Publications

2023
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.
Moritz Hunkeler, Cyrus Y. Jin, and Eric S. Fischer. 2/9/2023. “Structures of BIRC6-client complexes provide a mechanism of Smac-mediated release of caspases.” Science, Pp. eade5750. Publisher's VersionAbstract
Tight regulation of apoptosis is essential for metazoan development and prevents diseases such as cancer and neurodegeneration. Caspase activation is central to apoptosis and inhibitor of apoptosis (IAP) proteins are the principal actors that restrain caspase activity and are therefore attractive therapeutic targets. IAPs, in turn, are regulated by mitochondria-derived pro-apoptotic factors such as Smac and HtrA2. Through a series of cryo-electron microscopy (cryo-EM) structures of full-length human baculoviral IAP repeat-containing protein 6 (BIRC6) bound to Smac, caspases, and HtrA2, we provide a molecular understanding for BIRC6-mediated caspase inhibition and its release by Smac. The architecture of BIRC6, together with near-irreversible binding of Smac, elucidates how the IAP inhibitor Smac can effectively control a processive ubiquitin ligase to respond to apoptotic stimuli.
Bhaskar Paidimuddala, Jianhao Cao, Grady Nash, Qing Xie, Hao Wu, and Liman Zhang. 1/5/2023. “Mechanism of NAIP–-NLRC4 inflammasome activation revealed by cryo-EM structure of unliganded NAIP5.” Nature Structural & Molecular Biology, 30, 2, Pp. 159-166. Publisher's VersionAbstract
The nucleotide-binding domain (NBD), leucine rich repeat (LRR) domain containing protein family (NLR family) apoptosis inhibitory proteins (NAIPs) are cytosolic receptors that play critical roles in the host defense against bacterial infection. NAIPs interact with conserved bacterial ligands and activate the NLR family caspase recruitment domain containing protein 4 (NLRC4) to initiate the NAIP–-NLRC4 inflammasome pathway. Here we found the process of NAIP activation is completely different from NLRC4. Our cryo-EM structure of unliganded mouse NAIP5 adopts an unprecedented wide-open conformation, with the nucleating surface fully exposed and accessible to recruit inactive NLRC4. Upon ligand binding, the winged helix domain (WHD) of NAIP5 undergoes roughly 20° rotation to form a steric clash with the inactive NLRC4, which triggers the conformational change of NLRC4 from inactive to active state. We also show the rotation of WHD places the 17–18 loop at a position that directly bind the active NLRC4 and stabilize the NAIP5–NLRC4 complex. Overall, these data provide structural mechanisms of inactive NAIP5, the process of NAIP5 activation and NAIP-dependent NLRC4 activation.
Jiazhi Li, Longfei Wang, Quentin Hahn, Radosław P. Nowak, Thibault Viennet, Esteban A. Orellana, Shourya S. Roy Burman, Hong Yue, Moritz Hunkeler, Pietro Fontana, Hao Wu, Haribabu Arthanari, Eric S. Fischer, and Richard I. Gregory. 1/4/2023. “Structural basis of regulated m7G tRNA modification by METTL1–WDR4.” Nature, 613, 7943, Pp. 391-397. Publisher's VersionAbstract
Chemical modifications of RNA have key roles in many biological processes1–3. N7-methylguanosine (m7G) is required for integrity and stability of a large subset of tRNAs4–7. The methyltransferase 1–WD repeat-containing protein 4 (METTL1–WDR4) complex is the methyltransferase that modifies G46 in the variable loop of certain tRNAs, and its dysregulation drives tumorigenesis in numerous cancer types8–14. Mutations in WDR4 cause human developmental phenotypes including microcephaly15–17. How METTL1–WDR4 modifies tRNA substrates and is regulated remains elusive18. Here we show,  through structural, biochemical and cellular studies of human METTL1–WDR4, that WDR4 serves as a scaffold for METTL1 and the tRNA T-arm. Upon tRNA binding, the $\alpha$C region of METTL1 transforms into a helix, which together with the $\alpha$6 helix secures both ends of the tRNA variable loop. Unexpectedly, we find that the predicted disordered N-terminal region of METTL1 is part of the catalytic pocket and essential for methyltransferase activity. Furthermore, we reveal that S27 phosphorylation in the METTL1 N-terminal region inhibits methyltransferase activity by locally disrupting the catalytic centre. Our results provide a molecular understanding of tRNA substrate recognition and phosphorylation-mediated regulation of METTL1–WDR4, and reveal the presumed disordered N-terminal region of METTL1 as a nexus of methyltransferase activity.
