1
|
Rivalta A, Fedorenko A, Le Scornet A, Thompson S, Halfon Y, Breiner Goldstein E, Çavdaroglu S, Melenitzky T, Hiregange DG, Zimmerman E, Bashan A, Yap MNF, Yonath A. Structural studies on ribosomes of differentially macrolide-resistant Staphylococcus aureus strains. Life Sci Alliance 2025; 8:e202503325. [PMID: 40490363 DOI: 10.26508/lsa.202503325] [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: 03/25/2025] [Revised: 05/06/2025] [Accepted: 05/07/2025] [Indexed: 06/11/2025] Open
Abstract
Antimicrobial resistance is a major global health challenge, diminishing the efficacy of many antibiotics, including macrolides. In Staphylococcus aureus, an opportunistic pathogen, macrolide resistance is primarily mediated by Erm-family methyltransferases, which mono- or dimethylate A2058 in the 23S ribosomal RNA, reducing drug binding. Although macrolide-ribosome interactions have been characterized in nonpathogenic species, their structural basis in clinically relevant pathogens remains limited. In this study, we investigate the impact of ermB-mediated resistance on drug binding by analyzing ribosomes from S. aureus strains with varying levels of ermB expression and activity. Using cryo-electron microscopy, we determined the high-resolution structures of solithromycin-bound ribosomes, including those with dimethylated A2058. Our structural analysis reveals the specific interactions that enable solithromycin binding despite double methylation and resistance, as corroborated by microbiological and biochemical data, suggesting that further optimization of ketolide-ribosome interactions could enhance macrolide efficacy against resistant S. aureus strains.
Collapse
Affiliation(s)
- André Rivalta
- Department of Chemical and Structural Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Aliza Fedorenko
- Department of Chemical and Structural Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Alexandre Le Scornet
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Sophie Thompson
- Department of Chemical and Structural Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Yehuda Halfon
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK
| | | | - Sude Çavdaroglu
- Department of Emergency Medicine, Arnavutköy State Hospital, Istanbul, Turkey
| | - Tal Melenitzky
- Department of Chemical and Structural Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Disha-Gajanan Hiregange
- Department of Chemical and Structural Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Ella Zimmerman
- Department of Chemical and Structural Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Anat Bashan
- Department of Chemical and Structural Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Mee-Ngan Frances Yap
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Ada Yonath
- Department of Chemical and Structural Biology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
2
|
Jiang L, Zhu B, Long W, Xu J, Wu Y, Li YW. A review of denoising methods in single-particle cryo-EM. Micron 2025; 194:103817. [PMID: 40164016 DOI: 10.1016/j.micron.2025.103817] [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: 12/09/2024] [Revised: 02/08/2025] [Accepted: 03/11/2025] [Indexed: 04/02/2025]
Abstract
Cryo-EM has become a vital technique for resolving macromolecular structures at near-atomic resolution, enabling the visualization of proteins and large molecular complexes. However, the images are often accompanied by extremely low SNR, which poses significant challenges for subsequent processes such as particle picking and 3D reconstruction. Effective denoising methods can substantially improve SNR, making downstream analyzes more accurate and reliable. Thus, image denoising is an essential step in cryo-EM data processing. This paper comprehensively reviews recent advances in image denoising methods for single-particle analysis, covering approaches from traditional filtering methods to the latest deep learning-based strategies. By analyzing and comparing mainstream denoising methods, this review aims to advance the field of single-particle cryo-EM denoising, facilitate the acquisition of higher-quality images, and offer valuable insights for researchers new to the field.
Collapse
Affiliation(s)
- Linhua Jiang
- School of Information Engineering, Huzhou University, Huzhou, China; ISEP-Sorbonne Joint Research Lab, 10 Rue de Vanves, Paris 92130, France.
| | - Bo Zhu
- School of Information Engineering, Huzhou University, Huzhou, China.
| | - Wei Long
- School of Information Engineering, Huzhou University, Huzhou, China.
| | - Jiahao Xu
- School of Information Engineering, Huzhou University, Huzhou, China.
| | - Yi Wu
- School of Information Engineering, Huzhou University, Huzhou, China.
| | - Yao-Wang Li
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, China.
| |
Collapse
|
3
|
Gupta R, Sharma S, Saroj A, Madhukalya R, Kumar V, Agarwal V, Kumar D, Mangala Prasad V, Kumar R. Kyasanur Forest disease virus non-structural protein NS1 forms multimers in solution, with a distinctly identifiable tetrameric state. Biochimie 2025; 234:89-94. [PMID: 40252820 DOI: 10.1016/j.biochi.2025.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Revised: 03/30/2025] [Accepted: 04/17/2025] [Indexed: 04/21/2025]
Abstract
Kyasanur Forest Disease Virus (KFDV), a flavivirus, is predominantly present in the tropical region of southern India and is responsible for viral hemorrhagic disease in primates and non-primate animals. KFDV infection is spread by tick bites. The other medically important viruses of Flaviviridae family are dengue (DENV), Zika (ZIKV), West Nile virus (WNV) and Japanese encephalitis virus (JEV). The flaviviruses are collectively responsible for diverse disease pathologies and account for a major global health burden. A major contributing factor to disease pathogenesis of flavivirus is the secreted form of non-structural protein 1 (NS1). However, in vivo studies using lethal flavivirus challenge have demonstrated the protective role of NS1-specific antibodies and complement the hypothesis to explore possibilities of NS1-based vaccine and therapeutic candidates. Recent structural studies on DENV, ZIKV, JEV and WNV NS1 antigen have shown that the sNS1 protein exists in high-order oligomeric states. However, structural insights about the high-order oligomeric states of sNS1 of tick-borne flaviviruses and their biological significance are poorly explored. In this study, we have expressed and purified the KFDV NS1 protein in the mammalian expression system. The KFDV sNS1 protein exhibits higher oligomeric conformation in solution as determined by size exclusion chromatography (SEC), and negative stain transmission electron microscopy (NS-TEM). Single-particle analysis of KFDV NS1 reveals tetrameric arrangements that are structurally similar to previously reported NS1 structures from other flaviviruses. Our study will help to develop a future roadmap of the rational design of broad-spectrum anti-NS1 antibodies and subunit vaccines effective against tick-borne flaviviruses.
Collapse
Affiliation(s)
- Rohit Gupta
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Roorkee, Uttarakhand, India
| | - Shruti Sharma
- Molecular Biophysics Unit, Indian Institute of Science, Bengaluru, Karnataka, India; Center for Infectious Diseases Research, Indian Institute of Science, Bengaluru, Karnataka, India
| | - Anjali Saroj
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Roorkee, Uttarakhand, India
| | - Rishav Madhukalya
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Roorkee, Uttarakhand, India
| | - Vivek Kumar
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Roorkee, Uttarakhand, India
| | - Vidushi Agarwal
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Roorkee, Uttarakhand, India
| | - Dilip Kumar
- Trivedi School of Biosciences, Ashoka University, Sonipat, Haryana, India
| | - Vidya Mangala Prasad
- Molecular Biophysics Unit, Indian Institute of Science, Bengaluru, Karnataka, India; Center for Infectious Diseases Research, Indian Institute of Science, Bengaluru, Karnataka, India.
| | - Rajesh Kumar
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Roorkee, Uttarakhand, India.
| |
Collapse
|
4
|
Morris DJ, Gorman J, Zhou T, Lora J, Connell AJ, Li H, Liu W, Roark RS, Campion MS, Carey JW, Habib R, Li Y, Martella CL, Park Y, Singh A, Sowers KJ, Teng IT, Wang S, Chohan N, Ding W, Lauer C, Lewis E, Mason RD, Rando JM, Peyton L, Schramm CA, Wagh K, Korber B, Seaman MS, Douek DC, Haynes BF, Kulp DW, Roederer M, Hahn BH, Kwong PD, Shaw GM. Transient glycan shield reduction induces CD4-binding site broadly neutralizing antibodies in SHIV-infected macaques. Cell Rep 2025; 44:115848. [PMID: 40516049 DOI: 10.1016/j.celrep.2025.115848] [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: 12/19/2024] [Revised: 04/09/2025] [Accepted: 05/26/2025] [Indexed: 06/16/2025] Open
Abstract
Broadly neutralizing antibodies (bNAbs) targeting the HIV-1 CD4-binding site (CD4bs) occur infrequently in macaques and humans and have not been reproducibly elicited in any outbred animal model. To address this challenge, we first isolated RHA10, an infection-induced rhesus bNAb with 51% breadth. The cryoelectron microscopy (cryo-EM) structure of RHA10 with the HIV-1 envelope (Env) resembled prototypic human CD4bs bNAbs with CDR-H3-dominated binding. Env-antibody co-evolution revealed transient elimination of two Env CD4bs-proximal glycans near the time of RHA10-lineage initiation, and these glycan-deficient Envs bound preferentially to early RHA10 intermediates, suggesting that glycan deletions in infecting SHIVs could induce CD4bs bNAbs. To test this hypothesis, we constructed SHIV.CH505 variants with CD4bs-proximal glycan deletions. Infection of 11 macaques resulted in accelerated CD4bs bNAb responses in 9 compared with 1 of 115 control macaques. Glycan hole-based immunofocusing coupled to Env-Ab co-evolution can consistently induce broad CD4bs responses in macaques and serve as a model for HIV vaccine design.
Collapse
Affiliation(s)
- Daniel J Morris
- Departments of Medicine and Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jason Gorman
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA; Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Tongqing Zhou
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jinery Lora
- Departments of Medicine and Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Andrew J Connell
- Departments of Medicine and Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Hui Li
- Departments of Medicine and Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Weimin Liu
- Departments of Medicine and Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ryan S Roark
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Mary S Campion
- Departments of Medicine and Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - John W Carey
- Departments of Medicine and Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rumi Habib
- Departments of Medicine and Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, USA
| | - Yingying Li
- Departments of Medicine and Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Christian L Martella
- Departments of Medicine and Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Younghoon Park
- Departments of Medicine and Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ajay Singh
- Departments of Medicine and Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kirsten J Sowers
- Departments of Medicine and Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - I-Ting Teng
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Shuyi Wang
- Departments of Medicine and Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Neha Chohan
- Departments of Medicine and Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Wenge Ding
- Departments of Medicine and Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Craig Lauer
- Departments of Medicine and Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Emily Lewis
- Departments of Medicine and Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rosemarie D Mason
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Juliette M Rando
- Departments of Medicine and Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lowrey Peyton
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Chaim A Schramm
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Kshitij Wagh
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | | | - Michael S Seaman
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Daniel C Douek
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Barton F Haynes
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA; Departments of Immunology and Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Daniel W Kulp
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, USA
| | - Mario Roederer
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Beatrice H Hahn
- Departments of Medicine and Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Peter D Kwong
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA; Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA.
| | - George M Shaw
- Departments of Medicine and Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
5
|
Liu Y, Liao M. Conformational cycle and small-molecule inhibition mechanism of a plant ABCB transporter in lipid membranes. SCIENCE ADVANCES 2025; 11:eadv9721. [PMID: 40512840 PMCID: PMC12164952 DOI: 10.1126/sciadv.adv9721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 05/09/2025] [Indexed: 06/16/2025]
Abstract
In plants, ATP-binding cassette (ABC) transporters are crucial for nutrient uptake, phytohormone transport, and environmental response. It is of great interest to understand the mechanisms of these transporters and develop small-molecule modulators to regulate plant growth. Arabidopsis ABCB19 was recently shown to transport brassinosteroid, shaping hormone dynamics and plant architecture. However, the conformational cycle and inhibitor mechanism of ABCB transporters remain elusive. We reconstituted ABCB19 into lipid nanodiscs, where activity was drastically higher than in detergents, and determined its cryo-electron microscopy structures in substrate-free, substrate-bound, vanadate-trapped, and inhibitor-bound states. Inward-facing ABCB19 moved inward upon substrate binding and fully closed with vanadate trapping, unexpectedly temperature dependent. Two inhibitor molecules locked ABCB19 in the inward-facing conformation. Mutagenesis identified key residues for substrate and inhibitor binding, revealing differential contributions to transporter function and inhibition. These results deepen knowledge of plant ABCB transporters, laying a foundation for targeted manipulation to enhance plant resilience and productivity.
Collapse
Affiliation(s)
- Yong Liu
- Department of Chemical Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Maofu Liao
- Department of Chemical Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
- Institute for Biological Electron Microscopy, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
6
|
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
|
7
|
Dingeldein L, Silva-Sánchez D, Evans L, D'Imprima E, Grigorieff N, Covino R, Cossio P. Amortized template matching of molecular conformations from cryoelectron microscopy images using simulation-based inference. Proc Natl Acad Sci U S A 2025; 122:e2420158122. [PMID: 40465628 DOI: 10.1073/pnas.2420158122] [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: 10/01/2024] [Accepted: 04/30/2025] [Indexed: 06/18/2025] Open
Abstract
Characterizing the conformational ensemble of biomolecular systems is key to understand their functions. Cryoelectron microscopy (cryo-EM) captures two-dimensional snapshots of biomolecular ensembles, giving in principle access to thermodynamics. However, these images are very noisy and show projections of the molecule in unknown orientations, making it very difficult to identify the biomolecule's conformation in each individual image. Here, we introduce cryo-EM simulation-based inference (cryoSBI) to infer the conformations of biomolecules and the uncertainties associated with the inference from individual cryo-EM images. CryoSBI builds on simulation-based inference, a merger of physics-based simulations and probabilistic deep learning, allowing us to use Bayesian inference even when likelihoods are too expensive to calculate. We begin with an ensemble of conformations, templates from experiments, and molecular modeling, serving as structural hypotheses. We train a neural network approximating the Bayesian posterior using simulated images from these templates and then use it to accurately infer the conformation of the biomolecule from each experimental image. Training is only done once on simulations, and after that, it takes just a few milliseconds to make inference on an image, making cryoSBI suitable for arbitrarily large datasets and direct analysis on micrographs. CryoSBI eliminates the need to estimate particle pose and imaging parameters, significantly enhancing the computational speed compared to explicit likelihood methods. Importantly, we obtain interpretable machine learning models by integrating physics-based approaches with deep neural networks, ensuring that our results are transparent and reliable. We illustrate and benchmark cryoSBI on synthetic data and showcase its promise on experimental single-particle cryo-EM data.
