401
|
Song W, Wang J, Han Z, Zhang Y, Zhang H, Wang W, Chang J, Xia B, Fan S, Zhang D, Wang J, Wang HW, Chai J. Structural basis for specific recognition of single-stranded RNA by Toll-like receptor 13. Nat Struct Mol Biol 2015; 22:782-7. [PMID: 26323037 DOI: 10.1038/nsmb.3080] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 08/06/2015] [Indexed: 12/16/2022]
Abstract
Toll-like receptors (TLRs) have crucial roles in innate immunity, functioning as pattern-recognition receptors. TLR13 recognizes a conserved sequence from bacterial 23S rRNA and then triggers an immune response. Here we report the crystal structure of the mouse TLR13 ectodomain bound by a 13-nt single-stranded (ss) RNA derived from 23S rRNA. The ssRNA induces TLR13 dimerization but assumes a stem-loop-like structure that is completely different from that in the bacterial ribosome but nevertheless is crucial for TLR13 recognition. Most of the RNA nucleotides are splayed out to make base-specific contacts with the concave surface of TLR13, and RNA-specific interactions are important to allow TLR13 to distinguish RNA from DNA. Interestingly, a viral-derived 16-nt ssRNA predicted to form a similar stem-loop-like structure also induces TLR13 activation. Together, our results reveal the structural mechanism of TLR13's sequence- and conformation-specific recognition of ssRNA.
Collapse
Affiliation(s)
- Wen Song
- Ministry of Education Key Laboratory of Protein Science, Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing, China.,Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Jia Wang
- Ministry of Education Key Laboratory of Protein Science, Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing, China.,Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Zhifu Han
- Ministry of Education Key Laboratory of Protein Science, Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing, China.,Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Yifan Zhang
- Center for Infectious and Inflammatory Diseases, Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, Texas, USA
| | - Heqiao Zhang
- Ministry of Education Key Laboratory of Protein Science, Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing, China
| | - Weiguang Wang
- Ministry of Education Key Laboratory of Protein Science, Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing, China
| | - Junbiao Chang
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou, China
| | - Bingshu Xia
- Center for Infectious and Inflammatory Diseases, Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, Texas, USA
| | - Shilong Fan
- Ministry of Education Key Laboratory of Protein Science, Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing, China.,Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Dekai Zhang
- Center for Infectious and Inflammatory Diseases, Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, Texas, USA
| | - Jiawei Wang
- Ministry of Education Key Laboratory of Protein Science, Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing, China
| | - Hong-Wei Wang
- Ministry of Education Key Laboratory of Protein Science, Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing, China.,Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Jijie Chai
- Ministry of Education Key Laboratory of Protein Science, Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing, China.,Tsinghua-Peking Center for Life Sciences, Beijing, China
| |
Collapse
|
402
|
Ichikawa M, Saito K, Yanagisawa HA, Yagi T, Kamiya R, Yamaguchi S, Yajima J, Kushida Y, Nakano K, Numata O, Toyoshima YY. Axonemal dynein light chain-1 locates at the microtubule-binding domain of the γ heavy chain. Mol Biol Cell 2015; 26:4236-47. [PMID: 26399296 PMCID: PMC4642857 DOI: 10.1091/mbc.e15-05-0289] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 09/16/2015] [Indexed: 11/23/2022] Open
Abstract
Dynein light chain 1 (LC1) of the outer arm dynein (OAD) complex associates with the microtubule-binding domain (MTBD) of γ heavy chain inside the complex. LC1 is considered to regulate the OAD activity and ciliary/flagellar motion by modulating γ MTBD's affinity to the B-tubule of the doublet microtubule in the axoneme. The outer arm dynein (OAD) complex is the main propulsive force generator for ciliary/flagellar beating. In Chlamydomonas and Tetrahymena, the OAD complex comprises three heavy chains (α, β, and γ HCs) and >10 smaller subunits. Dynein light chain-1 (LC1) is an essential component of OAD. It is known to associate with the Chlamydomonas γ head domain, but its precise localization within the γ head and regulatory mechanism of the OAD complex remain unclear. Here Ni-NTA-nanogold labeling electron microscopy localized LC1 to the stalk tip of the γ head. Single-particle analysis detected an additional structure, most likely corresponding to LC1, near the microtubule-binding domain (MTBD), located at the stalk tip. Pull-down assays confirmed that LC1 bound specifically to the γ MTBD region. Together with observations that LC1 decreased the affinity of the γ MTBD for microtubules, we present a new model in which LC1 regulates OAD activity by modulating γ MTBD's affinity for the doublet microtubule.
