Publications by Year: 2023

2023
Calina Glynn, Joshua E. Chun, Cameron C. Donahue, Monica J. S. Nadler, Zhanyun Fan, and Bradley T. Hyman. 12/28/2023. “Reconstitution of the Alzheimer’s Disease Tau Core Structure from Recombinant Tau297–391 Yields Variable Quaternary Structures as Seen by Negative Stain and Cryo-EM.” Biochemistry, Pp. null. Publisher's Version
Joel Finney, Annie Park Moseman, Susan Kong, Akiko Watanabe, Shengli Song, Richard M. Walsh, Masayuki Kuraoka, Ryutaro Kotaki, E. Ashley Moseman, Kevin R. McCarthy, Dongmei Liao, Xiaoe Liang, Xiaoyan Nie, Olivia Lavidor, Richard Abbott, Stephen C. Harrison, and Garnett Kelsoe. 12/26/2023. “Protective human antibodies against a conserved epitope in pre- and postfusion influenza hemagglutinin.” Proceedings of the National Academy of Sciences, 121, 1, Pp. e2316964120. Publisher's VersionAbstract
Phylogenetically and antigenically distinct influenza A and B viruses (IAV and IBV) circulate in human populations, causing widespread morbidity. Antibodies (Abs) that bind epitopes conserved in both IAV and IBV hemagglutinins (HAs) could protect against disease by diverse virus subtypes. Only one reported HA Ab, isolated from a combinatorial display library, protects against both IAV and IBV. Thus, there has been so far no information on the likelihood of finding naturally occurring human Abs that bind HAs of diverse IAV subtypes and IBV lineages. We have now recovered from several unrelated human donors five clonal Abs that bind a conserved epitope preferentially exposed in the postfusion conformation of IAV and IVB HA2. These Abs lack neutralizing activity in vitro but in mice provide strong, IgG subtype–dependent protection against lethal IAV and IBV infections. Strategies to elicit similar Abs routinely might contribute to more effective influenza vaccines.
Jonathan W. Markert, Seychelle M. Vos, and Lucas Farnung. 12/8/2023. “Structure of the complete Saccharomyces cerevisiae Rpd3S-nucleosome complex.” Nature Communications, 14, 1, Pp. 8128. Publisher's VersionAbstract
Acetylation of histones is a key post-translational modification that guides gene expression regulation. In yeast, the class I histone deacetylase containing Rpd3S complex plays a critical role in the suppression of spurious transcription by removing histone acetylation from actively transcribed genes. The S. cerevisiae Rpd3S complex has five subunits (Rpd3, Sin3, Rco1, Eaf3, and Ume1) but its subunit stoichiometry and how the complex engages nucleosomes to achieve substrate specificity remains elusive. Here we report the cryo-EM structure of the complete Rpd3S complex bound to a nucleosome. Sin3 and two copies of subunits Rco1 and Eaf3 encircle the deacetylase subunit Rpd3 and coordinate the positioning of Ume1. The Rpd3S complex binds both trimethylated H3 tails at position lysine 36 and makes multiple additional contacts with the nucleosomal DNA and the H2A–H2B acidic patch. Direct regulation via the Sin3 subunit coordinates binding of the acetylated histone substrate to achieve substrate specificity.
Wenxiang Zhang, Fangwei Leng, Xi Wang, Ricardo N. Ramirez, Jinseok Park, Christophe Benoist, and Sun Hur. 11/29/2023. “FOXP3 recognizes microsatellites and bridges DNA through multimerization.” Nature. Publisher's VersionAbstract
FOXP3 is a transcription factor that is essential for the development of regulatory T cells, a branch of T cells that suppress excessive inflammation and autoimmunity1–5. However, the molecular mechanisms of FOXP3 remain unclear. Here we here show that FOXP3 uses the forkhead domain–-a DNA-binding domain that is commonly thought to function as a monomer or dimer–-to form a higher-order multimer after binding to TnG repeat microsatellites. The cryo-electron microscopy structure of FOXP3 in a complex with T3G repeats reveals a ladder-like architecture, whereby two double-stranded DNA molecules form the two `side rails' bridged by five pairs of FOXP3 molecules, with each pair forming a `rung'. Each FOXP3 subunit occupies TGTTTGT within the repeats in a manner that is indistinguishable from that of FOXP3 bound to the forkhead consensus motif (TGTTTAC). Mutations in the intra-rung interface impair TnG repeat recognition, DNA bridging and the cellular functions of FOXP3, all without affecting binding to the forkhead consensus motif. FOXP3 can tolerate variable inter-rung spacings, explaining its broad specificity for TnG-repeat-like sequences in vivo and in vitro. Both FOXP3 orthologues and paralogues show similar TnG repeat recognition and DNA bridging. These findings therefore reveal a mode of DNA recognition that involves transcription factor homomultimerization and DNA bridging, and further implicates microsatellites in transcriptional regulation and diseases.