Collapse
Affiliation(s)
- Lars Dingeldein
- Institute of Physics, Faculty of Physics, Goethe University Frankfurt, Frankfurt am Main 60438, Germany
- Frankfurt Institute for Advanced Studies, Frankfurt am Main 60438, Germany
| | | | - Luke Evans
- Center for Computational Mathematics, Flatiron Institute, New York, NY 10010
| | - Edoardo D'Imprima
- Istituto di Ricovero e Cura a Carattere Scientifico Humanitas Research Hospital, Unit of Correlative Light Emission Microscopy Core, Rozzano, Milan 20089, Italy
| | - Nikolaus Grigorieff
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, MA 01605
- HHMI, University of Massachusetts Chan Medical School, Worcester, MA 01605
| | - Roberto Covino
- Frankfurt Institute for Advanced Studies, Frankfurt am Main 60438, Germany
- Institute of Computer Science, Faculty of Computer Science and Mathematics, Goethe University Frankfurt, Frankfurt am Main 60325, Germany
| | - Pilar Cossio
- Center for Computational Mathematics, Flatiron Institute, New York, NY 10010
- Center for Computational Biology, Flatiron Institute, New York, NY 10010
| |
Collapse
|
8
|
Liu X, Krezel M, Wang W. Mechanism of human α3β GlyR regulation by intracellular M3/M4 loop phosphorylation and 2,6-di-tert-butylphenol interaction. Nat Commun 2025; 16:5242. [PMID: 40473619 PMCID: PMC12141631 DOI: 10.1038/s41467-025-60516-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 05/24/2025] [Indexed: 06/11/2025] Open
Abstract
α3β glycine receptor (GlyR) is a subtype of GlyRs that belongs to the Cys-loop receptor superfamily. It is highly expressed in the spinal dorsal horn where sensory information is integrated. Under inflammatory conditions, the large unstructured intracellular M3/M4 loops of the α3 subunit are phosphorylated through the prostaglandin E2 (PGE2) pathway, inhibiting ion conduction, and resulting in elevated pain sensation. A small molecule analgesic analog, 2,6-di-tert-butylphenol (2,6-DTBP) potentiates phosphorylated α3β GlyR through unclear mechanisms and relieves pain. Combining cryo-Electron Microscopy (cryo-EM) structures and single molecule Förster resonance energy transfer (smFRET) experiments, we show compaction of M3/M4 loop towards the ion conduction pore upon phosphorylation and further by 2,6-DTBP binding, which in turn modulates function through changing pore conformations and local electrostatics. We show that simultaneous interactions with the M3/M4 loop and the transmembrane domain (TM) is necessary for the potentiation of heteromeric α3β GlyR by 2,6-DTBP, while TM interaction alone is sufficient to potentiate homomeric α3 GlyR, explaining the mystery of why 2,6-DTBP potentiates only phosphorylated α3β GlyR. These findings show how post-translational modification of the unstructured intracellular M3/M4 loop may regulate Cys-loop receptor function, providing new perspectives in pain control and other pharmaceutical development targeting GlyRs and other Cys-loop receptors.
Collapse
Affiliation(s)
- Xiaofen Liu
- Departments of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Malgorzata Krezel
- Departments of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Weiwei Wang
- Departments of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
9
|
Lai-Kee-Him J, Trapani S, Boissinot S, Reinbold C, Fallet C, Ancelin A, Lecorre F, Hoh F, Ziegler-Graff V, Brault V, Bron P. Structure of the turnip yellows virus particles. Virology 2025; 607:110514. [PMID: 40179450 DOI: 10.1016/j.virol.2025.110514] [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: 11/28/2024] [Revised: 03/18/2025] [Accepted: 03/25/2025] [Indexed: 04/05/2025]
Abstract
Turnip yellows virus (TuYV) is a plant virus infecting important crops such as oilseed rape. TuYV is phloem-restricted and transmitted by aphids. The capsid contains two subunit types: the major capsid protein (CP) and a minor component (RTP∗) which arises from the C-terminal cleavage of a readthrough product (RTP). RTP∗ contains the CP sequence fused with a structured domain, denoted NRTD, which is a key determinant of virus transmission. Though both CP and RTP∗ are involved in virus movement and aphid transmission, how RTP∗ is incorporated into the capsid is poorly understood. We present here the structural characterisation, by immunogold labelling and 3D cryo-EM, of the wild-type TuYV and a mutant whose capsid contains the CP only. We show that incorporation of RTP∗ does not impair the capsid structure, and the NRTD does not adopt well-defined positions at the capsid surface. The number of incorporated RTP∗s suggests a random insertion.
Collapse
Affiliation(s)
- Joséphine Lai-Kee-Him
- Centre de Biologie Structurale (CBS), Univ Montpellier, CNRS, INSERM, Montpellier, France.
| | - Stefano Trapani
- Centre de Biologie Structurale (CBS), Univ Montpellier, CNRS, INSERM, Montpellier, France.
| | | | | | - Chloé Fallet
- Centre de Biologie Structurale (CBS), Univ Montpellier, CNRS, INSERM, Montpellier, France
| | - Aurélie Ancelin
- Centre de Biologie Structurale (CBS), Univ Montpellier, CNRS, INSERM, Montpellier, France.
| | - François Lecorre
- Centre de Biologie Structurale (CBS), Univ Montpellier, CNRS, INSERM, Montpellier, France.
| | - François Hoh
- Centre de Biologie Structurale (CBS), Univ Montpellier, CNRS, INSERM, Montpellier, France.
| | - Véronique Ziegler-Graff
- Institut de Biologie Moléculaire des Plantes, CNRS, Université de Strasbourg, Strasbourg, France.
| | | | - Patrick Bron
- Centre de Biologie Structurale (CBS), Univ Montpellier, CNRS, INSERM, Montpellier, France.
| |
Collapse
|
10
|
Cogan DP, Soohoo AM, Chen M, Liu Y, Brodsky KL, Khosla C. Structural basis for intermodular communication in assembly-line polyketide biosynthesis. Nat Chem Biol 2025; 21:876-882. [PMID: 39179672 PMCID: PMC11909739 DOI: 10.1038/s41589-024-01709-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 07/24/2024] [Indexed: 08/26/2024]
Abstract
Assembly-line polyketide synthases (PKSs) are modular multi-enzyme systems with considerable potential for genetic reprogramming. Understanding how they selectively transport biosynthetic intermediates along a defined sequence of active sites could be harnessed to rationally alter PKS product structures. To investigate functional interactions between PKS catalytic and substrate acyl carrier protein (ACP) domains, we employed a bifunctional reagent to crosslink transient domain-domain interfaces of a prototypical assembly line, the 6-deoxyerythronolide B synthase, and resolved their structures by single-particle cryogenic electron microscopy (cryo-EM). Together with statistical per-particle image analysis of cryo-EM data, we uncovered interactions between ketosynthase (KS) and ACP domains that discriminate between intra-modular and inter-modular communication while reinforcing the relevance of conformational asymmetry during the catalytic cycle. Our findings provide a foundation for the structure-based design of hybrid PKSs comprising biosynthetic modules from different naturally occurring assembly lines.
Collapse
Affiliation(s)
- Dillon P Cogan
- Department of Chemistry, Stanford University, Stanford, CA, USA.
- Department of Pharmacology and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA, USA.
| | - Alexander M Soohoo
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
| | - Muyuan Chen
- Division of CryoEM and Bioimaging, Stanford Synchrotron Radiation Lightsource, SLAC National Accelerator Laboratory, Stanford University, Menlo Park, CA, USA
| | - Yan Liu
- Division of CryoEM and Bioimaging, Stanford Synchrotron Radiation Lightsource, SLAC National Accelerator Laboratory, Stanford University, Menlo Park, CA, USA
| | | | - Chaitan Khosla
- Department of Chemistry, Stanford University, Stanford, CA, USA.
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA.
- Stanford ChEM-H, Stanford, CA, USA.
| |
Collapse
|
11
|
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
|
12
|
Nandi S, Chakrabarty S, Bandopadhyay P, Azaharuddin M, Pal A, Das A, Basu T. Synthesis of terbium-labelled tetracycline-loaded calcium phosphate nanoparticle and its mode of action on multi-drug-resistant pathogenic bacteria Escherichia coli and Salmonella kentucky. Biochem Biophys Res Commun 2025; 764:151842. [PMID: 40253911 DOI: 10.1016/j.bbrc.2025.151842] [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: 11/26/2024] [Revised: 04/09/2025] [Accepted: 04/16/2025] [Indexed: 04/22/2025]
Abstract
This study dealt with synthesis of a luminescent nano-form of tetracycline, characterization of its important physico-chemical properties, and molecular mechanism of its antibacterial action on tetracycline-resistant bacterial species. Nanonization was done by entrapping tetracycline (Tet) molecules within calcium phosphate nanoparticles (CPNPs) and doping them with fluorescent terbium (Tb) ions. To characterize the particles, techniques like AFM, SEM, TEM, DLS, absorption-fluorescence-FTIR spectrometry and dialysis were used and to investigate their antibacterial potency and mechanism of action, techniques of agar plating, Ni2+-NTA chromatography, absorption-fluorescence-CD spectroscopy, gel electrophoresis and NS-TEM were used. Tet-Tb-CPNPs, prepared as colloidal suspension, were highly mono-dispersed, moderately stable, spherical in shape, ∼30 nm in size and ∼220 kDa in MW; entrapment efficiency of tetracycline within the nanocomposite particles was about 55 % and its release from them was sustained, bringing out above 95 % of entrapped tetracycline over seven days. The bactericidal concentration of Tet-Tb-CPNP on diarrhoea-causing MDR (including tetracycline) bacteria E. coli and S. kentucky was about 40-45 μg/mL. Binding of Tet-Tb-CPNPs with bacterial ribosome resulted in disruption and degradation of ribosomal proteins and RNAs; such ribosomal degradation was the root cause of its antibacterial action. Moreover, the nanonized tetracycline had no significant toxicity on human neuroblastoma SH-SY5Y cells at its antibacterial dose. Therefore, further pharmacological and clinical investigations are utmost important before commercializing Tet-Tb-CPNP as a potential nano-antibiotic.
Collapse
Affiliation(s)
- Susmita Nandi
- Department of Biochemistry and Biophysics, University of Kalyani, Kalyani, 741 235, West Bengal, India
| | - Soumajit Chakrabarty
- Department of Biochemistry and Biophysics, University of Kalyani, Kalyani, 741 235, West Bengal, India
| | - Pathikrit Bandopadhyay
- Department of Biochemistry and Biophysics, University of Kalyani, Kalyani, 741 235, West Bengal, India
| | - Md Azaharuddin
- Department of Biochemistry and Biophysics, University of Kalyani, Kalyani, 741 235, West Bengal, India
| | - Anabadya Pal
- Department of Biochemistry and Biophysics, University of Kalyani, Kalyani, 741 235, West Bengal, India
| | - Abhijit Das
- Department of Biochemistry and Biophysics, University of Kalyani, Kalyani, 741 235, West Bengal, India
| | - Tarakdas Basu
- Department of Biochemistry and Biophysics, University of Kalyani, Kalyani, 741 235, West Bengal, India.
| |
Collapse
|
13
|
Xue J, Zeng W, John S, Attiq N, Ottolia M, Jiang Y. Structural mechanisms of PIP 2 activation and SEA0400 inhibition in human cardiac sodium-calcium exchanger NCX1. eLife 2025; 14:RP105396. [PMID: 40433952 PMCID: PMC12119087 DOI: 10.7554/elife.105396] [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] [Indexed: 05/29/2025] Open
Abstract
Na+/Ca2+ exchangers (NCXs) transport Ca2+ across the plasma membrane in exchange for Na+ and play a vital role in maintaining cellular Ca2+ homeostasis. Our previous structural study of human cardiac NCX1 (HsNCX1) reveals the overall architecture of the eukaryotic exchanger and the formation of the inactivation assembly by the intracellular regulatory domain that underlies the cytosolic Na+-dependent inactivation and Ca2+ activation of NCX1. Here, we present the cryo-EM structures of HsNCX1 in complex with a physiological activator phosphatidylinositol 4,5-bisphosphate (PIP2), or pharmacological inhibitor SEA0400, that enhances the inactivation of the exchanger. We demonstrate that PIP2 binding stimulates NCX1 activity by inducing a conformational change at the interface between the transmembrane (TM) and cytosolic domains that destabilizes the inactivation assembly. In contrast, SEA0400 binding in the TM domain of NCX1 stabilizes the exchanger in an inward-facing conformation that facilitates the formation of the inactivation assembly, thereby promoting the Na+-dependent inactivation of NCX1. Thus, this study reveals the structural basis of PIP2 activation and SEA0400 inhibition of NCX1 and provides some mechanistic understandings of cellular regulation and pharmacology of NCX family proteins.
Collapse
Affiliation(s)
- Jing Xue
- Department of Physiology, The University of Texas Southwestern Medical CenterDallasUnited States
- Department of Biophysics, The University of Texas Southwestern Medical CenterDallasUnited States
- Howard Hughes Medical InstituteChevy ChaseUnited States
| | - Weizhong Zeng
- Department of Physiology, The University of Texas Southwestern Medical CenterDallasUnited States
- Department of Biophysics, The University of Texas Southwestern Medical CenterDallasUnited States
- Howard Hughes Medical InstituteChevy ChaseUnited States
| | - Scott John
- Department of Medicine (Cardiology), UCLALos AngelesUnited States
| | - Nicole Attiq
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular Medicine, David Geffen School of Medicine, University of California Los AngelesLos AngelesUnited States
| | - Michela Ottolia
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular Medicine, David Geffen School of Medicine, University of California Los AngelesLos AngelesUnited States
| | - Youxing Jiang
- Department of Physiology, The University of Texas Southwestern Medical CenterDallasUnited States
- Department of Biophysics, The University of Texas Southwestern Medical CenterDallasUnited States
- Howard Hughes Medical InstituteChevy ChaseUnited States
| |
Collapse
|
14
|
Kucharska I, Ivanochko D, Hailemariam S, Inklaar MR, Kim HR, Teelen K, Stoter R, van de Vegte-Bolmer M, van Gemert GJ, Semesi A, McLeod B, Ki A, Lee WK, Rubinstein JL, Jore MM, Julien JP. Structural elucidation of full-length Pfs48/45 in complex with potent monoclonal antibodies isolated from a naturally exposed individual. Nat Struct Mol Biol 2025:10.1038/s41594-025-01532-6. [PMID: 40404982 DOI: 10.1038/s41594-025-01532-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 03/12/2025] [Indexed: 05/24/2025]
Abstract
Biomedical interventions that block the transmission of Plasmodium falciparum (Pf) from humans to mosquitoes may be critical for malaria elimination. Pfs48/45, a gamete-surface protein essential for Pf development in the mosquito midgut, is a target of clinical-stage transmission-blocking vaccines and monoclonal antibodies (mAbs) that disrupt Pf transmission to mosquitoes. Antibodies directed to domain 3 of Pfs48/45 have been structurally and functionally described; however, in-depth information about other inhibitory epitopes on Pfs48/45 is currently limited. Here, we present a cryo-electron microscopy structure of full-length Pfs48/45 in complex with potent human mAbs targeting all three domains. Our data indicate that although Pfs48/45 domains 1 and 2 are rigidly coupled, there is substantial conformational flexibility between domains 2 and 3. Characterization of mAbs against domain 1 revealed the presence of a conformational epitope class that is largely conserved across Pf field isolates and is associated with recognition by potent antibodies. Our study provides insights into epitopes across full-length Pfs48/45 and has implications for the design of next-generation malaria interventions.