Collapse
Affiliation(s)
- Muneyoshi Ichikawa
- Department of Life Sciences, Graduate School of Arts and Sciences, University of Tokyo, Tokyo 153-8902, Japan
| | - Kei Saito
- Department of Life Sciences, Graduate School of Arts and Sciences, University of Tokyo, Tokyo 153-8902, Japan
| | - Haru-Aki Yanagisawa
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Tokyo 113-0033, Japan
| | - Toshiki Yagi
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Tokyo 113-0033, Japan
| | - Ritsu Kamiya
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Tokyo 113-0033, Japan
| | - Shin Yamaguchi
- Department of Life Sciences, Graduate School of Arts and Sciences, University of Tokyo, Tokyo 153-8902, Japan
| | - Junichiro Yajima
- Department of Life Sciences, Graduate School of Arts and Sciences, University of Tokyo, Tokyo 153-8902, Japan
| | - Yasuharu Kushida
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Ibaraki 305-8572, Japan
| | - Kentaro Nakano
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Ibaraki 305-8572, Japan
| | - Osamu Numata
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Ibaraki 305-8572, Japan
| | - Yoko Y Toyoshima
- Department of Life Sciences, Graduate School of Arts and Sciences, University of Tokyo, Tokyo 153-8902, Japan
| |
Collapse
|
403
|
A Molecular Toolkit to Visualize Native Protein Assemblies in the Context of Human Disease. Sci Rep 2015; 5:14440. [PMID: 26395823 PMCID: PMC4585766 DOI: 10.1038/srep14440] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 08/27/2015] [Indexed: 12/18/2022] Open
Abstract
We present a new molecular toolkit to investigate protein assemblies natively formed in the context of human disease. The system employs tunable microchips that can be decorated with switchable adaptor molecules to select for target proteins of interest and analyze them using molecular microscopy. Implementing our new streamlined microchip approach, we could directly visualize BRCA1 gene regulatory complexes from patient-derived cancer cells for the first time.
Collapse
|
404
|
Dai A, Ye F, Taylor DW, Hu G, Ginsberg MH, Taylor KA. The Structure of a Full-length Membrane-embedded Integrin Bound to a Physiological Ligand. J Biol Chem 2015; 290:27168-27175. [PMID: 26391523 DOI: 10.1074/jbc.m115.682377] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Indexed: 11/06/2022] Open
Abstract
Increased ligand binding to integrin ("activation") underpins many biological processes, such as leukocyte trafficking, cell migration, host-pathogen interaction, and hemostasis. Integrins exist in several conformations, ranging from compact and bent to extended and open. However, the exact conformation of membrane-embedded, full-length integrin bound to its physiological macromolecular ligand is still unclear. Integrin αIIbβ3, the most abundant integrin in platelets, has been a prototype for integrin activation studies. Using negative stain electron microscopy and nanodisc-embedding to provide a membrane-like environment, we visualized the conformation of full-length αIIbβ3 in both a Mn(2+)-activated, ligand-free state and a Mn(2+)-activated, fibrin-bound state. Activated but ligand-free integrins exist mainly in the compact conformation, whereas fibrin-bound αIIbβ3 predominantly exists in a fully extended, headpiece open conformation. Our results show that membrane-embedded, full-length integrin adopts an extended and open conformation when bound to its physiological macromolecular ligand.
Collapse
Affiliation(s)
- Aguang Dai
- Institute of Molecular Biophysics, Florida State University, Tallahassee, Florida 32306-4380 and
| | - Feng Ye
- Department of Hematology and Oncology, University of California at San Diego, La Jolla, California 92093-0726
| | - Dianne W Taylor
- Institute of Molecular Biophysics, Florida State University, Tallahassee, Florida 32306-4380 and
| | - Guiqing Hu
- Institute of Molecular Biophysics, Florida State University, Tallahassee, Florida 32306-4380 and
| | - Mark H Ginsberg
- Department of Hematology and Oncology, University of California at San Diego, La Jolla, California 92093-0726
| | - Kenneth A Taylor
- Institute of Molecular Biophysics, Florida State University, Tallahassee, Florida 32306-4380 and.
| |
Collapse
|
405
|
DiMaio F, Yu X, Rensen E, Krupovic M, Prangishvili D, Egelman EH. Virology. A virus that infects a hyperthermophile encapsidates A-form DNA. Science 2015; 348:914-7. [PMID: 25999507 DOI: 10.1126/science.aaa4181] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Extremophiles, microorganisms thriving in extreme environmental conditions, must have proteins and nucleic acids that are stable at extremes of temperature and pH. The nonenveloped, rod-shaped virus SIRV2 (Sulfolobus islandicus rod-shaped virus 2) infects the hyperthermophilic acidophile Sulfolobus islandicus, which lives at 80°C and pH 3. We have used cryo-electron microscopy to generate a three-dimensional reconstruction of the SIRV2 virion at ~4 angstrom resolution, which revealed a previously unknown form of virion organization. Although almost half of the capsid protein is unstructured in solution, this unstructured region folds in the virion into a single extended α helix that wraps around the DNA. The DNA is entirely in the A-form, which suggests a common mechanism with bacterial spores for protecting DNA in the most adverse environments.