2022
Shimi Meleppattu, Haixia Zhou, Jin Dai, Miao Gui, and Alan Brown. 12/22/2022. “Mechanism of IFT-A polymerization into trains for ciliary transport.” Cell, 185, 26, Pp. 4986-4998.e12. Publisher's Version
Le Xiao, Venkat Giri Magupalli, and Hao Wu. 11/28/2022. “Cryo-EM structures of the active NLRP3 inflammasome disk.” Nature. Publisher's VersionAbstract
Inflammasomes are cytosolic innate immune complexes that activate caspase-1 upon detection of pathogenic and endogenous dangers1-5, and NLRP3 is an inflammasome sensor of membrane damage highly important in inducing inflammation2,6,7. Here we report cryo-EM structures of disk-shaped active NLRP3 oligomers in complex with ATP&\#x1D6FE;S, the centrosomal kinase NEK7, and the adaptor protein ASC which recruits caspase-1. In these NLRP3-NEK7-ASC complexes, the central NACHT domain of NLRP3 assumes an ATP-bound conformation in which two of its subdomains rotate by \textasciitilde85 ° relative to the ADP-bound inactive conformation8-12. The FISNA domain conserved in NLRP3 but absent in most NLRPs13 becomes ordered in its key regions to stabilize the active NACHT conformation and mediate most interactions in the disk. Mutations on these interactions compromise NLRP3-mediated caspase-1 activation. The N-terminal PYDs from all the NLRP3 subunits gather together to form a PYD filament that recruits ASC PYD to elicit downstream signalling. Surprisingly, the C-terminal LRR domain and the LRR-bound NEK7 do not participate in disk interfaces. Together with previous structures of inactive NLRP3 cage in which LRR-LRR interactions play an important role8-11, we propose that the role of NEK7 is to break the inactive cage to transform NLRP3 into the active NLRP3 inflammasome disk.
José A. Velilla, Matthew R. Volpe, Grace E. Kenney, Richard M. Walsh, Emily P. Balskus, and Rachelle Gaudet. 10/17/2022. “Structural basis of colibactin activation by the ClbP peptidase.” Nature Chemical Biology. Publisher's VersionAbstract
Colibactin, a DNA cross-linking agent produced by gut bacteria, is implicated in colorectal cancer. Its biosynthesis uses a prodrug resistance mechanism: a non-toxic precursor assembled in the cytoplasm is activated after export to the periplasm. This activation is mediated by ClbP, an inner-membrane peptidase with an N-terminal periplasmic catalytic domain and a C-terminal three-helix transmembrane domain. Although the transmembrane domain is required for colibactin activation, its role in catalysis is unclear. Our structure of full-length ClbP bound to a product analog reveals an interdomain interface important for substrate binding and enzyme stability and interactions that explain the selectivity of ClbP for the N-acyl-d-asparagine prodrug motif. Based on structural and biochemical evidence, we propose that ClbP dimerizes to form an extended substrate-binding site that can accommodate a pseudodimeric precolibactin with its two terminal prodrug motifs in the two ClbP active sites, thus enabling the coordinated activation of both electrophilic warheads.
Ying Dong, Xiong Pi, Frauke Bartels-Burgahn, Deniz Saltukoglu, Zhuoyi Liang, Jianying Yang, Frederick W. Alt, Michael Reth, and Hao Wu. 10/14/2022. “Structural principles of B-cell antigen receptor assembly.” Nature. Publisher's VersionAbstract
The B-cell antigen receptor (BCR) is composed of a membrane-bound immunoglobulin (mIg) of class M, D, G, E or A for antigen recognition1–3 and a disulfide-linked Ig$\alpha$ and Ig$\beta$ heterodimer (Ig$\alpha$/$\beta$) that functions as the signalling entity through their intracellular immunoreceptor tyrosine-based activation motifs (ITAMs)4,5. The organizing principle of the BCR remains elusive. Here we report cryogenic electron microscopy structures of mouse full-length IgM BCR at 8.2 \AA resolution and its Fab-deleted form at 3.3 \AA resolution. At the ectodomain (ECD), the Ig$\alpha$/$\beta$ heterodimer mainly uses Ig$\alpha$ to associate with Cµ3-Cµ4 domains of one heavy chain (µHC) while leaving the other heavy chain (µHC') empty. The transmembrane domain (TMD) helices of the two µHCs interact with those of the Ig$\alpha$/$\beta$ heterodimer to form a tight 4-helix bundle. The asymmetry at the TMD prevents the recruitment of two Ig$\alpha$/$\beta$ heterodimers. Surprisingly, the connecting peptide between the ECD and TMD of µHC intervenes in between those of Ig$\alpha$ and Ig$\beta$ to guide the TMD assembly through striking charge complementarity. Weaker but distinct density for the Ig$\beta$ ITAMs nestles next to the TMD, suggesting potential autoinhibition of ITAM phosphorylation. Interfacial analyses suggest that all BCR classes utilize a general organizational architecture. Our studies provide a structural platform for understanding B-cell signalling and designing rational therapies against BCR-mediated diseases.
Miao Gui, Jacob T. Croft, Davide Zabeo, Vajradhar Acharya, Justin M. Kollman, Thomas Burgoyne, Johanna L. Höög, and Alan Brown. 10/3/2022. “SPACA9 is a lumenal protein of human ciliary singlet and doublet microtubules.” Proceedings of the National Academy of Sciences, 119, 41, Pp. e2207605119. Publisher's VersionAbstract
The cilium-centrosome complex contains triplet, doublet, and singlet microtubules. The lumenal surfaces of each microtubule within this diverse array are decorated by microtubule inner proteins (MIPs). Here, we used single-particle cryo-electron microscopy methods to build atomic models of two types of human ciliary microtubule: the doublet microtubules of multiciliated respiratory cells and the distal singlet microtubules of monoflagellated human spermatozoa. We discover that SPACA9 is a polyspecific MIP capable of binding both microtubule types. SPACA9 forms intralumenal striations in the B tubule of respiratory doublet microtubules and noncontinuous spirals in sperm singlet microtubules. By acquiring new and reanalyzing previous cryo-electron tomography data, we show that SPACA9-like intralumenal striations are common features of different microtubule types in animal cilia. Our structures provide detailed references to help rationalize ciliopathy-causing mutations and position cryo-EM as a tool for the analysis of samples obtained directly from ciliopathy patients.

Pages