Sadie P. Antine, Alex G. Johnson, Sarah E. Mooney, Azita Leavitt, Megan L. Mayer, Erez Yirmiya, Gil Amitai, Rotem Sorek, and Philip J. Kranzusch. 11/22/2023. “Structural basis of Gabija anti-phage defence and viral immune evasion.” Nature. Publisher's VersionAbstract
Bacteria encode hundreds of diverse defense systems that protect from viral infection and inhibit phage propagation1–5. Gabija is one of the most prevalent anti-phage defense systems, occurring in >15% of all sequenced bacterial and archaeal genomes1,6,7, but the molecular basis of how Gabija defends cells from viral infection remains poorly understood. Here we use X-ray crystallography and cryo-EM to define how Gabija proteins assemble into an \textasciitilde500 kDa supramolecular complex that degrades phage DNA. Gabija protein A (GajA) is a DNA endonuclease that tetramerizes to form the core of the anti-phage defense complex. Two sets of Gabija protein B (GajB) dimers dock at opposite sides of the complex and create a 4:4 GajAB assembly that is essential for phage resistance in vivo. We show that a phage-encoded protein Gabija anti-defense 1 (Gad1) directly binds the Gabija GajAB complex and inactivates defense. A cryo-EM structure of the virally inhibited state reveals that Gad1 forms an octameric web that encases the GajAB complex and inhibits DNA recognition and cleavage. Our results reveal the structural basis of assembly of the Gabija anti-phage defense complex and define a unique mechanism of viral immune evasion.
Pascal Devant, Ying Dong, Julian Mintseris, Weiyi Ma, Steven P Gygi, Hao Wu, and Jonathan C Kagan. 11/22/2023. “Structural insights into cytokine cleavage by inflammatory caspase-4.” Nature, 624, 7991, Pp. 451–459.Abstract
Inflammatory caspases are key enzymes in mammalian innate immunity that control the processing and release of interleukin-1 (IL-1)-family cytokines1,2. Despite the biological importance, the structural basis for inflammatory caspase-mediated cytokine processing has remained unclear. To date, catalytic cleavage of IL-1-family members, including pro-IL-1$\beta$ and pro-IL-18, has been attributed primarily to caspase-1 activities within canonical inflammasomes3. Here we demonstrate that the lipopolysaccharide receptor caspase-4 from humans and other mammalian species (except rodents) can cleave pro-IL-18 with an efficiency similar to pro-IL-1$\beta$ and pro-IL-18 cleavage by the prototypical IL-1-converting enzyme caspase-1. This ability of caspase-4 to cleave pro-IL-18, combined with its previously defined ability to cleave and activate the lytic pore-forming protein gasdermin D (GSDMD)4,5, enables human cells to bypass the need for canonical inflammasomes and caspase-1 for IL-18 release. The structure of the caspase-4–pro-IL-18 complex determined using cryogenic electron microscopy reveals that pro-lL-18 interacts with caspase-4 through two distinct interfaces: a protease exosite and an interface at the caspase-4 active site involving residues in the pro-domain of pro-IL-18, including the tetrapeptide caspase-recognition sequence6. The mechanisms revealed for cytokine substrate capture and cleavage differ from those observed for the caspase substrate GSDMD7,8. These findings provide a structural framework for the discussion of caspase activities in health and disease.
Carol Cho, Christian Ganser, Takayuki Uchihashi, Koichi Kato, and Ji-Joon Song. 9/28/2023. “Structure of the human ATAD2 AAA+ histone chaperone reveals mechanism of regulation and inter-subunit communication.” Communications Biology, 6, 1, Pp. 993.Abstract
ATAD2 is a non-canonical ATP-dependent histone chaperone and a major cancer target. Despite widespread efforts to design drugs targeting the ATAD2 bromodomain, little is known about the overall structural organization and regulation of ATAD2. Here, we present the 3.1 \AA cryo-EM structure of human ATAD2 in the ATP state, showing a shallow hexameric spiral that binds a peptide substrate at the central pore. The spiral conformation is locked by an N-terminal linker domain (LD) that wedges between the seam subunits, thus limiting ATP-dependent symmetry breaking of the AAA+ ring. In contrast, structures of the ATAD2-histone H3/H4 complex show the LD undocked from the seam, suggesting that H3/H4 binding unlocks the AAA+ spiral by allosterically releasing the LD. These findings, together with the discovery of an inter-subunit signaling mechanism, reveal a unique regulatory mechanism for ATAD2 and lay the foundation for developing new ATAD2 inhibitors.
