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Young-Jun Park
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Joel Quispe
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Matthew C Chan
- Division of Basic Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Jack T Brown
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Jimin Lee
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Melody G Campbell
- Division of Basic Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | | | - David Veesler
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
28
|
Stevens A, Kashyap S, Crofut E, Alvarez-Cabrera AL, Jih J, Liu Y, Zhou ZH. Structure of a new capsid form and comparison with A-, B- and C-capsids clarify herpesvirus assembly and DNA translocation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.19.644230. [PMID: 40166288 PMCID: PMC11957103 DOI: 10.1101/2025.03.19.644230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Three capsid types have been recognized from the nuclei of herpesvirus-infected cells: empty A-capsids, scaffolding-containing B-capsids, and DNA-filled C-capsids. Despite great progress in determining the structures of these capsids and extracellular virions in recent years, debate persists concerning the origins and temporal relationships among these capsids during capsid assembly and genome packaging. Here, we have imaged over 300,000 capsids of herpes simplex virus type 1 by cryogenic electron microscopy (cryoEM) and exhaustively classified them to characterize the structural heterogeneity of the DNA-translocating portal complex and their functional states. The resultant atomic structures reveal not only the expected A-, B-, and C-capsids, but also capsids with portal vertices similar to C-capsids but no resolvable genome in the capsid lumen, which we named D-capsids. The dodecameric dsDNA-translocating portal complex varies in their radial positions in the icosahedral capsids and structural conformations among these capsids. In D-capsids, terminal DNA density exists in multiple conformations including one reminiscent to that in C-capsids, suggesting D-capsids are products of failed DNA retention. This interpretation is supported by varying amounts of DNA outside individual D-capsids and by correlation of capsid counts observed in situ of infected cell nuclei and those after purification. Additionally, an "anchoring" segment of the scaffold protein is resolved interacting with the portal baskets of A- and B-capsids but not D- and C-capsids. Taken together, our data indicate that A-capsids arise from failed DNA packaging and D-capsids from failed genome retention, clarifying the origins of empty capsids in herpesvirus assembly.
Collapse
|
29
|
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
|
30
|
Grill-Walcher S, Schäffer C. A new age in structural S-layer biology - Experimental and in silico milestones. J Biol Chem 2025:110205. [PMID: 40345586 DOI: 10.1016/j.jbc.2025.110205] [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: 12/20/2024] [Revised: 04/23/2025] [Accepted: 04/25/2025] [Indexed: 05/11/2025] Open
Abstract
Surface (S-) layer proteins, considered as the most abundant proteins in nature, perform diverse and essential biological roles in many bacteria and most archaea. Their functions range from providing structural support, maintaining cell shape, and protecting against extreme environments to acting as a cell surface display matrix for biologically active molecules, such as S-layer protein-bound glycans, which facilitate interspecies interactions and cellular communication in both health and disease. The intricate, symmetric, nanometer-scale patterns of S-layer lattices have long fascinated structural biologists, yet only recent methodological advances have revealed detailed molecular insights. These advances include a deeper understanding of domain organization, cell wall anchoring mechanisms, and how nascent proteins are incorporated into existing lattices. Significant progress in sample preparation and high-resolution imaging has led to the precise structural characterization of S-layers across various bacterial and archaeal species. Furthermore, the advent of deep learning-based structure prediction has enabled modeling of S-layer proteins in several largely uncultured microbial lineages. This review summarizes major achievements in S-layer protein structural research over the past five years, presenting them with a typical workflow for the experimental structure determination. For the first time, it also explores recent breakthroughs in computational S-layer modelling and offers an outlook on how in silico methods may further advance our understanding of S-layer protein architecture.
Collapse
Affiliation(s)
- Stephanie Grill-Walcher
- Department of Natural Sciences and Sustainable Resources, Institute of Biochemistry, NanoGlycobiology Research Group, BOKU University, Vienna, Austria
| | - Christina Schäffer
- Department of Natural Sciences and Sustainable Resources, Institute of Biochemistry, NanoGlycobiology Research Group, BOKU University, Vienna, Austria.
| |
Collapse
|
31
|
Penzes JJ, Kaelber JT. Capsid Structure of the Fish Pathogen Syngnathus Scovelli Chapparvovirus Offers a New Perspective on Parvovirus Structural Biology. Viruses 2025; 17:679. [PMID: 40431691 PMCID: PMC12115719 DOI: 10.3390/v17050679] [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: 03/22/2025] [Revised: 05/02/2025] [Accepted: 05/03/2025] [Indexed: 05/29/2025] Open
Abstract
Chapparvoviruses (ChPVs) comprise a divergent lineage of the Parvoviridae ssDNA virus family and evolved to infect vertebrate animals independently from the Parvovirinae subfamily. Despite being pathogenic and widespread in environmental samples and metagenomic assemblies, their structural characterization has proven challenging. Here, we report the first structural analysis of a ChPV, represented by the fish pathogen, Syngnathus scovelli chapparvovirus (SsChPV). We show through the SsChPV structure that the lineage harbors a surface morphology, subunit structure, and multimer interactions that are unique among parvoviruses. The SsChPV capsid evolved a threefold-related depression of α-helices that is analogous to the β-annulus pore of denso- and hamaparvoviruses and may play a role in monomer oligomerization during assembly. As interacting β-strands are absent from the twofold symmetry axis, the viral particle lacks the typical stability and resilience of parvovirus capsids. Although all parvoviruses thus far rely on the threading of large, flexible N-terminal domains to the capsid surface for their intracellular trafficking, our results show that ChPVs completely lack any such N-terminal sequences. This led to the subsequent degradation of their fivefold channel, the site of N-terminus externalization. These findings suggest that ChPVs harbor an infectious pathway that significantly deviates from the rest of the Parvoviridae.