Collapse
Affiliation(s)
- Frank DiMaio
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Xiong Yu
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA 22908, USA
| | - Elena Rensen
- Institut Pasteur, Department of Microbiology, 25 rue du Dr. Roux, Paris 75015, France
| | - Mart Krupovic
- Institut Pasteur, Department of Microbiology, 25 rue du Dr. Roux, Paris 75015, France
| | - David Prangishvili
- Institut Pasteur, Department of Microbiology, 25 rue du Dr. Roux, Paris 75015, France.
| | - Edward H Egelman
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA 22908, USA.
| |
Collapse
|
406
|
Matheisl S, Berninghausen O, Becker T, Beckmann R. Structure of a human translation termination complex. Nucleic Acids Res 2015; 43:8615-26. [PMID: 26384426 PMCID: PMC4605324 DOI: 10.1093/nar/gkv909] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 08/12/2015] [Indexed: 12/02/2022] Open
Abstract
In contrast to bacteria that have two release factors, RF1 and RF2, eukaryotes only possess one unrelated release factor eRF1, which recognizes all three stop codons of the mRNA and hydrolyses the peptidyl-tRNA bond. While the molecular basis for bacterial termination has been elucidated, high-resolution structures of eukaryotic termination complexes have been lacking. Here we present a 3.8 Å structure of a human translation termination complex with eRF1 decoding a UAA(A) stop codon. The complex was formed using the human cytomegalovirus (hCMV) stalling peptide, which perturbs the peptidyltransferase center (PTC) to silence the hydrolysis activity of eRF1. Moreover, unlike sense codons or bacterial stop codons, the UAA stop codon adopts a U-turn-like conformation within a pocket formed by eRF1 and the ribosome. Inducing the U-turn conformation for stop codon recognition rationalizes how decoding by eRF1 includes monitoring geometry in order to discriminate against sense codons.
Collapse
Affiliation(s)
- Sarah Matheisl
- Gene Center and Center for integrated Protein Science Munich, Department of Biochemistry, Feodor-Lynen-Str. 25, University of Munich, 81377 Munich, Germany
| | - Otto Berninghausen
- Gene Center and Center for integrated Protein Science Munich, Department of Biochemistry, Feodor-Lynen-Str. 25, University of Munich, 81377 Munich, Germany
| | - Thomas Becker
- Gene Center and Center for integrated Protein Science Munich, Department of Biochemistry, Feodor-Lynen-Str. 25, University of Munich, 81377 Munich, Germany
| | - Roland Beckmann
- Gene Center and Center for integrated Protein Science Munich, Department of Biochemistry, Feodor-Lynen-Str. 25, University of Munich, 81377 Munich, Germany
| |
Collapse
|
407
|
Imai H, Shima T, Sutoh K, Walker ML, Knight PJ, Kon T, Burgess SA. Direct observation shows superposition and large scale flexibility within cytoplasmic dynein motors moving along microtubules. Nat Commun 2015; 6:8179. [PMID: 26365535 PMCID: PMC4579568 DOI: 10.1038/ncomms9179] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 07/25/2015] [Indexed: 12/16/2022] Open
Abstract
Cytoplasmic dynein is a dimeric AAA(+) motor protein that performs critical roles in eukaryotic cells by moving along microtubules using ATP. Here using cryo-electron microscopy we directly observe the structure of Dictyostelium discoideum dynein dimers on microtubules at near-physiological ATP concentrations. They display remarkable flexibility at a hinge close to the microtubule binding domain (the stalkhead) producing a wide range of head positions. About half the molecules have the two heads separated from one another, with both leading and trailing motors attached to the microtubule. The other half have the two heads and stalks closely superposed in a front-to-back arrangement of the AAA(+) rings, suggesting specific contact between the heads. All stalks point towards the microtubule minus end. Mean stalk angles depend on the separation between their stalkheads, which allows estimation of inter-head tension. These findings provide a structural framework for understanding dynein's directionality and unusual stepping behaviour.