Richard M. Walsh, Shaun Rawson, Helena M. Schnell, Benjamin Velez, Tamayanthi Rajakumar, and John Hanna. 8/31/2023. “Structure of the preholoproteasome reveals late steps in proteasome core particle biogenesis.” Nature Structural & Molecular Biology. Publisher's VersionAbstract
Assembly of the proteasome's core particle (CP), a barrel-shaped chamber of four stacked rings, requires five chaperones and five subunit propeptides. Fusion of two half-CP precursors yields a complete structure but remains immature until active site maturation. Here, using Saccharomyces cerevisiae, we report a high-resolution cryogenic electron microscopy structure of preholoproteasome, a post-fusion assembly intermediate. Our data reveal how CP midline-spanning interactions induce local changes in structure, facilitating maturation. Unexpectedly, we find that cleavage may not be sufficient for propeptide release, as residual interactions with chaperones such as Ump1 hold them in place. We evaluated previous models proposing that dynamic conformational changes in chaperones drive CP fusion and autocatalytic activation by comparing preholoproteasome to pre-fusion intermediates. Instead, the data suggest a scaffolding role for the chaperones Ump1 and Pba1/Pba2. Our data clarify key aspects of CP assembly, suggest that undiscovered mechanisms exist to explain CP fusion/activation, and have relevance for diseases of defective CP biogenesis.
Wang Zheng, Shaun Rawson, Zhangfei Shen, Elakkiya Tamilselvan, Harper E. Smith, Julia Halford, Chen Shen, Swetha E. Murthy, Maximilian H. Ulbrich, Marcos Sotomayor, Tian-Min Fu, and Jeffrey R. Holt. 8/4/2023. “TMEM63 proteins function as monomeric high-threshold mechanosensitive ion channels.” Neuron. Publisher's VersionAbstract
OSCA/TMEM63s form mechanically activated (MA) ion channels in plants and animals, respectively. OSCAs and related TMEM16s and transmembrane channel-like (TMC) proteins form homodimers with two pores. Here, we uncover an unanticipated monomeric configuration of TMEM63 proteins. Structures of TMEM63A and TMEM63B (referred to as TMEM63s) revealed a single highly restricted pore. Functional analyses demonstrated that TMEM63s are bona fide mechanosensitive ion channels, characterized by small conductance and high thresholds. TMEM63s possess evolutionary variations in the intracellular linker IL2, which mediates dimerization in OSCAs. Replacement of OSCA1.2 IL2 with TMEM63A IL2 or mutations to key variable residues resulted in monomeric OSCA1.2 and MA currents with significantly higher thresholds. Structural analyses revealed substantial conformational differences in the mechano-sensing domain IL2 and gating helix TM6 between TMEM63s and OSCA1.2. Our studies reveal that mechanosensitivity in OSCA/TMEM63 channels is affected by oligomerization and suggest gating mechanisms that may be shared by OSCA/TMEM63, TMEM16, and TMC channels.
Eun Young Park, Shaun Rawson, Anna Schmoker, Byeong-Won Kim, Sehee Oh, KangKang Song, Hyesung Jeon, and Michael J. Eck. 7/29/2023. “Cryo-EM structure of a RAS/RAF recruitment complex.” Nature Communications, 14, 1, Pp. 4580. Publisher's VersionAbstract
RAF-family kinases are activated by recruitment to the plasma membrane by GTP-bound RAS, whereupon they initiate signaling through the MAP kinase cascade. Prior structural studies of KRAS with RAF have focused on the isolated RAS-binding and cysteine-rich domains of RAF (RBD and CRD, respectively), which interact directly with RAS. Here we describe cryo-EM structures of a KRAS bound to intact BRAF in an autoinhibited state with MEK1 and a 14-3-3 dimer. Analysis of this KRAS/BRAF/MEK1/14-3-3 complex reveals KRAS bound to the RAS-binding domain of BRAF, captured in two orientations. Core autoinhibitory interactions in the complex are unperturbed by binding of KRAS and in vitro activation studies confirm that KRAS binding is insufficient to activate BRAF, absent membrane recruitment. These structures illustrate the separability of binding and activation of BRAF by RAS and suggest stabilization of this pre-activation intermediate as an alternative therapeutic strategy to blocking binding of KRAS.