Collapse
Affiliation(s)
- Judit J. Penzes
- Institute for Quantitative Biomedicine, Rutgers, The State University of New Jersey, Piscataway, NJ 08812, USA;
- Department of Entomology, Texas A&M University, College Station, TX 77845, USA
| | - Jason T. Kaelber
- Institute for Quantitative Biomedicine, Rutgers, The State University of New Jersey, Piscataway, NJ 08812, USA;
| |
Collapse
|
32
|
Jin M, Hassan Z, Li Z, Liu Y, Marakhovskaia A, Wong AHM, Forman A, Nitz M, Gilbert M, Yu H, Chen X, Rini JM. Human coronavirus HKU1 spike structures reveal the basis for sialoglycan specificity and carbohydrate-promoted conformational changes. Nat Commun 2025; 16:4158. [PMID: 40324974 PMCID: PMC12053599 DOI: 10.1038/s41467-025-59137-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 04/10/2025] [Indexed: 05/07/2025] Open
Abstract
The human coronavirus HKU1 uses both sialoglycoconjugates and the protein transmembrane serine protease 2 (TMPRSS2) as receptors. Carbohydrate binding leads to the spike protein up conformation required for TMPRSS2 binding, an outcome suggesting a distinct mechanism for driving fusion of the viral and host cell membranes. Nevertheless, the conformational changes promoted by carbohydrate binding have not been fully elucidated and the basis for HKU1's carbohydrate binding specificity remains unknown. Reported here are high resolution cryo-EM structures of the HKU1 spike protein trimer in its apo form and in complex with the carbohydrate moiety of a candidate carbohydrate receptor, the 9-O-acetylated GD3 ganglioside. The structures show that the spike monomer can exist in four discrete conformational states and that progression through them would promote the up conformation upon carbohydrate binding. We also show that a six-amino-acid insert is a determinant of HKU1's specificity for gangliosides containing a 9-O-acetylated α2-8-linked disialic acid moiety and that HKU1 shows weak affinity for the 9-O-acetylated sialic acids found on decoy receptors such as mucins.
Collapse
Affiliation(s)
- Min Jin
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Zaky Hassan
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Zhijie Li
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Ying Liu
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | | | - Alan H M Wong
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Adam Forman
- Department of Chemistry, University of Toronto, Toronto, Ontario, Canada
| | - Mark Nitz
- Department of Chemistry, University of Toronto, Toronto, Ontario, Canada
| | - Michel Gilbert
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, Canada
| | - Hai Yu
- Department of Chemistry, University of California-Davis, Davis, CA, USA
| | - Xi Chen
- Department of Chemistry, University of California-Davis, Davis, CA, USA
| | - James M Rini
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada.
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
33
|
Vostal LE, Dahan NE, Reynolds MJ, Kronenberg LI, Kapoor TM. Structural insights into the coupling between VCP, an essential unfoldase, and a deubiquitinase. J Cell Biol 2025; 224:e202410148. [PMID: 40085114 PMCID: PMC11908451 DOI: 10.1083/jcb.202410148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 01/08/2025] [Accepted: 02/09/2025] [Indexed: 03/16/2025] Open
Abstract
Proteostasis involves degradation and recycling of proteins from organelles, membranes, and multiprotein complexes. These processes can depend on protein extraction and unfolding by the essential mechanoenzyme valosin-containing protein (VCP) and on ubiquitin chain remodeling by ubiquitin-specific proteases known as deubiquitinases (DUBs). How the activities of VCP and DUBs are coordinated is poorly understood. Here, we focus on the DUB VCPIP1, a VCP interactor required for post-mitotic Golgi and ER organization. We determine ∼3.3 Å cryogenic electron microscopy structures of VCP-VCPIP1 complexes in the absence of added nucleotide or the presence of an ATP analog. We find that up to 3 VCPIP1 protomers interact with the VCP hexamer to position VCPIP1's catalytic domain at the exit of VCP's central pore, poised to cleave ubiquitin following substrate unfolding. We observe competition between VCPIP1 and other cofactors for VCP binding and show that VCP stimulates VCPIP1's DUB activity. Together, our data suggest how the two enzyme activities can be coordinated to regulate proteostasis.
Collapse
Affiliation(s)
- Lauren E. Vostal
- Laboratory of Chemistry and Cell Biology, The Rockefeller University, New York, NY, USA
- Tri-Institutional PhD Program in Chemical Biology, New York, NY, USA
| | - Noa E. Dahan
- Laboratory of Chemistry and Cell Biology, The Rockefeller University, New York, NY, USA
- The David Rockefeller Graduate Program in Bioscience, The Rockefeller University, New York, NY, USA
| | - Matthew J. Reynolds
- Laboratory of Structural Biophysics and Mechanobiology, The Rockefeller University, New York, NY, USA
| | - Lily I. Kronenberg
- Laboratory of Chemistry and Cell Biology, The Rockefeller University, New York, NY, USA
| | - Tarun M. Kapoor
- Laboratory of Chemistry and Cell Biology, The Rockefeller University, New York, NY, USA
| |
Collapse
|
34
|
Guo Y, Yang G, Liu H, Chai J, Chen J, Shanklin J, Liu Q, Liu B, Lu M. Structure and mechanism of human vesicular polyamine transporter. Nat Commun 2025; 16:4142. [PMID: 40319071 PMCID: PMC12049414 DOI: 10.1038/s41467-025-59549-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 04/25/2025] [Indexed: 05/07/2025] Open
Abstract
Polyamines play essential roles in gene expression and modulate neuronal transmission in mammals. Vesicular polyamine transporters (VPAT) from the SLC18 family exploit the transmembrane H+ gradient to translocate polyamines into secretory vesicles, enabling the quantal release of polyamine neuromodulators and underpinning learning and memory formation. Here, we report the cryo-electron microscopy structures of human VPAT in complex with spermine, spermidine, H+, or tetrabenazine, elucidating discrete lumen-facing states of the antiporter and pivotal interactions between VPAT and its substrate or inhibitor. Leveraging structure-inspired mutagenesis studies and protein structure prediction, we deduce an unforeseen mechanism whereby the polyamine and H+ compete for multiple acidic protein residues both directly and indirectly, and rationalize how the antidopaminergic therapeutic tetrabenazine impedes vesicular transport of polyamines. This study unravels the mechanism of an H+-coupled polyamine antiporter, reveals mechanistic diversity between VPAT and other SLC18 antiporters, and raises new prospects for combating human disorders of polyamine homeostasis.