Collapse
Affiliation(s)
- Hiroshi Imai
- School of Molecular and Cellular Biology and Astbury Centre for Structural Molecular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Tomohiro Shima
- Quantitative Biology Center, Riken, 6-2-3 Furuedai, Suita, Osaka 565-0874, Japan
| | - Kazuo Sutoh
- Faculty of Science and Engineering, Waseda University, Takada 1-17-22, Toshima-ku, Tokyo 171-0033, Japan
| | | | - Peter J. Knight
- School of Molecular and Cellular Biology and Astbury Centre for Structural Molecular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Takahide Kon
- Department of Biological Sciences, Graduate School of Science, Osaka University, 1-1 Machikaneyama-cho, Toyonaka, 560-0043 Osaka, Japan
- Japan Science and Technology Agency, Precursory Research for Embryonic Science and Technology, Kawaguchi, 332-0012 Saitama, Japan
| | - Stan A. Burgess
- School of Molecular and Cellular Biology and Astbury Centre for Structural Molecular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| |
Collapse
|
408
|
Abstract
Alzheimer's disease (AD) is a fatal neurodegenerative disorder in humans and the main cause of dementia in aging societies. The disease is characterized by the aberrant formation of β-amyloid (Aβ) peptide oligomers and fibrils. These structures may damage the brain and give rise to cerebral amyloid angiopathy, neuronal dysfunction, and cellular toxicity. Although the connection between AD and Aβ fibrillation is extensively documented, much is still unknown about the formation of these Aβ aggregates and their structures at the molecular level. Here, we combined electron cryomicroscopy, 3D reconstruction, and integrative structural modeling methods to determine the molecular architecture of a fibril formed by Aβ(1-42), a particularly pathogenic variant of Aβ peptide. Our model reveals that the individual layers of the Aβ fibril are formed by peptide dimers with face-to-face packing. The two peptides forming the dimer possess identical tilde-shaped conformations and interact with each other by packing of their hydrophobic C-terminal β-strands. The peptide C termini are located close to the main fibril axis, where they produce a hydrophobic core and are surrounded by the structurally more flexible and charged segments of the peptide N termini. The observed molecular architecture is compatible with the general chemical properties of Aβ peptide and provides a structural basis for various biological observations that illuminate the molecular underpinnings of AD. Moreover, the structure provides direct evidence for a steric zipper within a fibril formed by full-length Aβ peptide.
Collapse
|
409
|
Translational arrest by a prokaryotic signal recognition particle is mediated by RNA interactions. Nat Struct Mol Biol 2015; 22:767-73. [PMID: 26344568 DOI: 10.1038/nsmb.3086] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Accepted: 08/13/2015] [Indexed: 12/25/2022]
Abstract
The signal recognition particle (SRP) recognizes signal sequences of nascent polypeptides and targets ribosome-nascent chain complexes to membrane translocation sites. In eukaryotes, translating ribosomes are slowed down by the Alu domain of SRP to allow efficient targeting. In prokaryotes, however, little is known about the structure and function of Alu domain-containing SRPs. Here, we report a complete molecular model of SRP from the Gram-positive bacterium Bacillus subtilis, based on cryo-EM. The SRP comprises two subunits, 6S RNA and SRP54 or Ffh, and it facilitates elongation slowdown similarly to its eukaryotic counterpart. However, protein contacts with the small ribosomal subunit observed for the mammalian Alu domain are substituted in bacteria by RNA-RNA interactions of 6S RNA with the α-sarcin-ricin loop and helices H43 and H44 of 23S rRNA. Our findings provide a structural basis for cotranslational targeting and RNA-driven elongation arrest in prokaryotes.
Collapse
|
410
|
des Georges A, Dhote V, Kuhn L, Hellen CUT, Pestova TV, Frank J, Hashem Y. Structure of mammalian eIF3 in the context of the 43S preinitiation complex. Nature 2015; 525:491-5. [PMID: 26344199 DOI: 10.1038/nature14891] [Citation(s) in RCA: 190] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2015] [Accepted: 07/06/2015] [Indexed: 12/11/2022]
Abstract
During eukaryotic translation initiation, 43S complexes, comprising a 40S ribosomal subunit, initiator transfer RNA and initiation factors (eIF) 2, 3, 1 and 1A, attach to the 5'-terminal region of messenger RNA and scan along it to the initiation codon. Scanning on structured mRNAs also requires the DExH-box protein DHX29. Mammalian eIF3 contains 13 subunits and participates in nearly all steps of translation initiation. Eight subunits having PCI (proteasome, COP9 signalosome, eIF3) or MPN (Mpr1, Pad1, amino-terminal) domains constitute the structural core of eIF3, to which five peripheral subunits are flexibly linked. Here we present a cryo-electron microscopy structure of eIF3 in the context of the DHX29-bound 43S complex, showing the PCI/MPN core at ∼6 Å resolution. It reveals the organization of the individual subunits and their interactions with components of the 43S complex. We were able to build near-complete polyalanine-level models of the eIF3 PCI/MPN core and of two peripheral subunits. The implications for understanding mRNA ribosomal attachment and scanning are discussed.