Collapse
Affiliation(s)
- Iga Kucharska
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Danton Ivanochko
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Sophia Hailemariam
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Maartje R Inklaar
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Hee Ryung Kim
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Karina Teelen
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Rianne Stoter
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | | | - Geert-Jan van Gemert
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Anthony Semesi
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Brandon McLeod
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Ahyoung Ki
- Structural Analysis Team, New Drug Development Center, KBIO Osong Medical Innovation Foundation, Osong, Republic of Korea
| | - Won-Kyu Lee
- Structural Analysis Team, New Drug Development Center, KBIO Osong Medical Innovation Foundation, Osong, Republic of Korea
| | - John L Rubinstein
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Matthijs M Jore
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Jean-Philippe Julien
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada.
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada.
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
15
|
Risi CM, Landim-Vieira M, Belknap B, Chase PB, Pinto JR, Galkin VE. The role of the troponin T interactions with actin in regulation of cardiac thin filament revealed by the troponin T pathogenic variant Ile79Asn. J Mol Cell Cardiol 2025; 204:55-67. [PMID: 40412797 DOI: 10.1016/j.yjmcc.2025.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 04/28/2025] [Accepted: 05/19/2025] [Indexed: 05/27/2025]
Abstract
Cardiac muscle contraction/relaxation cycle depends on the rising and falling Ca2+ levels in sarcomeres that control the extent of interactions between myosin-based thick and actin-based thin filaments. Cardiac thin filament (cTF) consists of actin, tropomyosin (Tm) that regulates myosin binding to actin, and troponin complex that governs Tm position upon Ca2+-binding. Troponin has three subunits - Ca2+-binding troponin C (TnC), Tm stabilizing troponin T (TnT), and inhibitory troponin I (TnI). TnT N-terminus (TnT1) interactions with actin stabilize the inhibited state of cTF. TnC, TnI, and Tm work in concert to control actomyosin interactions. Cryo-electron microscopy (cryo-EM) provided factual structures of healthy cTF, but structures of cTF carrying missense mutations linked to human cardiomyopathy are unknown. Variant Ile79Asn in human cardiac TnT (TnT-I79N) increases myofilament Ca2+ sensitivity and slows cross-bridge kinetics, leading to severe hypertrophic/restrictive cardiomyopathy. Here, we used TnT-I79N mutation as a tool to examine the role of TnT1 in the complex mechanism of cTF regulation. Comparison of the cryo-EM structures of murine wild type and TnT-I79N native cTFs at systolic Ca2+ levels (pCa = 5.8) demonstrates that TnT-I79N causes 1) dissociation of the TnT1 loop from its actin interface that results in Tm release to a more activated position, 2) reduced interaction of TnI C-terminus with actin-Tm, and 3) increased frequency of Ca2+-bound regulatory units. Our data indicate that the TnT1 loop is a crucial element of the allosteric regulatory network that couples Tn subunits and Tm to maintain adequate cTF response to physiological Ca2+ levels during a heartbeat.
Collapse
Affiliation(s)
- Cristina M Risi
- Department of Biomedical and Translational Sciences, Macon & Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, VA 23507, USA
| | - Maicon Landim-Vieira
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL, USA
| | - Betty Belknap
- Department of Biomedical and Translational Sciences, Macon & Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, VA 23507, USA
| | - P Bryant Chase
- Department of Biological Science, Florida State University, Tallahassee, FL, USA
| | - Jose R Pinto
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL, USA
| | - Vitold E Galkin
- Department of Biomedical and Translational Sciences, Macon & Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, VA 23507, USA.
| |
Collapse
|
16
|
Chen Q, Schafer CT, Mukherjee S, Wang K, Gustavsson M, Fuller JR, Tepper K, Lamme TD, Aydin Y, Agrawal P, Terashi G, Yao XQ, Kihara D, Kossiakoff AA, Handel TM, Tesmer JJG. Effect of phosphorylation barcodes on arrestin binding to a chemokine receptor. Nature 2025:10.1038/s41586-025-09024-9. [PMID: 40399676 DOI: 10.1038/s41586-025-09024-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Accepted: 04/15/2025] [Indexed: 05/23/2025]
Abstract
Unique phosphorylation 'barcodes' installed in different regions of an active seven-transmembrane receptor by different G-protein-coupled receptor (GPCR) kinases (GRKs) have been proposed to promote distinct cellular outcomes1, but it is unclear whether or how arrestins differentially engage these barcodes. Here, to address this, we developed an antigen-binding fragment (Fab7) that recognizes both active arrestin2 (β-arrestin1) and arrestin3 (β-arrestin2) without interacting with bound receptor polypeptides. We used Fab7 to determine the structures of both arrestins in complex with atypical chemokine receptor 3 (ACKR3) phosphorylated in different regions of its C-terminal tail by either GRK2 or GRK5 (ref. 2). The GRK2-phosphorylated ACKR3 resulted in more heterogeneous 'tail-mode' assemblies, whereas phosphorylation by GRK5 resulted in more rigid 'ACKR3-adjacent' assemblies. Unexpectedly, the finger loops of both arrestins engaged the micelle surface rather than the receptor intracellular pocket, with arrestin3 being more dynamic, partly because of its lack of a membrane-anchoring motif. Thus, both the region of the barcode and the arrestin isoform involved can alter the structure and dynamics of GPCR-arrestin complexes, providing a possible mechanistic basis for unique downstream cellular effects, such as the efficiency of chemokine scavenging and the robustness of arrestin binding in ACKR3.
Collapse
Affiliation(s)
- Qiuyan Chen
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA.
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA.
| | - Christopher T Schafer
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA
- Department of Medicinal Chemistry, Amsterdam Institute for Molecular and Life Sciences, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Somnath Mukherjee
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, USA
| | - Kai Wang
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Martin Gustavsson
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Katelyn Tepper
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Thomas D Lamme
- Department of Medicinal Chemistry, Amsterdam Institute for Molecular and Life Sciences, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Yasmin Aydin
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Parth Agrawal
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, USA
| | - Genki Terashi
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
| | - Xin-Qiu Yao
- Department of Chemistry, University of Nebraska Omaha, Omaha, NE, USA
| | - Daisuke Kihara
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
| | - Anthony A Kossiakoff
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, USA
| | - Tracy M Handel
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA.
| | - John J G Tesmer
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA.
| |
Collapse
|
17
|
Petrovic I, Tatli M, Desai S, Grahl A, Ni D, Stahlberg H, Spang A, Grzesiek S, Abiko LA. Arrestin recognizes GPCRs independently of the receptor state. Proc Natl Acad Sci U S A 2025; 122:e2501487122. [PMID: 40372433 DOI: 10.1073/pnas.2501487122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 03/31/2025] [Indexed: 05/16/2025] Open
Abstract
Only two nonvisual arrestins recognize many hundreds of different, intracellularly phosphorylated G protein-coupled receptors (GPCRs). Due to the highly dynamic nature of GPCR•arrestin complexes, the critical determinants of GPCR-arrestin recognition have remained largely unclear. We show here that arrestin2 recruitment to the β1-adrenergic receptor (β1AR) can be induced by an arrestin-activating phosphopeptide that is not covalently linked to the receptor and that the recruitment is independent of the presence and type of the orthosteric receptor ligand. Apparently, the arrestin-receptor interaction is driven by the conformational switch within arrestin induced by the phosphopeptide, whereas the electrostatic attraction toward the receptor phosphosites may only play an auxiliary role. Extensive NMR observations show that in contrast to previous static GPCR•arrestin complex structures, the β1AR complex with the beta-blocker carvedilol and arrestin2 is in a G protein-inactive conformation. The insensitivity to the specific receptor conformation provides a rationale for arrestin's promiscuous recognition of GPCRs and explains the arrestin-biased agonism of carvedilol, which largely blocks G protein binding, while still enabling arrestin engagement.
Collapse
Affiliation(s)
- Ivana Petrovic
- Department of Biozentrum, University of Basel, Basel CH-4056, Switzerland
| | - Meltem Tatli
- Laboratory of Biological Electron Microscopy, Institute of Physics, School of Basic Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne CH-1015, Switzerland
- Laboratory of Biological Electron Microscopy, Department of Fundamental Microbiology, Faculty of Biology and Medicine, University of Lausanne, Lausanne CH-1015, Switzerland
| | - Samit Desai
- Department of Biozentrum, University of Basel, Basel CH-4056, Switzerland
| | - Anne Grahl
- Department of Biozentrum, University of Basel, Basel CH-4056, Switzerland
| | - Dongchun Ni
- Laboratory of Biological Electron Microscopy, Institute of Physics, School of Basic Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne CH-1015, Switzerland
- Laboratory of Biological Electron Microscopy, Department of Fundamental Microbiology, Faculty of Biology and Medicine, University of Lausanne, Lausanne CH-1015, Switzerland
| | - Henning Stahlberg
- Laboratory of Biological Electron Microscopy, Institute of Physics, School of Basic Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne CH-1015, Switzerland
- Laboratory of Biological Electron Microscopy, Department of Fundamental Microbiology, Faculty of Biology and Medicine, University of Lausanne, Lausanne CH-1015, Switzerland
| | - Anne Spang
- Department of Biozentrum, University of Basel, Basel CH-4056, Switzerland
| | - Stephan Grzesiek
- Department of Biozentrum, University of Basel, Basel CH-4056, Switzerland
| | - Layara Akemi Abiko
- Department of Biozentrum, University of Basel, Basel CH-4056, Switzerland
| |
Collapse
|
18
|
Ma B, Cao Z, Ding W, Zhang X, Xiang Y, Cao D. Structural basis for the recognition of two different types of receptors by Western equine encephalitis virus. Cell Rep 2025; 44:115724. [PMID: 40402741 DOI: 10.1016/j.celrep.2025.115724] [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: 01/15/2025] [Revised: 03/25/2025] [Accepted: 04/29/2025] [Indexed: 05/24/2025] Open
Abstract
Western equine encephalitis virus (WEEV) enters cells via various receptors. Here, we report the cryoelectron microscopy (cryo-EM) structures of WEEV in complex with its receptors PCDH10 and very-low-density lipoprotein receptor (VLDLR). Structural analysis shows that PCDH10 binds in the cleft formed by adjacent E2-E1 heterodimers of WEEV through its EC1 ectodomain. Residues of viral envelope proteins involved in the interactions with PCDH10 EC1 are unique to WEEV. The strain-specific receptor VLDLR binds WEEV strain McMillan through two consecutive ecto-LDLR class A (LA) repeats. LA1-2, LA2-3, LA3-4, LA4-5, and LA5-6 of VLDLR all have detectable interactions with WEEV. Detailed structures of WEEV in complex with LA1-2 and LA2-3 show that the N-terminal LA repeat binds in the cleft and that the C-terminal LA repeat is attached to the E2 B domain. The acquisition of a single E2 mutation (V265F) allows WEEV strain 71V-1658, originally unable to bind VLDLR, to gain this receptor-binding ability. The binding of VLDLR to WEEV is in a mode different from those of other alphaviruses.
Collapse
Affiliation(s)
- Bingting Ma
- Beijing Frontier Research Center for Biological Structure, Center for Infectious Disease Research, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, P.R. China; SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Taiyuan 030001, P.R. China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, P.R. China
| | - Ziyi Cao
- National Laboratory of Biomacromolecules, Key Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences (CAS), Beijing 100101, P.R. China; University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Weijia Ding
- Beijing Frontier Research Center for Biological Structure, Center for Infectious Disease Research, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, P.R. China; SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Taiyuan 030001, P.R. China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, P.R. China
| | - Xinzheng Zhang
- National Laboratory of Biomacromolecules, Key Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences (CAS), Beijing 100101, P.R. China; University of Chinese Academy of Sciences, Beijing 100049, P.R. China.
| | - Ye Xiang
- Beijing Frontier Research Center for Biological Structure, Center for Infectious Disease Research, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, P.R. China; SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Taiyuan 030001, P.R. China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, P.R. China.
| | - Duanfang Cao
- National Laboratory of Biomacromolecules, Key Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences (CAS), Beijing 100101, P.R. China; University of Chinese Academy of Sciences, Beijing 100049, P.R. China.
| |
Collapse
|
19
|
Arimura Y, Konishi HA, Funabiki H. MagIC-Cryo-EM, structural determination on magnetic beads for scarce macromolecules in heterogeneous samples. eLife 2025; 13:RP103486. [PMID: 40390365 PMCID: PMC12092007 DOI: 10.7554/elife.103486] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/21/2025] Open
Abstract
Cryo-EM single-particle analyses typically require target macromolecule concentration at 0.05~5.0 mg/ml, which is often difficult to achieve. Here, we devise Magnetic Isolation and Concentration (MagIC)-cryo-EM, a technique enabling direct structural analysis of targets captured on magnetic beads, thereby reducing the targets' concentration requirement to <0.0005 mg/mL. Adapting MagIC-cryo-EM to a Chromatin Immunoprecipitation protocol, we characterized structural variations of the linker histone H1.8-associated nucleosomes that were isolated from interphase and metaphase chromosomes in Xenopus egg extract. Combining Duplicated Selection To Exclude Rubbish particles (DuSTER), a particle curation method that excludes low signal-to-noise ratio particles, we also resolved the 3D cryo-EM structures of nucleoplasmin NPM2 co-isolated with the linker histone H1.8 and revealed distinct open and closed structural variants. Our study demonstrates the utility of MagIC-cryo-EM for structural analysis of scarce macromolecules in heterogeneous samples and provides structural insights into the cell cycle-regulation of H1.8 association to nucleosomes.