Collapse
Affiliation(s)
- Yi Guo
- Center for Proteomics & Molecular Therapeutics, Rosalind Franklin University of Medicine & Science, 3333 Green Bay Road, North Chicago, IL, 60064, USA
| | - Ge Yang
- The Hormel Institute, University of Minnesota, 801 16th Ave NE, Austin, MN, 55921, USA
| | - Haijiao Liu
- Biology Department, Brookhaven National Laboratory, Bldg. 463, Upton, NY, 11973, USA
- Department of Materials Sciences & Chemical Engineering, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Jin Chai
- Biology Department, Brookhaven National Laboratory, Bldg. 463, Upton, NY, 11973, USA
| | - Jie Chen
- The Hormel Institute, University of Minnesota, 801 16th Ave NE, Austin, MN, 55921, USA
| | - John Shanklin
- Biology Department, Brookhaven National Laboratory, Bldg. 463, Upton, NY, 11973, USA
| | - Qun Liu
- Biology Department, Brookhaven National Laboratory, Bldg. 463, Upton, NY, 11973, USA.
| | - Bin Liu
- The Hormel Institute, University of Minnesota, 801 16th Ave NE, Austin, MN, 55921, USA.
| | - Min Lu
- Center for Proteomics & Molecular Therapeutics, Rosalind Franklin University of Medicine & Science, 3333 Green Bay Road, North Chicago, IL, 60064, USA.
| |
Collapse
|
35
|
Penchev I, Gumbin S, Scavone F, Berninghausen O, Becker T, Kopito R, Beckmann R. UFMylation orchestrates spatiotemporal coordination of RQC at the ER. SCIENCE ADVANCES 2025; 11:eadv0435. [PMID: 40315331 PMCID: PMC12047416 DOI: 10.1126/sciadv.adv0435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 03/27/2025] [Indexed: 05/04/2025]
Abstract
Degradation of arrest peptides from endoplasmic reticulum (ER) translocon-bound 60S ribosomal subunits via the ribosome-associated quality control (ER-RQC) pathway requires covalent modification of RPL26/uL24 on 60S ribosomal subunits with UFM1. However, the underlying mechanism that coordinates the UFMylation and RQC pathways remains elusive. Structural analysis of ER-RQC intermediates revealed concomitant binding and direct interaction of the UFMylation and RQC machineries on the 60S. In the presence of an arrested peptidyl-transfer RNA, the RQC factor NEMF and the UFM1 E3 ligase (E3UFM1) form a direct interaction via the UFL1 subunit of E3UFM1, and UFL1 adopts a conformation distinct from that previously observed for posttermination 60S. While this concomitant binding occurs on translocon-bound 60S, LTN1 recruitment and arrest peptide degradation require UFMylation-dependent 60S dissociation from the translocon. These data reveal a mechanism by which the UFMylation cycle orchestrates ER-RQC.
Collapse
Affiliation(s)
- Ivan Penchev
- Department of Biochemistry, Gene Center, Feodor-Lynen-Str. 25, University of Munich, 81377, Munich, Germany
| | - Samantha Gumbin
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | | | - Otto Berninghausen
- Department of Biochemistry, Gene Center, Feodor-Lynen-Str. 25, University of Munich, 81377, Munich, Germany
| | - Thomas Becker
- Department of Biochemistry, Gene Center, Feodor-Lynen-Str. 25, University of Munich, 81377, Munich, Germany
| | - Ron Kopito
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Roland Beckmann
- Department of Biochemistry, Gene Center, Feodor-Lynen-Str. 25, University of Munich, 81377, Munich, Germany
| |
Collapse
|
36
|
Jespersen N, Prajapati JD, Singhal A, Sanbonmatsu KY. Cryo-EM reveals remodeling of a tandem riboswitch at 2.9 Å resolution. RESEARCH SQUARE 2025:rs.3.rs-6422592. [PMID: 40343338 PMCID: PMC12060979 DOI: 10.21203/rs.3.rs-6422592/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 05/11/2025]
Abstract
Riboswitches are non-coding RNA sequences that control cellular processes through ligand binding. Conformational heterogeneity is fundamental to riboswitch functionality, yet this same attribute makes structural characterization of these mRNA elements challenging. Here, we use a combination of molecular dynamics and cryo-electron microscopy to expound the flexible nature of the glycine riboswitch tandem aptamers and characterize diMerent structural populations. We find that Mg2+ partially stabilizes the fully folded state, resulting in one-third of the particles adopting a unique "walking man" conformation, consisting of a rigidified core and two dynamic helices, and two-thirds adopting distinct, partially folded states. Glycine interactions double the relative population of fully folded particles by stabilizing a conserved inter-aptamer Hoogsteen base pair, enabling our capture of a 2.9 Å structure for this RNA-only system. The population data show that glycine and Mg2+ operate synergistically: glycine enhances Mg2+ occupancy, while Mg2+ drives glycine specificity. Our findings indicate that cryo-electron microscopy oMers a promising avenue to characterize RNA folding ensembles.
Collapse
Affiliation(s)
- Nathan Jespersen
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, NM 87545, United States
| | | | - Ankush Singhal
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, NM 87545, United States
| | - Karissa Y Sanbonmatsu
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, NM 87545, United States
- New Mexico Consortium, Los Alamos, NM 87544, United States
| |
Collapse
|
37
|
Bürmann F, Clifton B, Koekemoer S, Wilkinson OJ, Kimanius D, Dillingham MS, Löwe J. Mechanism of DNA capture by the MukBEF SMC complex and its inhibition by a viral DNA mimic. Cell 2025; 188:2465-2479.e14. [PMID: 40168993 DOI: 10.1016/j.cell.2025.02.032] [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: 10/16/2024] [Revised: 12/18/2024] [Accepted: 02/26/2025] [Indexed: 04/03/2025]
Abstract
Ring-like structural maintenance of chromosome (SMC) complexes are crucial for genome organization and operate through mechanisms of DNA entrapment and loop extrusion. Here, we explore the DNA loading process of the bacterial SMC complex MukBEF. Using cryoelectron microscopy (cryo-EM), we demonstrate that ATP binding opens one of MukBEF's three potential DNA entry gates, exposing a DNA capture site that positions DNA at the open neck gate. We discover that the gp5.9 protein of bacteriophage T7 blocks this capture site by DNA mimicry, thereby preventing DNA loading and inactivating MukBEF. We propose a comprehensive and unidirectional loading mechanism in which DNA is first captured at the complex's periphery and then ingested through the DNA entry gate, powered by a single cycle of ATP hydrolysis. These findings illuminate a fundamental aspect of how ubiquitous DNA organizers are primed for genome maintenance and demonstrate how this process can be disrupted by viruses.