Collapse
Affiliation(s)
- Amedee des Georges
- HHMI, Department of Biochemistry and Molecular Biophysics, Columbia University, New York, New York 10032, USA
| | - Vidya Dhote
- Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, New York 11203, USA
| | - Lauriane Kuhn
- CNRS, Proteomic Platform Strasbourg - Esplanade, Strasbourg 67084, France
| | - Christopher U T Hellen
- Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, New York 11203, USA
| | - Tatyana V Pestova
- Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, New York 11203, USA
| | - Joachim Frank
- HHMI, Department of Biochemistry and Molecular Biophysics, Columbia University, New York, New York 10032, USA.,Department of Biological Sciences, Columbia University, New York, New York 10032, USA
| | - Yaser Hashem
- CNRS, Architecture et Réactivité de l'ARN, Université de Strasbourg, Strasbourg 67084, France
| |
Collapse
|
411
|
Transmission electron microscopy in molecular structural biology: A historical survey. Arch Biochem Biophys 2015; 581:3-18. [DOI: 10.1016/j.abb.2014.11.011] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Revised: 11/14/2014] [Accepted: 11/21/2014] [Indexed: 01/21/2023]
|
412
|
Maheshwari A, Obbineni J, Bui K, Shibata K, Toyoshima Y, Ishikawa T. α- and β-Tubulin Lattice of the Axonemal Microtubule Doublet and Binding Proteins Revealed by Single Particle Cryo-Electron Microscopy and Tomography. Structure 2015. [DOI: 10.1016/j.str.2015.06.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
413
|
Nilsson OB, Hedman R, Marino J, Wickles S, Bischoff L, Johansson M, Müller-Lucks A, Trovato F, Puglisi JD, O'Brien EP, Beckmann R, von Heijne G. Cotranslational Protein Folding inside the Ribosome Exit Tunnel. Cell Rep 2015; 12:1533-40. [PMID: 26321634 PMCID: PMC4571824 DOI: 10.1016/j.celrep.2015.07.065] [Citation(s) in RCA: 191] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Revised: 07/17/2015] [Accepted: 07/29/2015] [Indexed: 12/25/2022] Open
Abstract
At what point during translation do proteins fold? It is well established that proteins can fold cotranslationally outside the ribosome exit tunnel, whereas studies of folding inside the exit tunnel have so far detected only the formation of helical secondary structure and collapsed or partially structured folding intermediates. Here, using a combination of cotranslational nascent chain force measurements, inter-subunit fluorescence resonance energy transfer studies on single translating ribosomes, molecular dynamics simulations, and cryoelectron microscopy, we show that a small zinc-finger domain protein can fold deep inside the vestibule of the ribosome exit tunnel. Thus, for small protein domains, the ribosome itself can provide the kind of sheltered folding environment that chaperones provide for larger proteins. Cotranslational folding is studied by arrest-peptide-mediated force measurements Single-molecule measurements show that a pulling force prevents ribosome stalling A ribosome-tethered zinc-finger domain is visualized by cryo-EM (electron microscopy) The zinc-finger domain is shown to fold deep inside the ribosome exit tunnel
Collapse
Affiliation(s)
- Ola B Nilsson
- Department of Biochemistry and Biophysics, Center for Biomembrane Research, Stockholm University, 106 91 Stockholm, Sweden
| | - Rickard Hedman
- Department of Biochemistry and Biophysics, Center for Biomembrane Research, Stockholm University, 106 91 Stockholm, Sweden
| | - Jacopo Marino
- Gene Center and Center for Integrated Protein Science Munich, CiPS-M, Feodor-Lynen-Strasse 25, University of Munich, 81377 Munich, Germany
| | - Stephan Wickles
- Gene Center and Center for Integrated Protein Science Munich, CiPS-M, Feodor-Lynen-Strasse 25, University of Munich, 81377 Munich, Germany
| | - Lukas Bischoff
- Gene Center and Center for Integrated Protein Science Munich, CiPS-M, Feodor-Lynen-Strasse 25, University of Munich, 81377 Munich, Germany
| | - Magnus Johansson
- Department of Cell and Molecular Biology, Biomedical Center, Uppsala University, Box 596, 751 24 Uppsala, Sweden
| | - Annika Müller-Lucks
- Department of Biochemistry and Biophysics, Center for Biomembrane Research, Stockholm University, 106 91 Stockholm, Sweden
| | - Fabio Trovato
- Department of Chemistry, Pennsylvania State University, University Park, PA 16802, USA
| | - Joseph D Puglisi
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA 94305-5126, USA; Stanford Magnetic Resonance Laboratory, Stanford University School of Medicine, Stanford, CA 94305-5126, USA
| | - Edward P O'Brien
- Department of Chemistry, Pennsylvania State University, University Park, PA 16802, USA
| | - Roland Beckmann
- Gene Center and Center for Integrated Protein Science Munich, CiPS-M, Feodor-Lynen-Strasse 25, University of Munich, 81377 Munich, Germany
| | - Gunnar von Heijne
- Department of Biochemistry and Biophysics, Center for Biomembrane Research, Stockholm University, 106 91 Stockholm, Sweden; Science for Life Laboratory, Stockholm University, Box 1031, 171 21 Solna, Sweden.