Colin H. Lipper, Emily D. Egan, Khal-Hentz Gabriel, and Stephen C. Blacklow. 7/28/2023. “Structural basis for membrane-proximal proteolysis of substrates by ADAM10.” Cell. Publisher's VersionAbstract
The endopeptidase ADAM10 is a critical catalyst for the regulated proteolysis of key drivers of mammalian development, physiology, and non-amyloidogenic cleavage of APP as the primary $\alpha$-secretase. ADAM10 function requires the formation of a complex with a C8-tetraspanin protein, but how tetraspanin binding enables positioning of the enzyme active site for membrane-proximal cleavage remains unknown. We present here a cryo-EM structure of a vFab-ADAM10-Tspan15 complex, which shows that Tspan15 binding relieves ADAM10 autoinhibition and acts as a molecular measuring stick to position the enzyme active site about 20 \AA from the plasma membrane for membrane-proximal substrate cleavage. Cell-based assays of N-cadherin shedding establish that the positioning of the active site by the interface between the ADAM10 catalytic domain and the bound tetraspanin influences selection of the preferred cleavage site. Together, these studies reveal the molecular mechanism underlying ADAM10 proteolysis at membrane-proximal sites and offer a roadmap for its modulation in disease.
Jun Zhang, Weichun Tang, Hailong Gao, Christy L. Lavine, Wei Shi, Hanqin Peng, Haisun Zhu, Krishna Anand, Matina Kosikova, Hyung Joon Kwon, Pei Tong, Avneesh Gautam, Sophia Rits-Volloch, Shaowei Wang, Megan L. Mayer, Duane R. Wesemann, Michael S. Seaman, Jianming Lu, Tianshu Xiao, Hang Xie, and Bing Chen. 7/10/2023. “Structural and functional characteristics of the SARS-CoV-2 Omicron subvariant BA.2 spike protein.” Nature Structural & Molecular Biology, 30, 7, Pp. 980-990. Publisher's VersionAbstract
The Omicron subvariant BA.2 has become the dominant circulating strain of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in many countries. Here, we have characterized structural, functional and antigenic properties of the full-length BA.2 spike (S) protein and compared replication of the authentic virus in cell culture and an animal model with previously prevalent variants. BA.2þinspace}S can fuse membranes slightly more efficiently than Omicron BA.1, but still less efficiently than other previous variants. Both BA.1 and BA.2 viruses replicated substantially faster in animal lungs than the early G614 (B.1) strain in the absence of pre-existing immunity, possibly explaining the increased transmissibility despite their functionally compromised spikes. As in BA.1, mutations in the BA.2þinspace}S remodel its antigenic surfaces, leading to strong resistance to neutralizing antibodies. These results suggest that both immune evasion and replicative advantage may contribute to the heightened transmissibility of the Omicron subvariants.
Irina Shlosman, Elayne M. Fivenson, Morgan S.A. Gilman, Tyler A. Sisley, Suzanne Walker, Thomas G. Bernhardt, Andrew C. Kruse, and Joseph J. Loparo. 6/10/2023. “Allosteric activation of cell wall synthesis during bacterial growth.” Nature Communications, 14, 1, Pp. 3439. Publisher's VersionAbstract
The peptidoglycan (PG) cell wall protects bacteria against osmotic lysis and determines cell shape, making this structure a key antibiotic target. Peptidoglycan is a polymer of glycan chains connected by peptide crosslinks, and its synthesis requires precise spatiotemporal coordination between glycan polymerization and crosslinking. However, the molecular mechanism by which these reactions are initiated and coupled is unclear. Here we use single-molecule FRET and cryo-EM to show that an essential PG synthase (RodA-PBP2) responsible for bacterial elongation undergoes dynamic exchange between closed and open states. Structural opening couples the activation of polymerization and crosslinking and is essential in vivo. Given the high conservation of this family of synthases, the opening motion that we uncovered likely represents a conserved regulatory mechanism that controls the activation of PG synthesis during other cellular processes, including cell division.