Collapse
Affiliation(s)
- Frank Bürmann
- MRC Laboratory of Molecular Biology, Structural Studies, Francis Crick Avenue, Cambridge CB2 0QH, UK; University of Oxford, Department of Biochemistry, South Parks Road, Oxford OX1 3QU, UK.
| | - Bryony Clifton
- University of Bristol, School of Biochemistry, DNA:Protein Interactions Unit, Bristol BS8 1TD, UK
| | - Sophie Koekemoer
- University of Bristol, School of Biochemistry, DNA:Protein Interactions Unit, Bristol BS8 1TD, UK
| | - Oliver J Wilkinson
- University of Bristol, School of Biochemistry, DNA:Protein Interactions Unit, Bristol BS8 1TD, UK
| | - Dari Kimanius
- MRC Laboratory of Molecular Biology, Structural Studies, Francis Crick Avenue, Cambridge CB2 0QH, UK; CZ Imaging Institute, 3400 Bridge Parkway, Redwood City, CA 94065, USA
| | - Mark S Dillingham
- University of Bristol, School of Biochemistry, DNA:Protein Interactions Unit, Bristol BS8 1TD, UK.
| | - Jan Löwe
- MRC Laboratory of Molecular Biology, Structural Studies, Francis Crick Avenue, Cambridge CB2 0QH, UK.
| |
Collapse
|
38
|
Shahid T, Danazumi AU, Tehseen M, Alhudhali L, Clark AR, Savva CG, Hamdan SM, De Biasio A. Structural dynamics of DNA unwinding by a replicative helicase. Nature 2025; 641:240-249. [PMID: 40108462 PMCID: PMC12043514 DOI: 10.1038/s41586-025-08766-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: 05/14/2024] [Accepted: 02/10/2025] [Indexed: 03/22/2025]
Abstract
Hexameric helicases are nucleotide-driven molecular machines that unwind DNA to initiate replication across all domains of life. Despite decades of intensive study, several critical aspects of their function remain unresolved1: the site and mechanism of DNA strand separation, the mechanics of unwinding propagation, and the dynamic relationship between nucleotide hydrolysis and DNA movement. Here, using cryo-electron microscopy (cryo-EM), we show that the simian virus 40 large tumour antigen (LTag) helicase assembles in the form of head-to-head hexamers at replication origins, melting DNA at two symmetrically positioned sites to establish bidirectional replication forks. Through continuous heterogeneity analysis2, we characterize the conformational landscape of LTag on forked DNA under catalytic conditions, demonstrating coordinated motions that drive DNA translocation and unwinding. We show that the helicase pulls the tracking strand through DNA-binding loops lining the central channel, while directing the non-tracking strand out of the rear, in a cyclic process. ATP hydrolysis functions as an 'entropy switch', removing blocks to translocation rather than directly powering DNA movement. Our structures show the allosteric couplings between nucleotide turnover and subunit motions that enable DNA unwinding while maintaining dedicated exit paths for the separated strands. These findings provide a comprehensive model for replication fork establishment and progression that extends from viral to eukaryotic systems. More broadly, they introduce fundamental principles of the mechanism by which ATP-dependent enzymes achieve efficient mechanical work through entropy-driven allostery.
Collapse
Affiliation(s)
- Taha Shahid
- Bioscience Program, Division of Biological and Environmental Sciences and Engineering, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
- Leicester Institute of Structural and Chemical Biology and Department of Molecular and Cell Biology, University of Leicester, Leicester, UK
| | - Ammar U Danazumi
- Bioscience Program, Division of Biological and Environmental Sciences and Engineering, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
| | - Muhammad Tehseen
- Bioscience Program, Division of Biological and Environmental Sciences and Engineering, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
| | - Lubna Alhudhali
- Bioscience Program, Division of Biological and Environmental Sciences and Engineering, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
| | - Alice R Clark
- Bioscience Program, Division of Biological and Environmental Sciences and Engineering, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
| | - Christos G Savva
- Bioscience Program, Division of Biological and Environmental Sciences and Engineering, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
| | - Samir M Hamdan
- Bioscience Program, Division of Biological and Environmental Sciences and Engineering, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia.
| | - Alfredo De Biasio
- Bioscience Program, Division of Biological and Environmental Sciences and Engineering, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia.
- Leicester Institute of Structural and Chemical Biology and Department of Molecular and Cell Biology, University of Leicester, Leicester, UK.
| |
Collapse
|
39
|
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
|
40
|
Armeev GA, Moiseenko AV, Motorin NA, Afonin DA, Zhao L, Vasilev VA, Oleinikov PD, Glukhov GS, Peters GS, Studitsky VM, Feofanov AV, Shaytan AK, Shi X, Sokolova OS. Structure and dynamics of a nucleosome core particle based on Widom 603 DNA sequence. Structure 2025; 33:948-959.e5. [PMID: 40101710 DOI: 10.1016/j.str.2025.02.007] [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: 07/22/2024] [Revised: 01/03/2025] [Accepted: 02/19/2025] [Indexed: 03/20/2025]
Abstract
Nucleosomes are fundamental elements of chromatin organization that participate in compacting genomic DNA and serve as targets for the binding of numerous regulatory proteins. Currently, over 500 different nucleosome structures are known. Despite the large number of nucleosome structures, all of them were formed on only about twenty different DNA sequences. Using cryo-electron microscopy, we determined the structure of the nucleosome formed on a high-affinity Widom 603 DNA sequence at 4 Å resolution; an atomic model was built. We proposed an integrative modeling approach to study the nucleosomal DNA unwrapping based on the cryoelectron microscopy (cryo-EM) data. We also demonstrated the DNA unwrapping of the Widom 603 nucleosome using small angle X-ray scattering and single particle Förster resonance energy transfer measurements. Our results are consistent with the asymmetry of nucleosomal DNA unwrapping. Our data revealed the dependence of nucleosome structure and dynamics on the sequence of nucleosomal DNA.