Collapse
Affiliation(s)
- Yasuhiro Arimura
- Laboratory of Chromosome and Cell Biology, The Rockefeller UniversityNew YorkUnited States
- Basic Sciences Division, Fred Hutchinson Cancer CenterSeattleUnited States
| | - Hide A Konishi
- Laboratory of Chromosome and Cell Biology, The Rockefeller UniversityNew YorkUnited States
| | - Hironori Funabiki
- Laboratory of Chromosome and Cell Biology, The Rockefeller UniversityNew YorkUnited States
| |
Collapse
|
20
|
Wu C, Song N, Zhao Y, Wang H, Ai Y, Wang Y, Wang Y, Yuan X, Liu T, Li N, Jaijyan DK, Li C, Zhang L, Zheng W, Yang Z, Zhu S, Liao HX. Structural basis of human cytomegalovirus neutralization by gB AD-5-specific potent antibodies. Cell Rep 2025; 44:115646. [PMID: 40382771 DOI: 10.1016/j.celrep.2025.115646] [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: 11/04/2024] [Revised: 02/25/2025] [Accepted: 04/11/2025] [Indexed: 05/20/2025] Open
Abstract
Human cytomegalovirus (hCMV) poses a severe threat to fetuses, newborns, and immunocompromised individuals. No approved vaccines and limited treatment options are current medical challenges. Here, we analyze the human B cell responses to glycoprotein B (gB) in three top hCMV neutralizers from a cohort of 283 individuals with latent-infected hCMV. By single-cell amplification of memory B cells, we identify a cluster of potent neutralizing monoclonal antibodies (nAbs) that competitively recognize an unknown vulnerable site on gB antigenic domain 5 (AD-5). This cluster of nAbs functionally outperforms the nAbs utilized in clinical trials. Cryoelectron microscopy (cryo-EM) unveils the structural basis of the neutralization mechanism of an antibody directly targeting the fusion subdomain on AD-5. Moreover, immunological analyses of human and mouse sera have preliminarily validated the potential superiority of AD-5-focused immune responses. Overall, our results will support the development of optimized gB-based vaccines and provide promising prophylactic and therapeutic antibody candidates against hCMV infection.
Collapse
Affiliation(s)
- Changwen Wu
- Trinomab Pharmaceutical Co., Ltd., Zhuhai 519040, China.
| | - Nan Song
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yizhen Zhao
- MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, School of Physics, Xi'an Jiaotong University, Xi'an 710049, China
| | - Han Wang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yuanbao Ai
- Trinomab Pharmaceutical Co., Ltd., Zhuhai 519040, China
| | - Yayu Wang
- Trinomab Pharmaceutical Co., Ltd., Zhuhai 519040, China
| | - Yueming Wang
- Trinomab Pharmaceutical Co., Ltd., Zhuhai 519040, China
| | - Xiaohui Yuan
- Trinomab Pharmaceutical Co., Ltd., Zhuhai 519040, China
| | - Tong Liu
- Trinomab Pharmaceutical Co., Ltd., Zhuhai 519040, China
| | - Nan Li
- Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China; Guangdong Provincial Key Laboratory of Bioengineering Medicine, Guangzhou 510632, China
| | - Dabbu Kumar Jaijyan
- Department of Microbiology and Molecular Genetics, New Jersey Medical School, Rutgers University, 225 Warren Street, Newark, NJ 070101, USA
| | - Chengming Li
- Trinomab Pharmaceutical Co., Ltd., Zhuhai 519040, China
| | - Lei Zhang
- MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, School of Physics, Xi'an Jiaotong University, Xi'an 710049, China
| | - Weihong Zheng
- Trinomab Pharmaceutical Co., Ltd., Zhuhai 519040, China
| | - Zhiwei Yang
- MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, School of Physics, Xi'an Jiaotong University, Xi'an 710049, China.
| | - Shujia Zhu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China.
| | - Hua-Xin Liao
- Trinomab Pharmaceutical Co., Ltd., Zhuhai 519040, China.
| |
Collapse
|
21
|
Wu H, Sun L, Huo T, Wensel TG, Horrigan FT, Wang Z. The identification of XPR1 as a voltage- and phosphate-activated phosphate-permeable ion channel. Nat Commun 2025; 16:4519. [PMID: 40374661 PMCID: PMC12081713 DOI: 10.1038/s41467-025-59678-2] [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: 12/10/2024] [Accepted: 04/29/2025] [Indexed: 05/17/2025] Open
Abstract
Maintaining a balance of inorganic phosphate (Pi) is vital for cellular functionality. Proper phosphate levels are managed through Pi import and export; and the processes governing Pi export remain the least understood. Xenotropic and Polytropic retrovirus Receptor 1 (XPR1) has been identified as the only known Pi export protein in mammals. In this study, we introduce the cryogenic electron microscopy structure of human XPR1 (hXPR1), unveiling a structural arrangement distinct from that of any known ion transporter. Our structural results suggest that hXPR1 may operate as an ion channel, a hypothesis supported by patch clamp recordings revealing hXPR1's voltage- and Pi-dependent activity and large unitary conductance. Further analyses, including the structure of hXPR1 in presence of Pi, and mutagenesis studies at one of the putative Pi binding sites, lead us to propose a plausible ion permeation pathway. Together, our results provide novel perspectives on the Pi transport mechanism of XPR1.
Collapse
Affiliation(s)
- Hongjiang Wu
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX, USA
| | - Liang Sun
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Tong Huo
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX, USA
| | - Theodore G Wensel
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX, USA
| | - Frank T Horrigan
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA.
| | - Zhao Wang
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX, USA.
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
- CryoEM Core (Advanced Technology Core), Baylor College of Medicine, Houston, TX, USA.
- Department of Materials Science and NanoEngineering, Rice University, Houston, TX, USA.
- Department of Molecular and Cellular Oncology, Division of Basic Science, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
22
|
Dam KMA, Gristick HB, Li YE, Yang Z, Gnanapragasam PNP, West AP, Seaman MS, Bjorkman PJ. Mapping essential somatic hypermutations in a CD4-binding site bNAb informs HIV-1 vaccine design. Cell Rep 2025; 44:115713. [PMID: 40378041 PMCID: PMC12117015 DOI: 10.1016/j.celrep.2025.115713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 03/17/2025] [Accepted: 04/25/2025] [Indexed: 05/18/2025] Open
Abstract
HIV-1 broadly neutralizing antibodies (bNAbs) targeting the CD4-binding site (CD4bs) contain rare features that pose challenges to elicit these bNAbs through vaccination. The IOMA class of CD4bs bNAbs includes fewer rare features and somatic hypermutations (SHMs) to achieve broad neutralization, thus presenting a potentially accessible pathway for vaccine-induced bNAb development. Here, we created a library of IOMA variants in which each SHM was individually reverted to the inferred germline counterpart to investigate the roles of SHMs in conferring IOMA's neutralization potency and breadth. Impacts on neutralization for each variant were evaluated, and this information was used to design minimally mutated IOMA-class variants (IOMAmin) that incorporated the fewest SHMs required for achieving IOMA's neutralization breadth. A cryoelectron microscopy (cryo-EM) structure of an IOMAmin variant bound to Env was used to further interpret characteristics of IOMA variants to elucidate how IOMA's structural features correlate with its neutralization mechanism, informing the design of IOMA-targeting immunogens.
Collapse
Affiliation(s)
- Kim-Marie A Dam
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Harry B Gristick
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Yancheng E Li
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA; Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Zhi Yang
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | | | - Anthony P West
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Michael S Seaman
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Pamela J Bjorkman
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA.
| |
Collapse
|
23
|
Veiveris D, Kopustas A, Sulskis D, Mikalauskaite K, Alsamsam MN, Tutkus M, Smirnovas V, Ziaunys M. Heterotypic Droplet Formation by Pro-Inflammatory S100A9 and Neurodegenerative Disease-Related α-Synuclein. Biomacromolecules 2025. [PMID: 40370127 DOI: 10.1021/acs.biomac.5c00130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2025]
Abstract
Liquid-liquid phase separation of proteins and nucleic acids is a rapidly emerging field of study, aimed at understanding the process of biomolecular condensate formation. Recently, it has been discovered that different neurodegenerative disease-related proteins, such as α-synuclein and amyloid-β are capable of forming heterotypic droplets. Other reports have also shown non-LLPS cross-interactions between various amyloidogenic proteins and the resulting influence on their amyloid fibril formation. This includes the new discovery of pro-inflammatory S100A9 affecting the aggregation of both amyloid-β, as well as α-synuclein. In this study, we explore the formation of heterotypic droplets by S100A9 and α-synuclein. We show that their mixture is capable of assembling into both homotypic and heterotypic condensates and that this cross-interaction alters the aggregation mechanism of α-synuclein. These results provide insight into the influence of S100A9 on the process of neurodegenerative disease-related protein LLPS and aggregation.
Collapse
Affiliation(s)
- Dominykas Veiveris
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius LT-10257, Lithuania
| | - Aurimas Kopustas
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius LT-10257, Lithuania
- Department of Molecular Compound Physics, Center for Physical Sciences and Technology, Vilnius LT-10257, Lithuania
| | - Darius Sulskis
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius LT-10257, Lithuania
| | - Kamile Mikalauskaite
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius LT-10257, Lithuania
| | - Mohammad Nour Alsamsam
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius LT-10257, Lithuania
- Department of Molecular Compound Physics, Center for Physical Sciences and Technology, Vilnius LT-10257, Lithuania
| | - Marijonas Tutkus
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius LT-10257, Lithuania
- Department of Molecular Compound Physics, Center for Physical Sciences and Technology, Vilnius LT-10257, Lithuania
| | - Vytautas Smirnovas
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius LT-10257, Lithuania
| | - Mantas Ziaunys
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius LT-10257, Lithuania
| |
Collapse
|
24
|
Zhan X, Zeng X, Uddin MR, Xu M. AITom: AI-guided cryo-electron tomography image analyses toolkit. J Struct Biol 2025; 217:108207. [PMID: 40378936 DOI: 10.1016/j.jsb.2025.108207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 04/20/2025] [Accepted: 04/28/2025] [Indexed: 05/19/2025]
Abstract
Cryo-electron tomography (cryo-ET) is an essential tool in structural biology, uniquely capable of visualizing three-dimensional macromolecular complexes within their native cellular environments, thereby providing profound molecular-level insights. Despite its significant promise, cryo-ET faces persistent challenges in the systematic localization, identification, segmentation, and structural recovery of three-dimensional subcellular components, necessitating the development of efficient and accurate large-scale image analysis methods. In response to these complexities, this paper introduces AITom, an open-source artificial intelligence platform tailored for cryo-ET researchers. AITom integrates a comprehensive suite of public and proprietary algorithms, supporting both traditional template-based and template-free approaches, alongside state-of-the-art deep learning methodologies for cryo-ET data analysis. By incorporating diverse computational strategies, AITom enables researchers to more effectively tackle the complexities inherent in cryo-ET, facilitating precise analysis and interpretation of complex biological structures. Furthermore, AITom provides extensive tutorials for each analysis module, offering valuable guidance to users in utilizing its comprehensive functionalities.
Collapse
Affiliation(s)
- Xueying Zhan
- Computational Biology Department, Carnegie Mellon University, Pittsburgh, PA, United States
| | - Xiangrui Zeng
- Computational Biology Department, Carnegie Mellon University, Pittsburgh, PA, United States
| | - Mostofa Rafid Uddin
- Computational Biology Department, Carnegie Mellon University, Pittsburgh, PA, United States
| | - Min Xu
- Computational Biology Department, Carnegie Mellon University, Pittsburgh, PA, United States.
| |
Collapse
|
25
|
Liu W, Soliman A, Emam HE, Zhang J, Bonventre JV, Lee LP, Nasr ML. Self-Assembly of Nanogold Triplets on Trimeric Viral Proteins for Infectious Disease Diagnosis. ACS NANO 2025; 19:17514-17524. [PMID: 40323304 DOI: 10.1021/acsnano.4c17685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2025]
Abstract
Timely and accurate diagnostics for infectious diseases are essential in preventing their worldwide spread. Though rapid diagnostic tests are favored for their speed, cost-effectiveness, and ease of use, most tests compromise sensitivity, which risks false-negative results. Here, we present the self-assembly of nanogold triplets on trimeric viral surface proteins for a sensitive colorimetric assay to identify viruses. Gold triplets were self-assembled on the viral trimeric surface proteins using ultrasmall gold nanoparticles. We observed a significant wavelength shift of 70 nm, enabling straightforward naked-eye detection through gold triplets that act as catalysts for producing nanoplasmonic viruses. We established the detection limit of 3 × 105 copies/ml using an effective colorimetric assay for detecting SARS-CoV-2. The self-assembly of gold triplets on trimeric viral surface proteins provides a reliable approach to the accurate and sensitive detection of viruses.
Collapse
Affiliation(s)
- Wenpeng Liu
- Renal Division and Division of Engineering in Medicine, Department of Medicine, Brigham Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Ahmed Soliman
- Renal Division and Division of Engineering in Medicine, Department of Medicine, Brigham Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Hagar E Emam
- Renal Division and Division of Engineering in Medicine, Department of Medicine, Brigham Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Jun Zhang
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Joseph V Bonventre
- Renal Division and Division of Engineering in Medicine, Department of Medicine, Brigham Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Luke P Lee
- Renal Division and Division of Engineering in Medicine, Department of Medicine, Brigham Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- Department of Bioengineering, University of California, Berkeley, Berkeley, California 94720, United States
- Department of Electrical Engineering and Computer Science, University of California, Berkeley, Berkeley, California 94720, United States
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon 16419, Korea
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul 03760, Korea
| | - Mahmoud L Nasr
- Renal Division and Division of Engineering in Medicine, Department of Medicine, Brigham Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai 505055, UAE
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| |
Collapse
|
26
|
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
|
27
|
Kim M, Kim J, Lee GS, Olinares PDB, Airan Y, Chow JL, Park J, Jeong Y, Park J, Chait BT, Herzon SB, Kim CS, Kang JY. Structural study on human microbiome-derived polyketide synthases that assemble genotoxic colibactin. Structure 2025:S0969-2126(25)00173-X. [PMID: 40381618 DOI: 10.1016/j.str.2025.04.017] [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: 01/13/2025] [Revised: 03/14/2025] [Accepted: 04/22/2025] [Indexed: 05/20/2025]
Abstract
Colibactin, a human microbiome-derived genotoxin, promotes colorectal cancer by damaging the host gut epithelial genomes. While colibactin is synthesized via a hybrid non-ribosomal peptide synthetase (NRPS)-polyketide synthase (PKS) pathway, known as pks or clb, the structural details of its biosynthetic enzymes remain limited, hindering our understanding of its biosynthesis and clinical application. In this study, we report the cryo-EM structures of two colibactin-producing PKS enzymes, ClbC and ClbI, captured in different reaction states using a substrate-mimic crosslinker. Our structural analysis revealed the binding sites of carrier protein (CP) domains of the ClbC and ClbI on their ketosynthase (KS) domains. Further, we identified a novel NRPS-PKS docking interaction between ClbI and its upstream enzyme, ClbH, mediated by the C-terminal peptide ClbH and the dimeric interface of ClbI, establishing a 1:2 stoichiometry. These findings advance our understanding of colibactin assembly line and provide broader insights into NRPS-PKS natural product biosynthesis mechanisms.