Wei Shi, Yongfei Cai, Haisun Zhu, Hanqin Peng, Jewel Voyer, Sophia Rits-Volloch, Hong Cao, Megan L. Mayer, KangKang Song, Chen Xu, Jianming Lu, Jun Zhang, and Bing Chen. 6/7/2023. “Cryo-EM structure of SARS-CoV-2 postfusion spike in membrane.” Nature, 619, 7969, Pp. 403-409. Publisher's VersionAbstract
The entry of SARS-CoV-2 into host cells depends on the refolding of the virus-encoded spike protein from a prefusion conformation, which is metastable after cleavage, to a lower-energy stable postfusion conformation1,2. This transition overcomes kinetic barriers for fusion of viral and target cell membranes3,4. Here we report a cryogenic electron microscopy (cryo-EM) structure of the intact postfusion spike in a lipid bilayer that represents the single-membrane product of the fusion reaction. The structure provides structural definition of the functionally critical membrane-interacting segments, including the fusion peptide and transmembrane anchor. The internal fusion peptide forms a hairpin-like wedge that spans almost the entire lipid bilayer and the transmembrane segment wraps around the fusion peptide at the last stage of membrane fusion. These results advance our understanding of the spike protein in a membrane environment and may guide development of intervention strategies.
Travis Walton, Miao Gui, Simona Velkova, Mahmoud R. Fassad, Robert A. Hirst, Eric Haarman, Christopher O'Callaghan, Mathieu Bottier, Thomas Burgoyne, Hannah M. Mitchison, and Alan Brown. 5/31/2023. “Axonemal structures reveal mechanoregulatory and disease mechanisms.” Nature, 618, 7965, Pp. 625-633. Publisher's VersionAbstract
Motile cilia and flagella beat rhythmically on the surface of cells to power the flow of fluid and to enable spermatozoa and unicellular eukaryotes to swim. In humans, defective ciliary motility can lead to male infertility and a congenital disorder called primary ciliary dyskinesia (PCD), in which impaired clearance of mucus by the cilia causes chronic respiratory infections1. Ciliary movement is generated by the axoneme, a molecular machine consisting of microtubules, ATP-powered dynein motors and regulatory complexes2. The size and complexity of the axoneme has so far prevented the development of an atomic model, hindering efforts to understand how it functions. Here we capitalize on recent developments in artificial intelligence-enabled structure prediction and cryo-electron microscopy (cryo-EM) to determine the structure of the 96-nm modular repeats of axonemes from the flagella of the alga Chlamydomonas reinhardtii and human respiratory cilia. Our atomic models provide insights into the conservation and specialization of axonemes, the interconnectivity between dyneins and their regulators, and the mechanisms that maintain axonemal periodicity. Correlated conformational changes in mechanoregulatory complexes with their associated axonemal dynein motors provide a mechanism for the long-hypothesized mechanotransduction pathway to regulate ciliary motility. Structures of respiratory-cilia doublet microtubules from four individuals with PCD reveal how the loss of individual docking factors can selectively eradicate periodically repeating structures.
Daniel T. D. Jones, Andrew N. Dates, Shaun D. Rawson, Maggie M. Burruss, Colin H. Lipper, and Stephen C. Blacklow. 4/29/2023. “Tethered agonist activated ADGRF1 structure and signalling analysis reveal basis for G protein coupling.” Nature Communications, 14, 1, Pp. 2490. Publisher's VersionAbstract
Adhesion G Protein Coupled Receptors (aGPCRs) have evolved an activation mechanism to translate extracellular force into liberation of a tethered agonist (TA) to effect cell signalling. We report here that ADGRF1 can signal through all major G protein classes and identify the structural basis for a previously reported G$\alpha$q preference by cryo-EM. Our structure shows that G$\alpha$q preference in ADGRF1 may derive from tighter packing at the conserved F569 of the TA, altering contacts between TM helix I and VII, with a concurrent rearrangement of TM helix VII and helix VIII at the site of G$\alpha$ recruitment. Mutational studies of the interface and of contact residues within the 7TM domain identify residues critical for signalling, and suggest that G$\alpha$s signalling is more sensitive to mutation of TA or binding site residues than G$\alpha$q. Our work advances the detailed molecular understanding of aGPCR TA activation, identifying features that potentially explain preferential signal modulation.