Collapse
Affiliation(s)
- Grigoriy A Armeev
- Department of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia.
| | - Andrey V Moiseenko
- Department of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Nikita A Motorin
- Department of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Dmitriy A Afonin
- Department of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia; Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Lei Zhao
- Faculty of Biology, Shenzhen MSU-BIT University, Shenzhen, Guangdong 518172, China
| | - Veniamin A Vasilev
- Department of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Pavel D Oleinikov
- Department of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Grigory S Glukhov
- Department of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia; Faculty of Biology, Shenzhen MSU-BIT University, Shenzhen, Guangdong 518172, China
| | - Georgy S Peters
- Faculty of Physics, Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Vasily M Studitsky
- Department of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia; Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Alexey V Feofanov
- Department of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia; Faculty of Biology, Shenzhen MSU-BIT University, Shenzhen, Guangdong 518172, China
| | - Alexey K Shaytan
- Department of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Xiangyan Shi
- Faculty of Biology, Shenzhen MSU-BIT University, Shenzhen, Guangdong 518172, China
| | - Olga S Sokolova
- Faculty of Biology, Shenzhen MSU-BIT University, Shenzhen, Guangdong 518172, China.
| |
Collapse
|
41
|
Banari A, Samanta AK, Munke A, Laugks T, Bajt S, Grünewald K, Marlovits TC, Küpper J, Maia FRNC, Chapman HN, Oberthür D, Seuring C. Advancing time-resolved structural biology: latest strategies in cryo-EM and X-ray crystallography. Nat Methods 2025:10.1038/s41592-025-02659-6. [PMID: 40312512 DOI: 10.1038/s41592-025-02659-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 03/11/2025] [Indexed: 05/03/2025]
Abstract
Structural biology offers a window into the functionality of molecular machines in health and disease. A fundamental challenge lies in capturing both the high-resolution structural details and dynamic changes that are essential for function. The high-resolution methods of X-ray crystallography and electron cryo-microscopy (cryo-EM) are mainly used to study ensembles of static conformations but can also capture crucial dynamic transition states. Here, we review the latest strategies and advancements in time-resolved structural biology with a focus on capturing dynamic changes. We describe recent technology developments for time-resolved sample preparation and delivery in the cryo-EM and X-ray fields and explore how these technologies could mutually benefit each other to advance our understanding of biology at the molecular and atomic scales.
Collapse
Affiliation(s)
- Amir Banari
- Centre for Structural Systems Biology, Hamburg, Germany
- The Hamburg Centre for Ultrafast Imaging, Hamburg, Germany
- Department of Physics, Universität Hamburg, Hamburg, Germany
| | - Amit K Samanta
- The Hamburg Centre for Ultrafast Imaging, Hamburg, Germany
- Center for Free-Electron Laser Science, Deutsches Elektronen-Synchrotron DESY, Hamburg, Germany
| | - Anna Munke
- Center for Free-Electron Laser Science, Deutsches Elektronen-Synchrotron DESY, Hamburg, Germany
- Laboratory of Molecular Biophysics, Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden
| | - Tim Laugks
- Centre for Structural Systems Biology, Hamburg, Germany
- Department of Chemistry, Universität Hamburg, Hamburg, Germany
| | - Saša Bajt
- The Hamburg Centre for Ultrafast Imaging, Hamburg, Germany
- Center for Free-Electron Laser Science, Deutsches Elektronen-Synchrotron DESY, Hamburg, Germany
| | - Kay Grünewald
- Centre for Structural Systems Biology, Hamburg, Germany
- The Hamburg Centre for Ultrafast Imaging, Hamburg, Germany
- Department of Chemistry, Universität Hamburg, Hamburg, Germany
- Department of Structural Cell Biology of Viruses, Leibniz Institute of Virology, Hamburg, Germany
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Thomas C Marlovits
- Centre for Structural Systems Biology, Hamburg, Germany
- University Medical Center Hamburg-Eppendorf (UKE), Institute of Microbial and Molecular Sciences, Hamburg, Germany
- Deutsches Elektronen-Synchrotron DESY, Hamburg, Germany
| | - Jochen Küpper
- The Hamburg Centre for Ultrafast Imaging, Hamburg, Germany
- Department of Physics, Universität Hamburg, Hamburg, Germany
- Center for Free-Electron Laser Science, Deutsches Elektronen-Synchrotron DESY, Hamburg, Germany
| | - Filipe R N C Maia
- Laboratory of Molecular Biophysics, Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden
| | - Henry N Chapman
- The Hamburg Centre for Ultrafast Imaging, Hamburg, Germany
- Department of Physics, Universität Hamburg, Hamburg, Germany
- Center for Free-Electron Laser Science, Deutsches Elektronen-Synchrotron DESY, Hamburg, Germany
| | - Dominik Oberthür
- Center for Free-Electron Laser Science, Deutsches Elektronen-Synchrotron DESY, Hamburg, Germany
| | - Carolin Seuring
- Centre for Structural Systems Biology, Hamburg, Germany.
- The Hamburg Centre for Ultrafast Imaging, Hamburg, Germany.
- Department of Chemistry, Universität Hamburg, Hamburg, Germany.
- Department of Structural Cell Biology of Viruses, Leibniz Institute of Virology, Hamburg, Germany.
| |
Collapse
|
42
|
Kumar Mondal A, Carrillo E, Jayaraman V, Twomey EC. Glutamate gating of AMPA-subtype iGluRs at physiological temperatures. Nature 2025; 641:788-796. [PMID: 40140570 PMCID: PMC12074995 DOI: 10.1038/s41586-025-08770-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 02/11/2025] [Indexed: 03/28/2025]
Abstract
Ionotropic glutamate receptors (iGluRs) are tetrameric ligand-gated ion channels that mediate most excitatory neurotransmission1. iGluRs are gated by glutamate, where on glutamate binding, they open their ion channels to enable cation influx into postsynaptic neurons, initiating signal transduction1,2. The structural mechanics of how glutamate gating occurs in full-length iGluRs is not well understood. Here, using the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid subtype iGluR (AMPAR), we identify the glutamate-gating mechanism. AMPAR activation by glutamate is augmented at physiological temperatures. By preparing AMPARs for cryogenic-electron microscopy at these temperatures, we captured the glutamate-gating mechanism. Activation by glutamate initiates ion channel opening that involves all ion channel helices hinging away from the pore axis in a motif that is conserved across all iGluRs. Desensitization occurs when the local dimer pairs decouple and enables closure of the ion channel below through restoring the channel hinges and refolding the channel gate. Our findings define how glutamate gates iGluRs, provide foundations for therapeutic design and demonstrate how physiological temperatures can alter iGluR function.
Collapse
Affiliation(s)
- Anish Kumar Mondal
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Elisa Carrillo
- Center for Membrane Biology, Department of Biochemistry and Molecular Biology, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Vasanthi Jayaraman
- Center for Membrane Biology, Department of Biochemistry and Molecular Biology, University of Texas Health Science Center at Houston, Houston, TX, USA.