Collapse
Affiliation(s)
- Minjae Kim
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Jinwoo Kim
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Gyu Sung Lee
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon, Republic of Korea
| | - Paul Dominic B Olinares
- Laboratory of Mass Spectrometry and Gaseous Ion Chemistry, The Rockefeller University, New York, NY, USA
| | - Yougant Airan
- Department of Chemistry, Yale University, New Haven, CT 06520, USA
| | - Jasmine L Chow
- Department of Chemistry, Yale University, New Haven, CT 06520, USA
| | - Jongseok Park
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Yujin Jeong
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Jiho Park
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Brian T Chait
- Laboratory of Mass Spectrometry and Gaseous Ion Chemistry, The Rockefeller University, New York, NY, USA
| | - Seth B Herzon
- Department of Chemistry, Yale University, New Haven, CT 06520, USA; Department of Pharmacology and Therapeutic Radiology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Chung Sub Kim
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon, Republic of Korea; School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea.
| | - Jin Young Kang
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.
| |
Collapse
|
28
|
Stautz J, Griwatz D, Kaltwasser S, Mehdipour AR, Ketter S, Thiel C, Wunnicke D, Schrecker M, Mills DJ, Hummer G, Vonck J, Hänelt I. A short intrinsically disordered region at KtrB's N-terminus facilitates allosteric regulation of K + channel KtrAB. Nat Commun 2025; 16:4252. [PMID: 40335548 PMCID: PMC12059179 DOI: 10.1038/s41467-025-59546-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 04/25/2025] [Indexed: 05/09/2025] Open
Abstract
K+ homeostasis is crucial for bacterial survival. The bacterial K+ channel KtrAB is regulated by the binding of ADP and ATP to the cytosolic RCK subunits KtrA. While the ligand-induced conformational changes in KtrA are well described, the transmission to the gating regions within KtrB is not understood. Here, we present a cryo-EM structure of the ADP-bound, inactive KtrAB complex from Vibrio alginolyticus, which resolves part of KtrB's N termini. They are short intrinsically disordered regions (IDRs) located at the interface of KtrA and KtrB. We reveal that these IDRs play a decisive role in ATP-mediated channel opening, while the closed ADP-bound state does not depend on the N-termini. We propose an allosteric mechanism, in which ATP-induced conformational changes within KtrA trigger an interaction of KtrB's N-terminal IDRs with the membrane, stabilizing the active and conductive state of KtrAB.
Collapse
Affiliation(s)
- Janina Stautz
- Institute of Biochemistry, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - David Griwatz
- Institute of Biochemistry, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Susann Kaltwasser
- Central Electron Microscopy Facility, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
| | - Ahmad Reza Mehdipour
- Center for Molecular Modeling, Ghent University, Zwijnaarde, Belgium
- Department of Theoretical Biophysics, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
| | - Sophie Ketter
- Institute of Biochemistry, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Celina Thiel
- Institute of Biochemistry, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Dorith Wunnicke
- Institute of Biochemistry, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Marina Schrecker
- Institute of Biochemistry, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Deryck J Mills
- Department of Structural Biology, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
| | - Gerhard Hummer
- Department of Theoretical Biophysics, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
- Institute for Biophysics, Goethe University Frankfurt, Frankfurt am Main, 60438, Germany
| | - Janet Vonck
- Department of Structural Biology, Max Planck Institute of Biophysics, Frankfurt am Main, Germany.
| | - Inga Hänelt
- Institute of Biochemistry, Goethe University Frankfurt, Frankfurt am Main, Germany.
| |
Collapse
|
29
|
Vigers MP, Lobo S, Najafi S, Dubose A, Tsay K, Ganguly P, Longhini AP, Jin Y, Buratto SK, Kosik KS, Shell MS, Shea JE, Han S. Water-directed pinning is key to tau prion formation. Proc Natl Acad Sci U S A 2025; 122:e2421391122. [PMID: 40294272 PMCID: PMC12067210 DOI: 10.1073/pnas.2421391122] [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: 10/24/2024] [Accepted: 03/10/2025] [Indexed: 04/30/2025] Open
Abstract
Tau forms fibrillar aggregates that are pathological hallmarks of a family of neurodegenerative diseases known as tauopathies. The synthetic replication of disease-specific fibril structures is a critical gap for developing diagnostic and therapeutic tools. This study debuts a strategy of identifying a critical and minimal folding motif in fibrils characteristic of tauopathies and generating seeding-competent fibrils from the isolated tau peptides. The 19-residue jR2R3 peptide (295 to 313) which spans the R2/R3 splice junction of tau, and includes the P301L mutation, is one such peptide that forms prion-competent fibrils. This tau fragment contains the hydrophobic VQIVYK hexapeptide that is part of the core of all known pathological tau fibril structures and an intramolecular counterstrand that stabilizes the strand-loop-strand (SLS) motif observed in 4R tauopathy fibrils. This study shows that P301L exhibits a duality of effects: it lowers the barrier for the peptide to adopt aggregation-prone conformations and enhances the local structuring of water around the mutation site to facilitate site-directed pinning and dewetting around sites 300-301 to achieve in-register stacking of tau to cross β-sheets. We solved a 3 Å cryo-EM structure of jR2R3-P301L fibrils in which each protofilament layer contains two jR2R3-P301L copies, of which one adopts a SLS fold found in 4R tauopathies and the other wraps around the SLS fold to stabilize it, reminiscent of the three- and fourfold structures observed in 4R tauopathies. These jR2R3-P301L fibrils are competent to template full-length 4R tau in a prion-like manner.
Collapse
Affiliation(s)
- Michael P. Vigers
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, CA93106
| | - Samuel Lobo
- Department of Chemical Engineering, University of California, Santa Barbara, CA93106
| | - Saeed Najafi
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, CA93106
| | - Austin Dubose
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, CA93106
| | - Karen Tsay
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, CA93106
| | - Pritam Ganguly
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, CA93106
- Department of Physics, University of California, Santa Barbara, CA93106
| | - Andrew P. Longhini
- Neuroscience Research Institute, University of California, Santa Barbara, CA93106
- Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, CA93106
| | - Yingying Jin
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, CA93106
| | - Steven K. Buratto
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, CA93106
| | - Kenneth S. Kosik
- Neuroscience Research Institute, University of California, Santa Barbara, CA93106
- Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, CA93106
| | - M. Scott Shell
- Department of Chemical Engineering, University of California, Santa Barbara, CA93106
| | - Joan-Emma Shea
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, CA93106
- Department of Physics, University of California, Santa Barbara, CA93106
| | - Songi Han
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, CA93106
- Department of Chemical Engineering, University of California, Santa Barbara, CA93106
- Neuroscience Research Institute, University of California, Santa Barbara, CA93106
- Department of Chemistry, Northwestern University, Evanston, IL60208
| |
Collapse
|
30
|
Wang G, Wang P, Zheng Z, Zhang Q, Xu C, Xu X, Jian L, Zhao Z, Cai G, Wang X. Molecular architecture and inhibition mechanism of human ATR-ATRIP. Sci Bull (Beijing) 2025:S2095-9273(25)00489-X. [PMID: 40379520 DOI: 10.1016/j.scib.2025.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Revised: 03/31/2025] [Accepted: 04/29/2025] [Indexed: 05/19/2025]
Abstract
The ataxia telangiectasia-mutated and Rad3-related (ATR) kinase is a master regulator of DNA damage response and replication stress in humans. Targeting ATR is the focus of oncology drug pipelines with a number of potent, selective ATR inhibitors currently in clinical development. Here, we determined the cryo-EM structures of the human ATR-ATRIP complex in the presence of VE-822 and RP-3500, two ATR inhibitors currently in Phase II clinical trials, achieving an overall resolution of approximately 3 Å. These structures yield a near-complete atomic model of the ATR-ATRIP complex, revealing subunit stoichiometry, intramolecular and intermolecular interactions, and critical regulatory sites including an insertion in the PIKK regulatory domain (PRD). Structural comparison provides insights into the modes of action and selectivity of ATR inhibitors. The divergent binding modes near the solvent side and in the rear pocket area of VE-822 and RP-3500, particularly their disparate binding orientations, lead to varying conformational changes in the active site. Surprisingly, one ATR-ATRIP complex binds four VE-822 molecules, with two in the ATR active site and two at the ATR-ATR dimer interface. The binding and selectivity of RP-3500 depend on two bound water molecules, which may be further enhanced by the substitution of these bound waters. Our study provides a structural framework for understanding ATR regulation and holds promise for assisting future efforts in rational drug design targeting ATR.
Collapse
Affiliation(s)
- Guangxian Wang
- Department of Radiation Oncology, the First Affiliated Hospital of USTC, MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230000, China
| | - Po Wang
- Department of Radiation Oncology, the First Affiliated Hospital of USTC, MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230000, China
| | - Zexuan Zheng
- Department of Radiation Oncology, the First Affiliated Hospital of USTC, MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230000, China
| | - Qingjun Zhang
- Department of Radiation Oncology, the First Affiliated Hospital of USTC, MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230000, China
| | - Chenchen Xu
- Department of Radiation Oncology, the First Affiliated Hospital of USTC, MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230000, China
| | - Xinyi Xu
- Department of Radiation Oncology, the First Affiliated Hospital of USTC, MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230000, China
| | - Lingfei Jian
- Department of Radiation Oncology, the First Affiliated Hospital of USTC, MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230000, China
| | - Zhanpeng Zhao
- Department of Radiation Oncology, the First Affiliated Hospital of USTC, MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230000, China
| | - Gang Cai
- Department of Radiation Oncology, the First Affiliated Hospital of USTC, MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230000, China; Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230000, China.
| | - Xuejuan Wang
- Department of Radiation Oncology, the First Affiliated Hospital of USTC, MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230000, China; Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230000, China.
| |
Collapse
|
31
|
Mallet V, Rapisarda C, Minoux H, Ovsjanikov M. Finding antibodies in cryo-EM maps with CrAI. Bioinformatics 2025; 41:btaf157. [PMID: 40203077 PMCID: PMC12077295 DOI: 10.1093/bioinformatics/btaf157] [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: 08/06/2024] [Revised: 03/31/2025] [Accepted: 04/07/2025] [Indexed: 04/11/2025] Open
Abstract
MOTIVATION Therapeutic antibodies have emerged as a prominent class of new drugs due to their high specificity and their ability to bind to several protein targets. Once an initial antibody has been identified, its design and characteristics are refined using structural information, when it is available. Cryo-EM is currently the most effective method to obtain 3D structures. It relies on well-established methods to process raw data into a 3D map, which may, however, be noisy and contain artifacts. To fully interpret these maps the number, position, and structure of antibodies and other proteins present must be determined. Unfortunately, existing automated methods addressing this step have limited accuracy, require additional inputs and high-resolution maps, and exhibit long running times. RESULTS We propose the first fully automatic and efficient method dedicated to finding antibodies in cryo-EM maps: CrAI. This machine learning approach leverages the conserved structure of antibodies and a dedicated novel database that we built to solve this problem. Running a prediction takes only a few seconds, instead of hours, and requires nothing but the cryo-EM map, seamlessly integrating within automated analysis pipelines. Our method can find the location and pose of both Fabs and VHHs at resolutions up to 10 Å and is significantly more reliable than existing approaches. AVAILABILITY AND IMPLEMENTATION We make our method available both in open source github.com/Sanofi-Public/crai and as a ChimeraX bundle (crai).
Collapse
Affiliation(s)
- Vincent Mallet
- LIX, Ecole Polytechnique, IPP Paris, Palaiseau, 91120, France
| | - Chiara Rapisarda
- Integrated Drug Discovery, Structural Biology and Biophysics, Sanofi, Vitry-sur-Seine, 94400, France
| | - Hervé Minoux
- Integrated Drug Discovery, Structural Biology and Biophysics, Sanofi, Vitry-sur-Seine, 94400, France
| | - Maks Ovsjanikov
- LIX, Ecole Polytechnique, IPP Paris, Palaiseau, 91120, France
| |
Collapse
|
32
|
Neiterman EH, Heimowitz A, Ben-Artzi G. A non-parametric approach to particle picking in all frames. J Struct Biol 2025; 217:108201. [PMID: 40334801 DOI: 10.1016/j.jsb.2025.108201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 04/09/2025] [Accepted: 04/10/2025] [Indexed: 05/09/2025]
Abstract
Single-particle cryo-electron microscopy (cryo-EM) has significantly advanced macromolecular structure reconstruction. However, a key limitation is the conventional reliance on micrographs obtained by motion correction and averaging, which inherently loses the richness of information contained within each frame of the original movie. The future of cryo-EM reconstruction ideally involves harnessing the raw signal from every frame to unlock potentially higher quality structures. In this paper, we present a first essential step toward this paradigm shift, that is, a novel, non-parametric method for detecting tomographic projections across all movie frames, using temporal consistency. Our method is inspired by Structure-from-Motion (SfM), and independent of motion correction, CTF estimation, and initial reconstruction. Our experimental results demonstrate reduced outlier rate and accurate particle localization comparable to existing approaches throughout the entire movie sequence.
Collapse
Affiliation(s)
| | - Ayelet Heimowitz
- Department of Electronics and Electrical Engineering, Ariel University, Ariel, Israel.
| | - Gil Ben-Artzi
- School of Computer Science, Ariel University, Ariel, Israel.
| |
Collapse
|
33
|
Wang K, Cao D, Liu L, Fan X, Lin Y, He W, Zhai Y, Xu P, Yan X, Wang H, Zhang X, Yang P. Identification of a nanobody able to catalyze the destruction of the spike-trimer of SARS-CoV-2. Front Med 2025:10.1007/s11684-025-1128-4. [PMID: 40317451 DOI: 10.1007/s11684-025-1128-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 12/16/2024] [Indexed: 05/07/2025]
Abstract
Neutralizing antibodies have been designed to specifically target and bind to the receptor binding domain (RBD) of spike (S) protein to block severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus from attaching to angiotensin converting enzyme 2 (ACE2). This study reports a distinctive nanobody, designated as VHH21, that directly catalyzes the S-trimer into an irreversible transition state through postfusion conformational changes. Derived from camels immunized with multiple antigens, a set of nanobodies with high affinity for the S1 protein displays abilities to neutralize pseudovirion infections with a broad resistance to variants of concern of SARS-CoV-2, including SARS-CoV and BatRaTG13. Importantly, a super-resolution screening and analysis platform based on visual fluorescence probes was designed and applied to monitor single proteins and protein subunits. A spontaneously occurring dimeric form of VHH21 was obtained to rapidly destroy the S-trimer. Structural analysis via cryogenic electron microscopy revealed that VHH21 targets specific conserved epitopes on the S protein, distinct from the ACE2 binding site on the RBD, which destabilizes the fusion process. This research highlights the potential of VHH21 as an abzyme-like nanobody (nanoabzyme) possessing broad-spectrum binding capabilities and highly effective anti-viral properties and offers a promising strategy for combating coronavirus outbreaks.