| |
Collapse
|
414
|
Nogales E, Scheres SHW. Cryo-EM: A Unique Tool for the Visualization of Macromolecular Complexity. Mol Cell 2015; 58:677-89. [PMID: 26000851 DOI: 10.1016/j.molcel.2015.02.019] [Citation(s) in RCA: 246] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
3D cryo-electron microscopy (cryo-EM) is an expanding structural biology technique that has recently undergone a quantum leap progression in its achievable resolution and its applicability to the study of challenging biological systems. Because crystallization is not required, only small amounts of sample are needed, and because images can be classified in a computer, the technique has the potential to deal with compositional and conformational mixtures. Therefore, cryo-EM can be used to investigate complete and fully functional macromolecular complexes in different functional states, providing a richness of biological insight. In this review, we underlie some of the principles behind the cryo-EM methodology of single particle analysis and discuss some recent results of its application to challenging systems of paramount biological importance. We place special emphasis on new methodological developments that are leading to an explosion of new studies, many of which are reaching resolutions that could only be dreamed of just a couple of years ago.
Collapse
Affiliation(s)
- Eva Nogales
- Molecular and Cell Biology Department, UC Berkeley, Berkeley, CA 94720-3220, USA; Howard Hughes Medical Institute, UC Berkeley, Berkeley, CA 94720-3220, USA; Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA.
| | - Sjors H W Scheres
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge Biomedical Campus, Cambridge CB2 0QH, UK
| |
Collapse
|
415
|
Byun S, Lim S, Mun JY, Kim KH, Ramadhar TR, Farrand L, Shin SH, Thimmegowda NR, Lee HJ, Frank DA, Clardy J, Lee SW, Lee KW. Identification of a Dual Inhibitor of Janus Kinase 2 (JAK2) and p70 Ribosomal S6 Kinase1 (S6K1) Pathways. J Biol Chem 2015; 290:23553-62. [PMID: 26242912 DOI: 10.1074/jbc.m115.662445] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Indexed: 01/06/2023] Open
Abstract
Bioactive phytochemicals can suppress the growth of malignant cells, and investigation of the mechanisms responsible can assist in the identification of novel therapeutic strategies for cancer therapy. Ginger has been reported to exhibit potent anti-cancer effects, although previous reports have often focused on a narrow range of specific compounds. Through a direct comparison of various ginger compounds, we determined that gingerenone A selectively kills cancer cells while exhibiting minimal toxicity toward normal cells. Kinase array screening revealed JAK2 and S6K1 as the molecular targets primarily responsible for gingerenone A-induced cancer cell death. The effect of gingerenone A was strongly associated with relative phosphorylation levels of JAK2 and S6K1, and administration of gingerenone A significantly suppressed tumor growth in vivo. More importantly, the combined inhibition of JAK2 and S6K1 by commercial inhibitors selectively induced apoptosis in cancer cells, whereas treatment with either agent alone did not. These findings provide rationale for dual targeting of JAK2 and S6K1 in cancer for a combinatorial therapeutic approach.