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX, USA.
| | - Edward C Twomey
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- The Beckman Center for Cryo-EM at Johns Hopkins, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Diana Helis Henry Medical Research Foundation, New Orleans, LA, USA.
| |
Collapse
|
43
|
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
|
44
|
Faure G, Saito M, Wilkinson ME, Quinones-Olvera N, Xu P, Flam-Shepherd D, Kim S, Reddy N, Zhu S, Evgeniou L, Koonin EV, Macrae RK, Zhang F. TIGR-Tas: A family of modular RNA-guided DNA-targeting systems in prokaryotes and their viruses. Science 2025; 388:eadv9789. [PMID: 40014690 PMCID: PMC12045711 DOI: 10.1126/science.adv9789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 02/15/2025] [Indexed: 03/01/2025]
Abstract
RNA-guided systems provide remarkable versatility, enabling diverse biological functions. Through iterative structural and sequence homology-based mining starting with a guide RNA-interaction domain of Cas9, we identified a family of RNA-guided DNA-targeting proteins in phage and parasitic bacteria. Each system consists of a tandem interspaced guide RNA (TIGR) array and a TIGR-associated (Tas) protein containing a nucleolar protein (Nop) domain, sometimes fused to HNH (TasH)- or RuvC (TasR)-nuclease domains. We show that TIGR arrays are processed into 36-nucleotide RNAs (tigRNAs) that direct sequence-specific DNA binding through a tandem-spacer targeting mechanism. TasR can be reprogrammed for precise DNA cleavage, including in human cells. The structure of TasR reveals striking similarities to box C/D small nucleolar ribonucleoproteins and IS110 RNA-guided transposases, providing insights into the evolution of diverse RNA-guided systems.
Collapse
Affiliation(s)
- Guilhem Faure
- Broad Institute of MIT and Harvard; Cambridge, USA
- McGovern Institute for Brain Research at MIT; Cambridge, USA
- Department of Brain and Cognitive Science, Massachusetts Institute of Technology; Cambridge, USA
- Department of Biological Engineering, Massachusetts Institute of Technology; Cambridge, USA
- Howard Hughes Medical Institute; Cambridge, USA
| | - Makoto Saito
- Broad Institute of MIT and Harvard; Cambridge, USA
- McGovern Institute for Brain Research at MIT; Cambridge, USA
- Department of Brain and Cognitive Science, Massachusetts Institute of Technology; Cambridge, USA
- Department of Biological Engineering, Massachusetts Institute of Technology; Cambridge, USA
- Howard Hughes Medical Institute; Cambridge, USA
| | - Max E. Wilkinson
- Broad Institute of MIT and Harvard; Cambridge, USA
- McGovern Institute for Brain Research at MIT; Cambridge, USA
- Department of Brain and Cognitive Science, Massachusetts Institute of Technology; Cambridge, USA
- Department of Biological Engineering, Massachusetts Institute of Technology; Cambridge, USA
- Howard Hughes Medical Institute; Cambridge, USA
| | - Natalia Quinones-Olvera
- Broad Institute of MIT and Harvard; Cambridge, USA
- McGovern Institute for Brain Research at MIT; Cambridge, USA
- Department of Brain and Cognitive Science, Massachusetts Institute of Technology; Cambridge, USA
- Department of Biological Engineering, Massachusetts Institute of Technology; Cambridge, USA
- Howard Hughes Medical Institute; Cambridge, USA
| | - Peiyu Xu
- Broad Institute of MIT and Harvard; Cambridge, USA
- McGovern Institute for Brain Research at MIT; Cambridge, USA
- Department of Brain and Cognitive Science, Massachusetts Institute of Technology; Cambridge, USA
- Department of Biological Engineering, Massachusetts Institute of Technology; Cambridge, USA
- Howard Hughes Medical Institute; Cambridge, USA
| | - Daniel Flam-Shepherd
- Broad Institute of MIT and Harvard; Cambridge, USA
- McGovern Institute for Brain Research at MIT; Cambridge, USA
- Department of Brain and Cognitive Science, Massachusetts Institute of Technology; Cambridge, USA
- Department of Biological Engineering, Massachusetts Institute of Technology; Cambridge, USA
- Howard Hughes Medical Institute; Cambridge, USA
| | - Stephanie Kim
- Broad Institute of MIT and Harvard; Cambridge, USA
- McGovern Institute for Brain Research at MIT; Cambridge, USA
- Department of Brain and Cognitive Science, Massachusetts Institute of Technology; Cambridge, USA
- Department of Biological Engineering, Massachusetts Institute of Technology; Cambridge, USA
- Howard Hughes Medical Institute; Cambridge, USA
| | - Nishith Reddy
- Broad Institute of MIT and Harvard; Cambridge, USA
- McGovern Institute for Brain Research at MIT; Cambridge, USA
- Department of Brain and Cognitive Science, Massachusetts Institute of Technology; Cambridge, USA
- Department of Biological Engineering, Massachusetts Institute of Technology; Cambridge, USA
- Howard Hughes Medical Institute; Cambridge, USA
| | - Shiyou Zhu
- Broad Institute of MIT and Harvard; Cambridge, USA
- McGovern Institute for Brain Research at MIT; Cambridge, USA
- Department of Brain and Cognitive Science, Massachusetts Institute of Technology; Cambridge, USA
- Department of Biological Engineering, Massachusetts Institute of Technology; Cambridge, USA
- Howard Hughes Medical Institute; Cambridge, USA
| | - Lilia Evgeniou
- Broad Institute of MIT and Harvard; Cambridge, USA
- McGovern Institute for Brain Research at MIT; Cambridge, USA
- Department of Brain and Cognitive Science, Massachusetts Institute of Technology; Cambridge, USA
- Department of Biological Engineering, Massachusetts Institute of Technology; Cambridge, USA
- Howard Hughes Medical Institute; Cambridge, USA
- Department of Systems Biology, Harvard University; Boston, USA
| | - Eugene V. Koonin
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, USA
| | - Rhiannon K. Macrae
- Broad Institute of MIT and Harvard; Cambridge, USA
- McGovern Institute for Brain Research at MIT; Cambridge, USA
- Department of Brain and Cognitive Science, Massachusetts Institute of Technology; Cambridge, USA
- Department of Biological Engineering, Massachusetts Institute of Technology; Cambridge, USA
- Howard Hughes Medical Institute; Cambridge, USA
| | - Feng Zhang
- Broad Institute of MIT and Harvard; Cambridge, USA
- McGovern Institute for Brain Research at MIT; Cambridge, USA
- Department of Brain and Cognitive Science, Massachusetts Institute of Technology; Cambridge, USA
- Department of Biological Engineering, Massachusetts Institute of Technology; Cambridge, USA
- Howard Hughes Medical Institute; Cambridge, USA
| |
Collapse
|
45
|
Stevens A, Kashyap S, Crofut EH, Wang SE, Muratore KA, Johnson PJ, Zhou ZH. Structures of Native Doublet Microtubules from Trichomonas vaginalis Reveal Parasite-Specific Proteins. Nat Commun 2025; 16:3996. [PMID: 40301421 PMCID: PMC12041511 DOI: 10.1038/s41467-025-59369-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 04/21/2025] [Indexed: 05/01/2025] Open
Abstract
Doublet microtubules (DMTs) are flagellar components required for the protist Trichomonas vaginalis (Tv) to swim through the human genitourinary tract to cause trichomoniasis, the most common non-viral sexually transmitted disease. Lack of structures of Tv's DMT (Tv-DMT) has prevented structure-guided drug design to manage Tv infection. Here, we determine the 16 nm, 32 nm, 48 nm and 96 nm-repeat structures of native Tv-DMT at resolution ranging from 3.4 to 4.4 Å by cryogenic electron microscopy (cryoEM) and built an atomic model for the entire Tv-DMT. These structures show that Tv-DMT is composed of 30 different proteins, including the α- and β-tubulin, 19 microtubule inner proteins (MIPs) and 9 microtubule outer proteins. While the A-tubule of Tv-DMT is simplistic compared to DMTs of other organisms, the B-tubule of Tv-DMT features parasite-specific proteins, such as TvFAP40 and TvFAP35. Notably, TvFAP40 and TvFAP35 form filaments near the inner and outer junctions, respectively, and interface with stabilizing MIPs. This atomic model of the Tv-DMT highlights diversity of eukaryotic motility machineries and provides a structural framework to inform rational design of therapeutics against trichomoniasis.