Collapse
Affiliation(s)
- Kai Wang
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Duanfang Cao
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Lanlan Liu
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiaoyi Fan
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yihuan Lin
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Wenting He
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yunze Zhai
- State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Pingyong Xu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiyun Yan
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Haikun Wang
- State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
| | - Xinzheng Zhang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Pengyuan Yang
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
34
|
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
|
35
|
Kelly DF. Liquid-Electron Microscopy and the Real-Time Revolution. Annu Rev Biophys 2025; 54:1-15. [PMID: 40327441 DOI: 10.1146/annurev-biophys-071624-095107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
Advances in imaging technology enable striking views of life's most minute details. A missing piece of the puzzle, however, is the direct atomic observation of biomolecules in action. Liquid-phase transmission electron microscopy (liquid-EM) is the room-temperature correlate to cryo-electron microscopy, which is leading the resolution revolution in biophysics. This article reviews current challenges and opportunities in the liquid-EM field while discussing technical considerations for specimen enclosures, devices and systems, and scientific data management. Since liquid-EM is gaining traction in the life sciences community, cross talk among the disciplines of materials and life sciences is needed to disseminate knowledge of best practices along with high-level user engagement. How liquid-EM technology is inspiring the real-time revolution in molecular microscopy is also discussed. Looking ahead, the new movement can be better supported through open resource sharing and partnerships among academic, industry, and federal organizations, which may benefit from the scientific equity foundational to the technique.
Collapse
|
36
|
Song XY, Xia Y, Zhang JT, Liu YJ, Qi H, Wei XY, Hu H, Xia Y, Liu X, Ma YF, Jia N. Bacterial reverse transcriptase synthesizes long poly-A-rich cDNA for antiphage defense. Science 2025:eads4639. [PMID: 40310939 DOI: 10.1126/science.ads4639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 01/27/2025] [Accepted: 04/22/2025] [Indexed: 05/03/2025]
Abstract
Prokaryotic defense-associated reverse transcriptases (DRTs) were recently identified with antiviral functions; however, their functional mechanisms remain largely unexplored. Here we show that DRT9 forms a hexameric complex with its upstream non-coding RNA (ncRNA) to mediate antiphage defense by inducing cell growth arrest via abortive infection. Upon phage infection, the phage-encoded ribonucleotide reductase NrdAB complex elevates intracellular dATP levels, activating DRT9 to synthesize long, poly-A-rich single-stranded cDNA, which likely sequesters the essential phage SSB protein and disrupts phage propagation. We further determined the cryo-electron microscopy structure of the DRT9-ncRNA hexamer complex, providing mechanistic insights into its cDNA synthesis. These findings highlight the diversity of RT-based antiviral defense mechanisms, expand our understanding of RT biological functions, and provide a structural basis for developing DRT9-based biotechnological tools.
Collapse
Affiliation(s)
- Xin-Yi Song
- Department of Biochemistry, SUSTech Homeostatic Medicine Institute, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Yushan Xia
- Department of Biochemistry, SUSTech Homeostatic Medicine Institute, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Jun-Tao Zhang
- Department of Biochemistry, SUSTech Homeostatic Medicine Institute, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Yu-Jun Liu
- Department of Biochemistry, SUSTech Homeostatic Medicine Institute, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Hua Qi
- Department of Biochemistry, SUSTech Homeostatic Medicine Institute, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Xin-Yang Wei
- Department of Biochemistry, SUSTech Homeostatic Medicine Institute, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Hailiang Hu
- Department of Biochemistry, SUSTech Homeostatic Medicine Institute, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Yu Xia
- School of Environmental Science and Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Xue Liu
- Department of Pathogen Biology, Base for International Science and Technology Cooperation: Carson Cancer Stem Cell Vaccines R&D Center, International Cancer Center,Shenzhen University Medical School, Shenzhen, Guangdong, China
| | - Ying-Fei Ma
- Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
| | - Ning Jia
- Department of Biochemistry, SUSTech Homeostatic Medicine Institute, School of Medicine, Southern University of Science and Technology, Shenzhen, China
- Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, Guangdong, China
- Key University Laboratory of Metabolism and Health of Guangdong, Institute for Biological Electron Microscopy, Southern University of Science and Technology, Shenzhen, Guangdong, China
| |
Collapse
|
37
|
Fatema K, Snowden JS, Watson A, Sherry L, Ranson NA, Stonehouse NJ, Rowlands DJ. A VLP vaccine platform comprising the core protein of hepatitis B virus with N-terminal antigen capture. Int J Biol Macromol 2025; 305:141152. [PMID: 39961558 DOI: 10.1016/j.ijbiomac.2025.141152] [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: 12/18/2024] [Revised: 02/05/2025] [Accepted: 02/14/2025] [Indexed: 02/25/2025]
Abstract
Nanoparticle presentation systems offer the potential to develop new vaccines rapidly in response to emerging diseases, a public health need that has become increasingly evident in the wake of the COVID-19 pandemic. Previously, we reported a nanoparticle scaffold system termed VelcroVax. This was constructed by insertion of a high affinity SUMO binding protein (Affimer), able to recognise a SUMO peptide tag, into the major immunodominant region of VLPs assembled from a tandem (fused dimer) form of hepatitis B virus (HBV) core protein (HBc). Here we describe an alternative form, termed N-VelcroVax, a VLP vaccine platform assembled from a monomeric HBc protein (N-anti-SUMO Affimer HBc 190) with the Affimer inserted at the N-terminus. In contrast to the tandem form of VelcroVax, N-VelcroVax VLPs were expressed well in E. coli. The VLPs effectively bound SUMO-tagged Junín virus glycoprotein, gp1 as assessed by structural and serological analyses. Cryo-EM characterisation of N-VelcroVax complexed with a SUMO-Junín gp1 showed continuous density attributable to the fused Affimer, in addition to evidence of target antigen capture. Collectively, these data suggest that N-VelcroVax has potential as a versatile next generation vaccine scaffold.
Collapse
Affiliation(s)
- Kaniz Fatema
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Joseph S Snowden
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Alexander Watson
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Lee Sherry
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Neil A Ranson
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Nicola J Stonehouse
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, United Kingdom.
| | - David J Rowlands
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, United Kingdom.
| |
Collapse
|
38
|
Rimkute I, Olia AS, Suleiman M, Woods KD, Bylund T, Morano NC, Tully ES, Verardi R, Bao S, Beddall MH, Chaimongkol N, Donaldson MM, Du R, Dulan CNM, Gorman J, Henry AR, Schramm CA, Sosnovtsev SV, Stephens T, Todd JP, Tsybovsky Y, Douek DC, Green KY, Rawi R, Shapiro L, Zhou T, Kwong PD, Roederer M. A broadly protective human antibody for GI genogroup noroviruses. Nat Microbiol 2025; 10:1227-1239. [PMID: 40211068 DOI: 10.1038/s41564-025-01952-6] [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: 08/06/2024] [Accepted: 02/06/2025] [Indexed: 04/12/2025]
Abstract
Noroviruses infect millions each year, and while effective countermeasures are eagerly sought, none have been reported for the GI genogroup, first described more than 50 years ago. Here, to provide insight into GI norovirus neutralization, we isolated a broad GI antibody, 16E10, from a human blood donor and showed it neutralizes noroviruses in human enteroid cultures and abrogates or reduces infection in rhesus macaques. The cryogenic electron microscopy reconstruction of 16E10 with a norovirus protruding-domain dimer at 2.56-Å resolution reveals an exceptionally large binding surface, overlapping an antibody supersite, distal from host receptor-binding or cofactor-binding sites. Cryogenic electron microscopy reconstructions with virus-like particles (VLPs) showed that 16E10 disrupts protruding domains on the VLP surface and disassembles VLPs, altering viral organization required for avidity. While its epitope was generally conserved, 16E10 recognized multiple sequence-divergent residues, binding to which was enabled by corresponding cavities in the 16E10-norovirus interface. Broad recognition of noroviruses can thus incorporate sequence-divergent residues, through a cavity-based mechanism of diversity tolerance.
Collapse
Affiliation(s)
- Inga Rimkute
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Adam S Olia
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Mehin Suleiman
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Kamron D Woods
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Tatsiana Bylund
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Nicholas C Morano
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
- Department of Biochemistry and Molecular Biophysics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Ena S Tully
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Raffaello Verardi
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Saran Bao
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Margaret H Beddall
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Natthawan Chaimongkol
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Mitzi M Donaldson
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Renguang Du
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Caitlyn N M Dulan
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Jason Gorman
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Amy R Henry
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Chaim A Schramm
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Stanislav V Sosnovtsev
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Tyler Stephens
- Electron Microscopy Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - John-Paul Todd
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Yaroslav Tsybovsky
- Electron Microscopy Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Daniel C Douek
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Kim Y Green
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Reda Rawi
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Lawrence Shapiro
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
- Department of Biochemistry and Molecular Biophysics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Tongqing Zhou
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Peter D Kwong
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA.
- Department of Biochemistry and Molecular Biophysics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA.
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA.
| | - Mario Roederer
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA.
| |
Collapse
|
39
|
Wollweber F, Xu J, Ponce-Toledo RI, Marxer F, Rodrigues-Oliveira T, Pössnecker A, Luo ZH, Malit JJL, Kokhanovska A, Wieczorek M, Schleper C, Pilhofer M. Microtubules in Asgard archaea. Cell 2025; 188:2451-2464.e26. [PMID: 40120574 DOI: 10.1016/j.cell.2025.02.027] [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: 06/23/2024] [Revised: 01/27/2025] [Accepted: 02/25/2025] [Indexed: 03/25/2025]
Abstract
Microtubules are a hallmark of eukaryotes. Archaeal and bacterial homologs of tubulins typically form homopolymers and non-tubular superstructures. The origin of heterodimeric tubulins assembling into microtubules remains unclear. Here, we report the discovery of microtubule-forming tubulins in Asgard archaea, the closest known relatives of eukaryotes. These Asgard tubulins (AtubA/B) are closely related to eukaryotic α/β-tubulins and the enigmatic bacterial tubulins BtubA/B. Proteomics of Candidatus Lokiarchaeum ossiferum showed that AtubA/B were highly expressed. Cryoelectron microscopy structures demonstrate that AtubA/B form eukaryote-like heterodimers, which assembled into 5-protofilament bona fide microtubules in vitro. The additional paralog AtubB2 lacks a nucleotide-binding site and competitively displaced AtubB. These AtubA/B2 heterodimers polymerized into 7-protofilament non-canonical microtubules. In a sub-population of Ca. Lokiarchaeum ossiferum cells, cryo-tomography revealed tubular structures, while expansion microscopy identified AtubA/B cytoskeletal assemblies. Our findings suggest a pre-eukaryotic origin of microtubules and provide a framework for understanding the fundamental principles of microtubule assembly.
Collapse
Affiliation(s)
- Florian Wollweber
- Department of Biology, Institute of Molecular Biology & Biophysics, Eidgenössische Technische Hochschule Zürich, Otto-Stern-Weg 5, 8093 Zürich, Switzerland
| | - Jingwei Xu
- Department of Biology, Institute of Molecular Biology & Biophysics, Eidgenössische Technische Hochschule Zürich, Otto-Stern-Weg 5, 8093 Zürich, Switzerland
| | - Rafael I Ponce-Toledo
- Department of Functional and Evolutionary Ecology, Archaea Biology and Ecogenomics Unit, University of Vienna, Djerassiplatz 1, 1030 Vienna, Austria
| | - Florina Marxer
- Department of Biology, Institute of Molecular Biology & Biophysics, Eidgenössische Technische Hochschule Zürich, Otto-Stern-Weg 5, 8093 Zürich, Switzerland
| | - Thiago Rodrigues-Oliveira
- Department of Functional and Evolutionary Ecology, Archaea Biology and Ecogenomics Unit, University of Vienna, Djerassiplatz 1, 1030 Vienna, Austria
| | - Anja Pössnecker
- Department of Biology, Institute of Molecular Biology & Biophysics, Eidgenössische Technische Hochschule Zürich, Otto-Stern-Weg 5, 8093 Zürich, Switzerland
| | - Zhen-Hao Luo
- Department of Functional and Evolutionary Ecology, Archaea Biology and Ecogenomics Unit, University of Vienna, Djerassiplatz 1, 1030 Vienna, Austria
| | - Jessie James Limlingan Malit
- Department of Biology, Institute of Molecular Biology & Biophysics, Eidgenössische Technische Hochschule Zürich, Otto-Stern-Weg 5, 8093 Zürich, Switzerland
| | - Anastasiia Kokhanovska
- Department of Biology, Institute of Molecular Biology & Biophysics, Eidgenössische Technische Hochschule Zürich, Otto-Stern-Weg 5, 8093 Zürich, Switzerland
| | - Michal Wieczorek
- Department of Biology, Institute of Molecular Biology & Biophysics, Eidgenössische Technische Hochschule Zürich, Otto-Stern-Weg 5, 8093 Zürich, Switzerland
| | - Christa Schleper
- Department of Functional and Evolutionary Ecology, Archaea Biology and Ecogenomics Unit, University of Vienna, Djerassiplatz 1, 1030 Vienna, Austria
| | - Martin Pilhofer
- Department of Biology, Institute of Molecular Biology & Biophysics, Eidgenössische Technische Hochschule Zürich, Otto-Stern-Weg 5, 8093 Zürich, Switzerland.
| |
Collapse
|
40
|
Xiong Q, Zhu Z, Li T, Li X, Zhou Z, Chao Y, Yang C, Feng S, Qu Q, Li D. Molecular architecture of human LYCHOS involved in lysosomal cholesterol signaling. Nat Struct Mol Biol 2025; 32:905-913. [PMID: 39824977 DOI: 10.1038/s41594-024-01474-5] [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: 04/22/2024] [Accepted: 12/10/2024] [Indexed: 01/20/2025]
Abstract
Lysosomal membrane protein LYCHOS (lysosomal cholesterol signaling) translates cholesterol abundance to mammalian target of rapamycin activation. Here we report the 2.11-Å structure of human LYCHOS, revealing a unique fusion architecture comprising a G-protein-coupled receptor (GPCR)-like domain and a transporter domain that mediates homodimer assembly. The NhaA-fold transporter harbors a previously uncharacterized intramembrane Na+ pocket. The GPCR-like domain is stabilized, by analogy to canonical GPCRs, in an inactive state through 'tethered antagonism' by a lumenal loop and strong interactions at the cytosol side preventing the hallmark swing of the sixth transmembrane helix seen in active GPCRs. A cholesterol molecule and an associated docosahexaenoic acid (DHA)-phospholipid are entrapped between the transporter and GPCR-like domains, with the DHA-phospholipid occupying a pocket previously implicated in cholesterol sensing, indicating inter-domain coupling via dynamic lipid-protein interactions. Our work provides a high-resolution framework for functional investigations of the understudied LYCHOS protein.