Collapse
Affiliation(s)
- Sanguine Byun
- From the Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts 02129, Advanced Institutes of Convergence Technology, Seoul National University, Suwon 443-270, Republic of Korea
| | - Semi Lim
- WCU Biomodulation Major, Department of Agricultural Biotechnology, Seoul National University, Seoul 151-742, Republic of Korea
| | - Ji Young Mun
- Department of Biomedical Laboratory Science, College of Health Sciences, Eulji University, Seongnam 461-463, Republic of Korea
| | - Ki Hyun Kim
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, School of Pharmacy, Sungkyunkwan University, Suwon 440-746, Republic of Korea
| | - Timothy R Ramadhar
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115
| | - Lee Farrand
- Yuhan Research Institute, Yuhan Corp., Yongin 446-902, Republic of Korea
| | - Seung Ho Shin
- Program in Biomedical Informatics and Computational Biology, University of Minnesota, Minneapolis, Minnesota 55455
| | - N R Thimmegowda
- Department of Chemistry, Government Sri Krishnarajendra Silver Jubilee Technological Institute, Bangalore 560001, India, World Class Institute, Korea Research Institute of Bioscience and Biotechnology, Ochang 363-883, Korea
| | - Hyong Joo Lee
- WCU Biomodulation Major, Department of Agricultural Biotechnology, Seoul National University, Seoul 151-742, Republic of Korea
| | - David A Frank
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02115, and
| | - Jon Clardy
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115,
| | - Sam W Lee
- From the Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts 02129,
| | - Ki Won Lee
- Advanced Institutes of Convergence Technology, Seoul National University, Suwon 443-270, Republic of Korea, WCU Biomodulation Major, Department of Agricultural Biotechnology, Seoul National University, Seoul 151-742, Republic of Korea,
| |
Collapse
|
416
|
Rosenberg MF, Bikadi Z, Hazai E, Starborg T, Kelley L, Chayen NE, Ford RC, Mao Q. Three-dimensional structure of the human breast cancer resistance protein (BCRP/ABCG2) in an inward-facing conformation. ACTA CRYSTALLOGRAPHICA. SECTION D, BIOLOGICAL CRYSTALLOGRAPHY 2015; 71:1725-35. [PMID: 26249353 PMCID: PMC4528803 DOI: 10.1107/s1399004715010676] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 06/02/2015] [Indexed: 11/10/2022]
Abstract
ABCG2 is an efflux drug transporter that plays an important role in drug resistance and drug disposition. In this study, the first three-dimensional structure of human full-length ABCG2 analysed by electron crystallography from two-dimensional crystals in the absence of nucleotides and transported substrates is reported at 2 nm resolution. In this state, ABCG2 forms a symmetric homodimer with a noncrystallographic twofold axis perpendicular to the two-dimensional crystal plane, as confirmed by subtomogram averaging. This configuration suggests an inward-facing configuration similar to murine ABCB1, with the nucleotide-binding domains (NBDs) widely separated from each other. In the three-dimensional map, densities representing the long cytoplasmic extensions from the transmembrane domains that connect the NBDs are clearly visible. The structural data have allowed the atomic model of ABCG2 to be refined, in which the two arms of the V-shaped ABCG2 homodimeric complex are in a more closed and narrower conformation. The structural data and the refined model of ABCG2 are compatible with the biochemical analysis of the previously published mutagenesis studies, providing novel insight into the structure and function of the transporter.
Collapse
Affiliation(s)
- Mark F. Rosenberg
- Faculty of Life Science, The University of Manchester, Michael Smith Building, Oxford Road, Manchester M13 9PT, England
- Computational and Systems Medicine, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Sir Alexander Fleming Building, London SW7 2AZ, England
| | | | - Eszter Hazai
- Faculty of Life Science, The University of Manchester, Michael Smith Building, Oxford Road, Manchester M13 9PT, England
| | - Tobias Starborg
- Faculty of Life Science, The University of Manchester, Michael Smith Building, Oxford Road, Manchester M13 9PT, England
| | - Lawrence Kelley
- Centre for Bioinformatics, Division of Molecular Biosciences, Department of Life Sciences, Faculty of Natural Sciences, Imperial College London, London SW7 2AZ, England
| | - Naomi E. Chayen
- Computational and Systems Medicine, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Sir Alexander Fleming Building, London SW7 2AZ, England
| | - Robert C. Ford
- Faculty of Life Science, The University of Manchester, Michael Smith Building, Oxford Road, Manchester M13 9PT, England
| | - Qingcheng Mao
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
417
|
Kuang Q, Purhonen P, Pattipaka T, Ayele YH, Hebert H, Koeck PJB. A Refined Single-Particle Reconstruction Procedure to Process Two-Dimensional Crystal Images from Transmission Electron Microscopy. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2015; 21:876-885. [PMID: 25990985 DOI: 10.1017/s1431927615000616] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Single-particle reconstruction (SPR) and electron crystallography (EC), two major applications in electron microscopy, can be used to determine the structure of membrane proteins. The three-dimensional (3D) map is obtained from separated particles in conventional SPR, but from periodic unit cells in EC. Here, we report a refined SPR procedure for processing 2D crystal images. The method is applied to 2D crystals of melibiose permease, a secondary transporter in Escherichia coli. The current procedure is improved from our previously published one in several aspects. The "gold standard Fourier shell correlation" resolution of our final reconstruction reaches 13 Å, which is significantly better than the previously obtained 17 Å resolution. The choices of different refinement parameters for reconstruction are discussed. Our refined SPR procedure could be applied to determine the structure of other membrane proteins in small or locally distorted 2D crystals, which are not ideal for EC.