Collapse
Affiliation(s)
- Alexander Stevens
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
- California NanoSystems Institute, UCLA, Los Angeles, CA, USA
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, CA, USA
| | - Saarang Kashyap
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
- California NanoSystems Institute, UCLA, Los Angeles, CA, USA
| | - Ethan H Crofut
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
- California NanoSystems Institute, UCLA, Los Angeles, CA, USA
| | - Shuqi E Wang
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Katherine A Muratore
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Patricia J Johnson
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles (UCLA), Los Angeles, CA, USA.
| | - Z Hong Zhou
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles (UCLA), Los Angeles, CA, USA.
- California NanoSystems Institute, UCLA, Los Angeles, CA, USA.
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, CA, USA.
| |
Collapse
|
46
|
Draper-Barr G, Defelipe LA, Ruiz-Carrillo D, Gustavsson E, Landau M, García-Alai M. Sla2 is a core interaction hub for clathrin light chain and the Pan1/End3/Sla1 complex. Structure 2025:S0969-2126(25)00147-9. [PMID: 40347949 DOI: 10.1016/j.str.2025.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 03/25/2025] [Accepted: 04/15/2025] [Indexed: 05/14/2025]
Abstract
The interaction network of Sla2, a vital endocytic mid-coat adaptor protein, undergoes constant rearrangement. Sla2 serves as a scaffold linking the membrane to the actin cytoskeleton, with its role modulated by the clathrin light chain (CLC), which inhibits Sla2's function under certain conditions. We show that Sla2 has two independent binding sites for CLC: one previously described in homologs of fungi (Sla2) and metazoa (Hip1R), and a second found only in Fungi. We present the structural model of the Sla2 actin-binding domains in the context of regulatory structural domains by cryoelectron microscopy. We provide an interaction map of Sla2 and the regulatory proteins Sla1 and Pan1, predicted by AI modeling and confirmed by molecular biophysics techniques. Pan1 may compete with CLC for the conserved Sla2-binding site. These results enhance the mapping of crucial interactions at endocytic checkpoints and highlight the divergence between Metazoa and Fungi in this vital process.
Collapse
Affiliation(s)
- George Draper-Barr
- European Molecular Biology Laboratory, DESY, Building 25a, Hamburg 22607, Germany; Centre for Structural Systems Biology (CSSB), DESY, Building 15, Hamburg 22607, Germany
| | - Lucas A Defelipe
- European Molecular Biology Laboratory, DESY, Building 25a, Hamburg 22607, Germany; Centre for Structural Systems Biology (CSSB), DESY, Building 15, Hamburg 22607, Germany
| | - David Ruiz-Carrillo
- European Molecular Biology Laboratory, DESY, Building 25a, Hamburg 22607, Germany; Centre for Structural Systems Biology (CSSB), DESY, Building 15, Hamburg 22607, Germany
| | - Emil Gustavsson
- Centre for Structural Systems Biology (CSSB), DESY, Building 15, Hamburg 22607, Germany
| | - Meytal Landau
- European Molecular Biology Laboratory, DESY, Building 25a, Hamburg 22607, Germany; Centre for Structural Systems Biology (CSSB), DESY, Building 15, Hamburg 22607, Germany; University Medical Center Hamburg-Eppendorf, Martinistraße 52, Hamburg 20251, Germany; Department of Biology, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Maria García-Alai
- European Molecular Biology Laboratory, DESY, Building 25a, Hamburg 22607, Germany; Centre for Structural Systems Biology (CSSB), DESY, Building 15, Hamburg 22607, Germany.
| |
Collapse
|
47
|
Ghanim GE, Hu H, Boulanger J, Nguyen THD. Structural mechanism of LINE-1 target-primed reverse transcription. Science 2025; 388:eads8412. [PMID: 40048554 DOI: 10.1126/science.ads8412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 02/11/2025] [Indexed: 04/26/2025]
Abstract
Long interspersed element-1 (LINE-1) retrotransposons are the only active autonomous transposable elements in humans. They propagate by reverse transcribing their messenger RNA into new genomic locations by a process called target-primed reverse transcription (TPRT). In this work, we present four cryo-electron microscopy structures of the human LINE-1 TPRT complex, revealing the conformational dynamics of open reading frame 2 protein (ORF2p) and its extensive remodeling of the target DNA for TPRT initiation. We observe nicking of the DNA second strand during reverse transcription of the first strand. Structure prediction identifies high-confidence binding sites for LINE-1-associated factors-namely proliferating cell nuclear antigen (PCNA) and cytoplasmic poly(A)-binding protein 1 (PABPC1)-on ORF2p. Together with our structural data, this suggests a mechanism by which these factors regulate retrotransposition and supports a model for TPRT that accounts for retrotransposition outcomes observed in cells.