Collapse
Affiliation(s)
- Qi Xiong
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Key Laboratory of RNA Innovation, Science, and Engineering; Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Zhini Zhu
- Institutes of Biomedical Sciences, Shanghai Key Laboratory of Medical Epigenetics, International Colaboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Department of Systems Biology for Medicine, Fudan University, Shanghai, China
| | - Tingting Li
- Key Laboratory of RNA Innovation, Science, and Engineering; Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Xiaotian Li
- Lipid Metabolism and Chemical Biology Unit, Key Laboratory of Immune Response and Immunotherapy, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zixuan Zhou
- Institutes of Biomedical Sciences, Shanghai Key Laboratory of Medical Epigenetics, International Colaboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Department of Systems Biology for Medicine, Fudan University, Shanghai, China
| | - Yulin Chao
- Institutes of Biomedical Sciences, Shanghai Key Laboratory of Medical Epigenetics, International Colaboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Department of Systems Biology for Medicine, Fudan University, Shanghai, China
| | - Chuanhui Yang
- Institutes of Biomedical Sciences, Shanghai Key Laboratory of Medical Epigenetics, International Colaboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Department of Systems Biology for Medicine, Fudan University, Shanghai, China
| | - Suihan Feng
- Lipid Metabolism and Chemical Biology Unit, Key Laboratory of Immune Response and Immunotherapy, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
| | - Qianhui Qu
- Institutes of Biomedical Sciences, Shanghai Key Laboratory of Medical Epigenetics, International Colaboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Department of Systems Biology for Medicine, Fudan University, Shanghai, China.
| | - Dianfan Li
- Key Laboratory of RNA Innovation, Science, and Engineering; Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China.
- School of Agriculture and Biotechnology, Sun Yat-Sen University, Shenzhen, China.
| |
Collapse
|
41
|
Choi W, Li C, Chen Y, Wang Y, Cheng Y. Structural dynamics of human fatty acid synthase in the condensing cycle. Nature 2025; 641:529-536. [PMID: 39978408 PMCID: PMC12058526 DOI: 10.1038/s41586-025-08782-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: 05/07/2024] [Accepted: 02/12/2025] [Indexed: 02/22/2025]
Abstract
Long-chain fatty acids are the building blocks of fat in human bodies. In mammals, fatty acid synthase (FASN) contains multiple enzymatic domains to catalyse all chemical reactions needed for de novo fatty acid synthesis1. Although the chemical reactions carried out by these enzymatic domains are well defined, how the dimeric FASN with an open architecture continuously catalyses such reactions to synthesize a complete fatty acid remains elusive. Here, using a strategy of tagging and purifying endogenous FASN in HEK293T cells for single-particle cryo-electron microscopy studies, we characterized the structural dynamics of endogenous human FASN. We captured conformational snapshots of various functional substates in the condensing cycle and developed a procedure to analyse the particle distribution landscape of FASN with different orientations between its condensing and modifying wings. Together, our findings reveal that FASN function does not require a large rotational motion between its two main functional domains during the condensing cycle, and that the catalytic reactions in the condensing cycle carried out by the two monomers are unsynchronized. Our data thus provide a new composite view of FASN dynamics during the fatty acid synthesis condensing cycle.
Collapse
Affiliation(s)
- Wooyoung Choi
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, USA
| | - Chengmin Li
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, USA
| | - Yifei Chen
- Howard Hughes Medical Institute, University of California San Francisco, San Francisco, CA, USA
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
| | - YongQiang Wang
- Howard Hughes Medical Institute, University of California San Francisco, San Francisco, CA, USA
| | - Yifan Cheng
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, USA.
- Howard Hughes Medical Institute, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
42
|
Kim J, Ko S, Choi C, Bae J, Byeon H, Seok C, Choi HJ. Structural insights into small-molecule agonist recognition and activation of complement receptor C3aR. EMBO J 2025; 44:2803-2826. [PMID: 40195498 DOI: 10.1038/s44318-025-00429-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: 10/02/2024] [Revised: 03/07/2025] [Accepted: 03/07/2025] [Indexed: 04/09/2025] Open
Abstract
The complement system plays crucial roles in innate immunity and inflammatory responses. The anaphylatoxin C3a mediates pro-inflammatory and chemotactic functions through the G protein-coupled receptor C3aR. While the active structure of the C3a-C3aR-Gi complex has been determined, the inactive conformation and activation mechanism of C3aR remain elusive. Here we report the cryo-EM structure of ligand-free, G protein-free C3aR, providing insights into its inactive conformation. In addition, we determine the structures of C3aR in complex with the synthetic small-molecule agonist JR14a in two distinct conformational states: a G protein-free intermediate, and a fully active Gi-bound state. The structure of the active JR14a-bound C3aR reveals that JR14a engages in highly conserved interactions with C3aR, similar to the binding of the C-terminal pentapeptide of C3a, along with JR14a-specific interactions. Structural comparison of C3aR in the apo, intermediate, and fully active states provides novel insights into the conformational landscape and activation mechanism of C3aR and defines a molecular basis explaining its high basal activity. Our results may aid in the rational design of therapeutics targeting complement-related inflammatory disorders.
Collapse
Affiliation(s)
- Jinuk Kim
- Department of Biological Sciences, Seoul National University, Seoul, 08826, Republic of Korea
- Division of Biological Science and Technology, Yonsei University, Wonju, 26493, Republic of Korea
| | - Saebom Ko
- Department of Biological Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Chulwon Choi
- Department of Biological Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jungnam Bae
- Department of Biological Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hyeonsung Byeon
- Department of Chemistry, Seoul National University, Seoul, 08826, Republic of Korea
| | - Chaok Seok
- Department of Chemistry, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hee-Jung Choi
- Department of Biological Sciences, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
43
|
Sung MW, Hu K, Hurlimann LM, Lees JA, Fennell KF, West MA, Costales C, Rodrigues AD, Zimmermann I, Dawson RJP, Liu S, Han S. Cyclosporine A sterically inhibits statin transport by solute carrier OATP1B1. J Biol Chem 2025; 301:108484. [PMID: 40199401 PMCID: PMC12127550 DOI: 10.1016/j.jbc.2025.108484] [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: 08/08/2024] [Revised: 03/24/2025] [Accepted: 03/31/2025] [Indexed: 04/10/2025] Open
Abstract
Members of the Organic Anion Transporter Polypeptides (OATP) are integral membrane proteins responsible for facilitating the transport of organic anions across the cell membrane. OATP1B1 (SLCO1B1), the prototypic OATP family member, is the most abundant uptake transporter in the liver and a key mediator of the hepatic uptake and clearance of numerous endogenous and xenobiotic compounds. It serves as a locus of important drug-drug interactions, such as those between statins and cyclosporine A, and carries the potential to enable liver-targeting therapeutics. In this study, we report cryo-EM structures of OATP1B1 and its complexes with one of its statin substrates, atorvastatin, and an inhibitor, cyclosporine A. This structural analysis has yielded insights into the mechanisms underlying the OATP1B1-mediated transport of statins and the inhibitory effect of cyclosporine A. These findings contribute to a better understanding of the molecular processes involved in drug transport and offer potential avenues for the development of targeted medications for liver-related conditions.
Collapse
Affiliation(s)
- Min Woo Sung
- Discovery Sciences, Discovery & Early Development, Pfizer Inc, Groton, Connecticut, USA
| | - Kuan Hu
- Discovery Sciences, Discovery & Early Development, Pfizer Inc, Groton, Connecticut, USA
| | | | - Joshua A Lees
- Discovery Sciences, Discovery & Early Development, Pfizer Inc, Groton, Connecticut, USA
| | - Kimberly F Fennell
- Discovery Sciences, Discovery & Early Development, Pfizer Inc, Groton, Connecticut, USA
| | - Mark A West
- Pharmacokinetics, Dynamics, and Metabolism, Discovery & Early Development, Pfizer Inc, Groton, Connecticut, USA
| | - Chester Costales
- Pharmacokinetics, Dynamics, and Metabolism, Discovery & Early Development, Pfizer Inc, Groton, Connecticut, USA
| | - Amilcar David Rodrigues
- Pharmacokinetics, Dynamics, and Metabolism, Discovery & Early Development, Pfizer Inc, Groton, Connecticut, USA
| | | | | | - Shenping Liu
- Discovery Sciences, Discovery & Early Development, Pfizer Inc, Groton, Connecticut, USA.
| | - Seungil Han
- Discovery Sciences, Discovery & Early Development, Pfizer Inc, Groton, Connecticut, USA.
| |
Collapse
|
44
|
Taniguchi R, Orniacki C, Kreysing JP, Zila V, Zimmerli CE, Böhm S, Turoňová B, Kräusslich HG, Doye V, Beck M. Nuclear pores safeguard the integrity of the nuclear envelope. Nat Cell Biol 2025; 27:762-775. [PMID: 40205196 DOI: 10.1038/s41556-025-01648-3] [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: 02/28/2024] [Accepted: 02/25/2025] [Indexed: 04/11/2025]
Abstract
Nuclear pore complexes (NPCs) mediate nucleocytoplasmic exchange, which is essential for eukaryotes. Mutations in the central scaffolding components of NPCs are associated with genetic diseases, but how they manifest only in specific tissues remains unclear. This is exemplified in Nup133-deficient mouse embryonic stem cells, which grow normally during pluripotency, but differentiate poorly into neurons. Here, using an innovative in situ structural biology approach, we show that Nup133-/- mouse embryonic stem cells have heterogeneous NPCs with non-canonical symmetries and missing subunits. During neuronal differentiation, Nup133-deficient NPCs frequently disintegrate, resulting in abnormally large nuclear envelope openings. We propose that the elasticity of the NPC scaffold has a protective function for the nuclear envelope and that its perturbation becomes critical under conditions that impose an increased mechanical load onto nuclei.
Collapse
Affiliation(s)
- Reiya Taniguchi
- Department of Molecular Sociology, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
- RIKEN Center for Integrative Medical Sciences, Tsurumi-ku, Yokohama, Japan
| | - Clarisse Orniacki
- Université Paris Cité, CNRS, Institut Jacques Monod, Paris, France
- The Neuro - Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec, Canada
| | - Jan Philipp Kreysing
- Department of Molecular Sociology, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
- IMPRS on Cellular Biophysics, Frankfurt am Main, Germany
| | - Vojtech Zila
- Department of Infectious Diseases, Virology, Heidelberg University, Heidelberg, Germany
- AskBio GmbH, Heidelberg, Germany
| | - Christian E Zimmerli
- Department of Molecular Sociology, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
- Institute of Physics, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Stefanie Böhm
- Department of Molecular Sociology, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
| | - Beata Turoňová
- Department of Molecular Sociology, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
| | - Hans-Georg Kräusslich
- Department of Infectious Diseases, Virology, Heidelberg University, Heidelberg, Germany
- German Centre for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany
| | - Valérie Doye
- Université Paris Cité, CNRS, Institut Jacques Monod, Paris, France.
| | - Martin Beck
- Department of Molecular Sociology, Max Planck Institute of Biophysics, Frankfurt am Main, Germany.
- Institute of Biochemistry, Goethe University Frankfurt, Frankfurt am Main, Germany.
| |
Collapse
|
45
|
Coleman B, Bedi S, Hill JH, Morris J, Manthei KA, Hart RC, He Y, Shah AS, Jerome WG, Vaisar T, Bornfeldt KE, Song H, Segrest JP, Heinecke JW, Aller SG, Tesmer JJG, Davidson WS. Lecithin:cholesterol acyltransferase binds a discontinuous binding site on adjacent apolipoprotein A-I belts in HDL. J Lipid Res 2025; 66:100786. [PMID: 40147634 PMCID: PMC12049944 DOI: 10.1016/j.jlr.2025.100786] [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: 01/21/2025] [Revised: 03/11/2025] [Accepted: 03/23/2025] [Indexed: 03/29/2025] Open
Abstract
Lecithin:cholesterol acyltransferase (LCAT) is a high-density lipoprotein (HDL) modifying protein that profoundly affects the composition and function of HDL subspecies. The cholesterol esterification activity of LCAT is dramatically increased by apolipoprotein A-I (APOA1) on HDL, but the mechanism remains unclear. Using site-directed mutagenesis, cross-linking, mass spectrometry, electron microscopy, protein engineering, and molecular docking, we identified two LCAT binding sites formed by helices 4 and 6 from two antiparallel APOA1 molecules in HDL. Although the reciprocating APOA1 "belts" form two ostensibly symmetrical binding locations, LCAT can adopt distinct orientations at each site, as shown by our 9.8 Å cryoEM envelope. In one case, LCAT membrane binding domains align with the APOA1 belts and, in the other, the HDL phospholipids. By introducing disulfide bonds between the APOA1 helical domains, we demonstrated that LCAT does not require helical separation during its reaction cycle. This indicates that LCAT, anchored to APOA1 belts, accesses substrates and deposits products through interactions with the planar lipid surface. This model of the LCAT/APOA1 interaction provides insights into how LCAT and possibly other HDL-modifying factors engage the APOA1 scaffold, offering potential strategies to enhance LCAT activity in individuals with genetic defects.
Collapse
Affiliation(s)
- Bethany Coleman
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH, USA
| | - Shimpi Bedi
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - John H Hill
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jamie Morris
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Kelly A Manthei
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | - Rachel C Hart
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Yi He
- Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Amy S Shah
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center and the University of Cincinnati, Cincinnati, OH, USA
| | - W Gray Jerome
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Tomas Vaisar
- Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Karin E Bornfeldt
- Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Hyun Song
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jere P Segrest
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jay W Heinecke
- Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Stephen G Aller
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - John J G Tesmer
- Departments of Biological Sciences and Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, USA
| | - W Sean Davidson
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, OH, USA.
| |
Collapse
|
46
|
An J, Imasaki T, Narita A, Niwa S, Sasaki R, Makino T, Nitta R, Kikkawa M. Dimerization of GAS2 mediates crosslinking of microtubules and F-actin. EMBO J 2025; 44:2997-3024. [PMID: 40169809 DOI: 10.1038/s44318-025-00415-2] [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: 09/12/2024] [Revised: 03/05/2025] [Accepted: 03/12/2025] [Indexed: 04/03/2025] Open
Abstract
The spectraplakin family protein GAS2 was originally identified as a growth arrest-specific protein, and recent studies have revealed its involvement in multiple cellular processes. Its dual interaction with actin filaments and microtubules highlights its essential role in cytoskeletal organization, such as cell division, apoptosis, and possibly tumorigenesis. However, the structural basis of cytoskeletal dynamics regulation by GAS2 remains unclear. In this study, we present cryo-electron microscopy structures of the GAS2 type 3 calponin homology domain (CH3) in complex with F-actin at 2.8 Å resolution, thus solving the first type CH3 domain structure bound to F-actin and confirming its actin-binding activity. We also provide the first near-atomic resolution cryo-EM structure of the GAS2-GAR domain bound to microtubules and identify conserved microtubule-binding residues. Our biochemical experiments show that GAS2 promotes microtubule nucleation and polymerization, and that its C-terminal region is essential for dimerization, bundling of both F-actin and microtubules, and microtubule nucleation. As mutations leading to expression of C-terminally truncated GAS2 have been linked to hearing loss, these findings suggest that the disruption of GAS2-dependent cytoskeletal organisation could underlie auditory dysfunction.