Sarah C Erlandson, Shaun Rawson, James Osei-Owusu, Kelly P Brock, Xinyue Liu, Joao A Paulo, Julian Mintseris, Steven P Gygi, Debora S Marks, Xiaojing Cong, and Andrew C Kruse. 4/20/2023. “The relaxin receptor RXFP1 signals through a mechanism of autoinhibition.” Nature Chemical Biology.Abstract
The relaxin family peptide receptor 1 (RXFP1) is the receptor for relaxin-2, an important regulator of reproductive and cardiovascular physiology. RXFP1 is a multi-domain G protein-coupled receptor (GPCR) with an ectodomain consisting of a low-density lipoprotein receptor class A (LDLa) module and leucine-rich repeats. The mechanism of RXFP1 signal transduction is clearly distinct from that of other GPCRs, but remains very poorly understood. In the present study, we determine the cryo-electron microscopy structure of active-state human RXFP1, bound to a single-chain version of the endogenous agonist relaxin-2 and the heterotrimeric Gs protein. Evolutionary coupling analysis and structure-guided functional experiments reveal that RXFP1 signals through a mechanism of autoinhibition. Our results explain how an unusual GPCR family functions, providing a path to rational drug development targeting the relaxin receptors.
Ashlee M. Plummer-Medeiros, Alan T. Culbertson, Claudio L. Morales-Perez, and Maofu Liao. 4/15/2023. “Activity and Structural Dynamics of Human ABCA1 in a Lipid Membrane.” Journal of Molecular Biology, 435, 8, Pp. 168038. Publisher's VersionAbstract
The human ATP-binding cassette (ABC) transporter ABCA1 plays a critical role in lipid homeostasis as it extracts sterols and phospholipids from the plasma membrane for excretion to the extracellular apolipoprotein A-I and subsequent formation of high-density lipoprotein (HDL) particles. Deleterious mutations of ABCA1 lead to sterol accumulation and are associated with atherosclerosis, poor cardiovascular outcomes, cancer, and Alzheimer’s disease. The mechanism by which ABCA1 drives lipid movement is poorly understood, and a unified platform to produce active ABCA1 protein for both functional and structural studies has been missing. In this work, we established a stable expression system for both a human cell-based sterol export assay and protein purification for in vitro biochemical and structural studies. ABCA1 produced in this system was active in sterol export and displayed enhanced ATPase activity after reconstitution into a lipid bilayer. Our single-particle cryo-EM study of ABCA1 in nanodiscs showed protein induced membrane curvature, revealed multiple distinct conformations, and generated a structure of nanodisc-embedded ABCA1 at 4.0-Å resolution representing a previously unknown conformation. Comparison of different ABCA1 structures and molecular dynamics simulations demonstrates both concerted domain movements and conformational variations within each domain. Taken together, our platform for producing and characterizing ABCA1 in a lipid membrane enabled us to gain important mechanistic and structural insights and paves the way for investigating modulators that target the functions of ABCA1.
Zhipeng A. Wang, Jonathan W. Markert, Samuel D. Whedon, Maheeshi Yapa Abeywardana, Kwangwoon Lee, Hanjie Jiang, Carolay Suarez, Hening Lin, Lucas Farnung, and Philip A. Cole. 3/17/2023. “Structural Basis of Sirtuin 6-Catalyzed Nucleosome Deacetylation.” Journal of the American Chemical Society, Pp. null. Publisher's Version
Brianna Duncan-Lowey, Nitzan Tal, Alex G. Johnson, Shaun Rawson, Megan L. Mayer, Shany Doron, Adi Millman, Sarah Melamed, Taya Fedorenko, Assaf Kacen, Alexander Brandis, Tevie Mehlman, Gil Amitai, Rotem Sorek, and Philip J. Kranzusch. 2/9/2023. “Cryo-EM structure of the RADAR supramolecular anti-phage defense complex.” Cell. Publisher's VersionAbstract
Summary RADAR is a two-protein bacterial defense system that was reported to defend against phage by “editing” messenger RNA. Here, we determine cryo-EM structures of the RADAR defense complex, revealing RdrA as a heptameric, two-layered AAA+ ATPase and RdrB as a dodecameric, hollow complex with twelve surface-exposed deaminase active sites. RdrA and RdrB join to form a giant assembly up to 10 MDa, with RdrA docked as a funnel over the RdrB active site. Surprisingly, our structures reveal an RdrB active site that targets mononucleotides. We show that RdrB catalyzes ATP-to-ITP conversion in vitro and induces the massive accumulation of inosine mononucleotides during phage infection in vivo, limiting phage replication. Our results define ATP mononucleotide deamination as a determinant of RADAR immunity and reveal supramolecular assembly of a nucleotide-modifying machine as a mechanism of anti-phage defense.

Pages