Collapse
Affiliation(s)
| | - Hongmiao Hu
- MRC Laboratory of Molecular Biology, Cambridge, UK
| | | | | |
Collapse
|
48
|
Yen IY, Whitfield GB, Rubinstein JL, Burrows LL, Brun YV, Howell PL. Conformational changes in the motor ATPase CpaF facilitate a rotary mechanism of Tad pilus assembly. Nat Commun 2025; 16:3839. [PMID: 40268890 PMCID: PMC12019362 DOI: 10.1038/s41467-025-59009-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 04/08/2025] [Indexed: 04/25/2025] Open
Abstract
The type IV pilus family uses PilT/VirB11-like ATPases to rapidly assemble and disassemble pilin subunits. Among these, the tight adherence (Tad) pilus performs both functions using a single bifunctional ATPase, CpaF. Here, we determine three conformationally distinct structures of CpaF hexamers with varying nucleotide occupancies by cryo-electron microscopy. Analysis of these structures suggest ATP binding and hydrolysis expand and rotate the hexamer pore clockwise while subsequent ADP release contracts the ATPase. Truncation of the intrinsically disordered region of CpaF in Caulobacter crescentus equally reduces pilus extension and retraction events observed using fluorescence microscopy, but does not reduce ATPase activity. AlphaFold3 modeling suggests that CpaF and other motors of the type IV filament superfamily employ conserved secondary structural features to engage their respective platform proteins. From these data, we propose that CpaF uses a clockwise, rotary mechanism of catalysis to assemble a right-handed, helical Tad pilus, a process broadly applicable to other single motor systems.
Collapse
Affiliation(s)
- Ian Y Yen
- Program in Molecular Medicine, Peter Gilgan Center for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Gregory B Whitfield
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC, Canada
| | - John L Rubinstein
- Program in Molecular Medicine, Peter Gilgan Center for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Lori L Burrows
- Biochemistry and Biomedical Sciences and the Michael G. DeGroote Centre for Infectious Disease Research, McMaster University, Hamilton, ON, Canada.
| | - Yves V Brun
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC, Canada.
| | - P Lynne Howell
- Program in Molecular Medicine, Peter Gilgan Center for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada.
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
49
|
Sherry J, Pawar KI, Dolat L, Smith E, Chang IC, Pha K, Kaake R, Swaney DL, Herrera C, McMahon E, Bastidas RJ, Johnson JR, Valdivia RH, Krogan NJ, Elwell CA, Verba K, Engel JN. The Chlamydia effector Dre1 binds dynactin to reposition host organelles during infection. Cell Rep 2025; 44:115509. [PMID: 40186871 DOI: 10.1016/j.celrep.2025.115509] [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: 08/27/2024] [Revised: 01/09/2025] [Accepted: 03/12/2025] [Indexed: 04/07/2025] Open
Abstract
The obligate intracellular pathogen Chlamydia trachomatis replicates in a specialized membrane-bound compartment where it repositions host organelles during infection to acquire nutrients and evade host surveillance. We describe a bacterial effector, Dre1, that binds specifically to dynactin associated with host microtubule organizing centers without globally impeding dynactin function. Dre1 is required to reposition the centrosome, mitotic spindle, Golgi apparatus, and primary cilia around the inclusion and contributes to pathogen fitness in cell-based and mouse models of infection. We utilized Dre1 to affinity purify the megadalton dynactin protein complex and determined the first cryoelectron microscopy (cryo-EM) structure of human dynactin. Our results suggest that Dre1 binds to the pointed end of dynactin and uncovers the first bacterial effector that modulates dynactin function. Our work highlights how a pathogen employs a single effector to evoke targeted, large-scale changes in host cell organization that facilitate pathogen growth without inhibiting host viability.
Collapse
Affiliation(s)
- Jessica Sherry
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Komal Ishwar Pawar
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Lee Dolat
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Erin Smith
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - I-Chang Chang
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Khavong Pha
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Robyn Kaake
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Danielle L Swaney
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Clara Herrera
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Eleanor McMahon
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Robert J Bastidas
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Jeffrey R Johnson
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Raphael H Valdivia
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Nevan J Krogan
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Cherilyn A Elwell
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA.
| | - Kliment Verba
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94143, USA.
| | - Joanne N Engel
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
50
|
Matsushita D, Toyoda Y, Lee Y, Aoi M, Matsuo H, Takada T, Nishizawa T. Structural basis of urate transport by glucose transporter 9. Cell Rep 2025; 44:115514. [PMID: 40186864 DOI: 10.1016/j.celrep.2025.115514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 01/17/2025] [Accepted: 03/13/2025] [Indexed: 04/07/2025] Open
Abstract
Glucose transporter 9 (GLUT9) is a critical urate transporter involved in renal reabsorption, playing a pivotal role in regulating physiological urate levels and representing a potential therapeutic target for gout. Despite such clinical significance, the structural basis of urate recognition and transport by GLUT9 remains elusive. Here, we present the cryoelectron microscopy (cryo-EM) structures of GLUT9 in the inward-open conformation in both apo and urate-bound states. Urate binds in a cleft between the N-terminal and C-terminal domains, interacting via hydrogen bonds and hydrophobic interactions. Structural comparison with sugar-transporting GLUTs highlights unique amino acid compositions in the substrate recognition pocket of GLUT9. Functional and mutational studies directly measuring GLUT9-mediated urate uptake further demonstrate the cooperative roles of multiple residues in urate recognition. Our findings elucidate the structural basis of urate transport by GLUT9 and provide valuable insights for the development of uricosuric drugs targeting GLUT9.
Collapse
Affiliation(s)
- Daiki Matsushita
- Graduate School of Medical Life Science, Yokohama City University, Yokohama 230-0045, Japan
| | - Yu Toyoda
- Department of Pharmacy, The University of Tokyo Hospital, Tokyo 113-8655, Japan; Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa 359-8513, Japan
| | - Yongchan Lee
- Graduate School of Medical Life Science, Yokohama City University, Yokohama 230-0045, Japan
| | - Maeda Aoi
- Graduate School of Medical Life Science, Yokohama City University, Yokohama 230-0045, Japan
| | - Hirotaka Matsuo
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa 359-8513, Japan
| | - Tappei Takada
- Department of Pharmacy, The University of Tokyo Hospital, Tokyo 113-8655, Japan
| | - Tomohiro Nishizawa
- Graduate School of Medical Life Science, Yokohama City University, Yokohama 230-0045, Japan.
| |
Collapse
|