Collapse
Affiliation(s)
- Jiancheng An
- Department of Cell Biology and Anatomy, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tsuyoshi Imasaki
- Division of Structural Medicine and Anatomy, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Akihiro Narita
- Structural Biology Research Center, Graduate School of Science, Nagoya University, Nagoya, Aichi, Japan
| | - Shinsuke Niwa
- Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | - Ryohei Sasaki
- Department of Cell Biology and Anatomy, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tsukasa Makino
- Department of Cell Biology and Anatomy, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Ryo Nitta
- Division of Structural Medicine and Anatomy, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Masahide Kikkawa
- Department of Cell Biology and Anatomy, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
47
|
Lu X, Yan M, Cai Y, Song X, Chen H, Du M, Wang Z, Li J, Niu L, Zeng F, Hao Q, Zhang H. A large, general and modular DARPin-apoferritin scaffold enables the visualization of small proteins by cryo-EM. IUCRJ 2025; 12:393-402. [PMID: 40277178 PMCID: PMC12044855 DOI: 10.1107/s2052252525003021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Accepted: 04/03/2025] [Indexed: 04/26/2025]
Abstract
Single-particle cryo-electron microscopy (cryo-EM) has emerged as an indispensable technique in structural biology that is pivotal for deciphering protein architectures. However, the medium-sized proteins (30-40 kDa) that are prevalent in both eukaryotic and prokaryotic organisms often elude the resolving capabilities of contemporary cryo-EM methods. To address this challenge, we engineered a scaffold strategy that securely anchors proteins of interest to a robust, symmetric base via a selective adapter. Our most efficacious constructs, namely models 4 and 6c, feature a designed ankyrin-repeat protein (DARPin) rigidly linked to an octahedral human apoferritin via a helical linker. By utilizing these large, highly symmetric scaffolds (∼1 MDa), we achieved near-atomic-resolution cryo-EM structures of green fluorescent protein (GFP) and maltose-binding protein (MBP), revealing nearly all side-chain densities of GFP and the distinct structural features of MBP. The modular design of our scaffold allows the adaptation of new DARPins through minor amino-acid-sequence modifications, enabling the binding and visualization of a diverse array of proteins. The high symmetry and near-spherical shape of the scaffold not only mitigates the prevalent challenge of preferred particle orientation in cryo-EM but also significantly reduces the demands of image collection and data processing. This approach presents a versatile solution, breaking through the size constraints that have traditionally limited single-particle cryo-EM.
Collapse
Affiliation(s)
- Xin Lu
- Institute of High Energy PhysicsChinese Academy of SciencesBeijing100000People’s Republic of China
- Spallation Neutron Source Science CenterChinese Academy of SciencesDongguan523000People’s Republic of China
- Department of Biology, Southern University of Science and Technology, Shenzhen518055, People’s Republic of China
- Bio-Tech Center, Shenzhen Medical Academy of Research and Translation (SMART), Shenzhen518107, People’s Republic of China
| | - Ming Yan
- Institute of High Energy PhysicsChinese Academy of SciencesBeijing100000People’s Republic of China
- Spallation Neutron Source Science CenterChinese Academy of SciencesDongguan523000People’s Republic of China
- Department of Biology, Southern University of Science and Technology, Shenzhen518055, People’s Republic of China
- School of Biomedical SciencesThe University of Hong KongHong KongPeople’s Republic of China
| | - Yang Cai
- Department of Biology, Southern University of Science and Technology, Shenzhen518055, People’s Republic of China
- The Chinese University of Hong Kong (Shenzhen)ShenzhenPeople’s Republic of China
| | - Xi Song
- Department of Biology, Southern University of Science and Technology, Shenzhen518055, People’s Republic of China
| | - Huan Chen
- Department of Biology, Southern University of Science and Technology, Shenzhen518055, People’s Republic of China
| | - Mengtan Du
- Department of Biology, Southern University of Science and Technology, Shenzhen518055, People’s Republic of China
| | - Zhenyi Wang
- Department of Biology, Southern University of Science and Technology, Shenzhen518055, People’s Republic of China
- Hefei National Laboratory for Physical Sciences at the Microscale and School of Life SciencesUniversity of Science and Technology of ChinaHefei230026People’s Republic of China
- Key Laboratory of Structural BiologyChinese Academy of SciencesHefei230026People’s Republic of China
| | - Jia’an Li
- Department of Biology, Southern University of Science and Technology, Shenzhen518055, People’s Republic of China
| | - Liwen Niu
- Hefei National Laboratory for Physical Sciences at the Microscale and School of Life SciencesUniversity of Science and Technology of ChinaHefei230026People’s Republic of China
- Key Laboratory of Structural BiologyChinese Academy of SciencesHefei230026People’s Republic of China
| | - Fuxing Zeng
- Department of Biology, Southern University of Science and Technology, Shenzhen518055, People’s Republic of China
| | - Quan Hao
- Institute of High Energy PhysicsChinese Academy of SciencesBeijing100000People’s Republic of China
- Spallation Neutron Source Science CenterChinese Academy of SciencesDongguan523000People’s Republic of China
- School of Biomedical SciencesThe University of Hong KongHong KongPeople’s Republic of China
| | - Hongmin Zhang
- Institute of High Energy PhysicsChinese Academy of SciencesBeijing100000People’s Republic of China
- Spallation Neutron Source Science CenterChinese Academy of SciencesDongguan523000People’s Republic of China
- Department of Biology, Southern University of Science and Technology, Shenzhen518055, People’s Republic of China
| |
Collapse
|
48
|
Ma Q, Ma K, Dong Y, Meng Y, Zhao J, Li R, Bai Q, Wu D, Jiang D, Sun J, Zhao Y. Binding mechanism and antagonism of the vesicular acetylcholine transporter VAChT. Nat Struct Mol Biol 2025; 32:818-827. [PMID: 39806024 DOI: 10.1038/s41594-024-01462-9] [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: 02/20/2024] [Accepted: 11/29/2024] [Indexed: 01/16/2025]
Abstract
The vesicular acetylcholine transporter (VAChT) has a pivotal role in packaging and transporting acetylcholine for exocytotic release, serving as a vital component of cholinergic neurotransmission. Dysregulation of its function can result in neurological disorders. It also serves as a target for developing radiotracers to quantify cholinergic neuron deficits in neurodegenerative conditions. Here we unveil the cryo-electron microscopy structures of human VAChT in its apo state, the substrate acetylcholine-bound state and the inhibitor vesamicol-bound state. These structures assume a lumen-facing conformation, offering a clear depiction of architecture of VAChT. The acetylcholine-bound structure provides a detailed understanding of how VAChT recognizes its substrate, shedding light on the coupling mechanism of protonation and substrate binding. Meanwhile, the vesamicol-bound structure reveals the binding mode of vesamicol to VAChT, laying the structural foundation for the design of the next generation of radioligands targeting VAChT.
Collapse
Affiliation(s)
- Qiao Ma
- Key Laboratory of Biomacromolecules (CAS), National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Kunpeng Ma
- Interdisciplinary Center for Brain Information, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yanli Dong
- Key Laboratory of Biomacromolecules (CAS), National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Yufei Meng
- Key Laboratory of Biomacromolecules (CAS), National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jun Zhao
- Peking University Institute of Advanced Agricultural Sciences, Shandong Laboratory of Advanced Agricultural Sciences at Weifang, Weifang, China
| | - Renjie Li
- Key Laboratory of Biomacromolecules (CAS), National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Qinru Bai
- Key Laboratory of Biomacromolecules (CAS), National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Di Wu
- University of Chinese Academy of Sciences, Beijing, China
- Laboratory of Soft Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing, China
| | - Daohua Jiang
- University of Chinese Academy of Sciences, Beijing, China
- Laboratory of Soft Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing, China
| | - Jianyuan Sun
- University of Chinese Academy of Sciences, Beijing, China
- Interdisciplinary Center for Brain Information, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yan Zhao
- Key Laboratory of Biomacromolecules (CAS), National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders and National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
49
|
Kinman LF, Carreira MV, Powell BM, Davis JH. Automated model-free analysis of cryo-EM volume ensembles with SIREn. Structure 2025; 33:974-987.e4. [PMID: 40068687 PMCID: PMC12055258 DOI: 10.1016/j.str.2025.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 12/16/2024] [Accepted: 02/12/2025] [Indexed: 03/19/2025]
Abstract
Cryogenic electron microscopy (cryo-EM) has the potential to capture snapshots of proteins in motion and generate hypotheses linking conformational states to biological function. This potential has been increasingly realized by the advent of machine learning models that allow 100s-1,000s of 3D density maps to be generated from a single dataset. How to identify distinct structural states within these volume ensembles and quantify their relative occupancies remain open questions. Here, we present an approach to inferring variable regions directly from a volume ensemble based on the statistical co-occupancy of voxels, as well as a 3D convolutional neural network that predicts binarization thresholds for volumes in an unbiased and automated manner. We show that these tools recapitulate known heterogeneity in a variety of simulated and real cryo-EM datasets and highlight how integrating these tools with existing data processing pipelines enables improved particle curation.
Collapse
Affiliation(s)
- Laurel F Kinman
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Maria V Carreira
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Barrett M Powell
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Joseph H Davis
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA; Computational and Systems Biology Graduate Program, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
50
|
Ruan W, Li T, Bang IH, Lee J, Deng W, Ma X, Luo C, Du F, Yoo SH, Kim B, Li J, Yuan X, Figarella K, An YA, Wang YY, Liang Y, DeBerge M, Zhang D, Zhou Z, Wang Y, Gorham JM, Seidman JG, Seidman CE, Aranki SF, Nair R, Li L, Narula J, Zhao Z, Gorfe AA, Muehlschlegel JD, Tsai KL, Eltzschig HK. BMAL1-HIF2A heterodimer modulates circadian variations of myocardial injury. Nature 2025; 641:1017-1028. [PMID: 40269168 PMCID: PMC12095075 DOI: 10.1038/s41586-025-08898-z] [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: 02/07/2024] [Accepted: 03/14/2025] [Indexed: 04/25/2025]
Abstract
Acute myocardial infarction is a leading cause of morbidity and mortality worldwide1. Clinical studies have shown that the severity of cardiac injury after myocardial infarction exhibits a circadian pattern, with larger infarcts and poorer outcomes in patients experiencing morning-onset events2-7. However, the molecular mechanisms underlying these diurnal variations remain unclear. Here we show that the core circadian transcription factor BMAL17-11 regulates circadian-dependent myocardial injury by forming a transcriptionally active heterodimer with a non-canonical partner-hypoxia-inducible factor 2 alpha (HIF2A)12-16-in a diurnal manner. To substantiate this finding, we determined the cryo-EM structure of the BMAL1-HIF2A-DNA complex, revealing structural rearrangements within BMAL1 that enable cross-talk between circadian rhythms and hypoxia signalling. BMAL1 modulates the circadian hypoxic response by enhancing the transcriptional activity of HIF2A and stabilizing the HIF2A protein. We further identified amphiregulin (AREG)16,17 as a rhythmic target of the BMAL1-HIF2A complex, critical for regulating daytime variations of myocardial injury. Pharmacologically targeting the BMAL1-HIF2A-AREG pathway provides cardioprotection, with maximum efficacy when aligned with the pathway's circadian phase. These findings identify a mechanism governing circadian variations of myocardial injury and highlight the therapeutic potential of clock-based pharmacological interventions for treating ischaemic heart disease.
Collapse
Affiliation(s)
- Wei Ruan
- Department of Anesthesiology, Critical Care and Pain Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA.
- Department of Anesthesiology, Second Xiangya Hospital, Central South University, Changsha, China.
| | - Tao Li
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - In Hyuk Bang
- Department of Anesthesiology, Critical Care and Pain Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Jaewoong Lee
- Department of Anesthesiology, Yale University School of Medicine, New Haven, CT, USA
| | - Wankun Deng
- Center for Precision Health, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Xinxin Ma
- Department of Anesthesiology, Critical Care and Pain Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Cong Luo
- Department of Anesthesiology, Critical Care and Pain Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
- Department of Anesthesiology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Fang Du
- Department of Anesthesiology, Critical Care and Pain Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Seung-Hee Yoo
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Boyun Kim
- Department of Anesthesiology, Critical Care and Pain Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
- Major in Aquaculture and Applied Life Sciences, College of Fisheries Science, Pukyong National University, Busan, Republic of Korea
| | - Jiwen Li
- Department of Anesthesiology, Critical Care and Pain Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
- Department of Cardiac Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaoyi Yuan
- Department of Anesthesiology, Critical Care and Pain Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Katherine Figarella
- Department of Anesthesiology, Critical Care and Pain Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Yu A An
- Department of Anesthesiology, Critical Care and Pain Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Yin-Ying Wang
- Center for Precision Health, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Yafen Liang
- Department of Anesthesiology, Critical Care and Pain Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
- Center for Outcomes Research, UTHealth Houston, Houston, TX, USA
| | - Matthew DeBerge
- Department of Anesthesiology, Critical Care and Pain Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Dongze Zhang
- Department of Anesthesiology, Critical Care and Pain Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Zhen Zhou
- Division of Medical Genetics, Department of Internal Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Yanyu Wang
- Department of Anesthesiology, Critical Care and Pain Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Joshua M Gorham
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | | | | | - Sary F Aranki
- Department of Surgery, Division of Cardiac Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ragini Nair
- Department of Anesthesiology, Critical Care and Pain Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Lei Li
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen, China
| | - Jagat Narula
- Division of Cardiology, Department of Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Memorial Hermann Hospital, Houston, TX, USA
| | - Zhongming Zhao
- Center for Precision Health, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Alemayehu A Gorfe
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Jochen D Muehlschlegel
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kuang-Lei Tsai
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA.
- MD Anderson Cancer Center, UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX, USA.
| | - Holger K Eltzschig
- Department of Anesthesiology, Critical Care and Pain Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA.
- Center for Outcomes Research, UTHealth Houston, Houston, TX, USA.
| |
Collapse
|