Collapse
Affiliation(s)
- Qie Kuang
- Karolinska Institutet,Department of Biosciences and Nutrition and KTH Royal Institute of Technology,School of Technology and Health,Novum,S-14183 Huddinge,Sweden
| | - Pasi Purhonen
- Karolinska Institutet,Department of Biosciences and Nutrition and KTH Royal Institute of Technology,School of Technology and Health,Novum,S-14183 Huddinge,Sweden
| | - Thirupathi Pattipaka
- Karolinska Institutet,Department of Biosciences and Nutrition and KTH Royal Institute of Technology,School of Technology and Health,Novum,S-14183 Huddinge,Sweden
| | - Yohannes H Ayele
- Karolinska Institutet,Department of Biosciences and Nutrition and KTH Royal Institute of Technology,School of Technology and Health,Novum,S-14183 Huddinge,Sweden
| | - Hans Hebert
- Karolinska Institutet,Department of Biosciences and Nutrition and KTH Royal Institute of Technology,School of Technology and Health,Novum,S-14183 Huddinge,Sweden
| | - Philip J B Koeck
- Karolinska Institutet,Department of Biosciences and Nutrition and KTH Royal Institute of Technology,School of Technology and Health,Novum,S-14183 Huddinge,Sweden
| |
Collapse
|
418
|
Conformational states of the full-length glucagon receptor. Nat Commun 2015; 6:7859. [PMID: 26227798 PMCID: PMC4532856 DOI: 10.1038/ncomms8859] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Accepted: 06/18/2015] [Indexed: 01/06/2023] Open
Abstract
Class B G protein-coupled receptors are composed of an extracellular domain (ECD) and a seven-transmembrane (7TM) domain, and their signalling is regulated by peptide hormones. Using a hybrid structural biology approach together with the ECD and 7TM domain crystal structures of the glucagon receptor (GCGR), we examine the relationship between full-length receptor conformation and peptide ligand binding. Molecular dynamics (MD) and disulfide crosslinking studies suggest that apo-GCGR can adopt both an open and closed conformation associated with extensive contacts between the ECD and 7TM domain. The electron microscopy (EM) map of the full-length GCGR shows how a monoclonal antibody stabilizes the ECD and 7TM domain in an elongated conformation. Hydrogen/deuterium exchange (HDX) studies and MD simulations indicate that an open conformation is also stabilized by peptide ligand binding. The combined studies reveal the open/closed states of GCGR and suggest that glucagon binds to GCGR by a conformational selection mechanism.
Collapse
|
419
|
Kishchenko GP, Danev R, Fisher R, He J, Hsieh C, Marko M, Sui H. Effect of fringe-artifact correction on sub-tomogram averaging from Zernike phase-plate cryo-TEM. J Struct Biol 2015. [PMID: 26210582 DOI: 10.1016/j.jsb.2015.07.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Zernike phase-plate (ZPP) imaging greatly increases contrast in cryo-electron microscopy, however fringe artifacts appear in the images. A computational de-fringing method has been proposed, but it has not been widely employed, perhaps because the importance of de-fringing has not been clearly demonstrated. For testing purposes, we employed Zernike phase-plate imaging in a cryo-electron tomographic study of radial-spoke complexes attached to microtubule doublets. We found that the contrast enhancement by ZPP imaging made nonlinear denoising insensitive to the filtering parameters, such that simple low-frequency band-pass filtering made the same improvement in map quality. We employed sub-tomogram averaging, which compensates for the effect of the "missing wedge" and considerably improves map quality. We found that fringes (caused by the abrupt cut-on of the central hole in the phase plate) can lead to incorrect representation of a structure that is well-known from the literature. The expected structure was restored by amplitude scaling, as proposed in the literature. Our results show that de-fringing is an important part of image-processing for cryo-electron tomography of macromolecular complexes with ZPP imaging.
Collapse
Affiliation(s)
- Gregory P Kishchenko
- Wadsworth Center, New York State Department of Health, Albany, NY 12201, United States
| | - Radostin Danev
- Max Planck Institute of Biochemistry, Department of Molecular Structural Biology, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Rebecca Fisher
- Wadsworth Center, New York State Department of Health, Albany, NY 12201, United States
| | - Jie He
- Wadsworth Center, New York State Department of Health, Albany, NY 12201, United States
| | - Chyongere Hsieh
- Wadsworth Center, New York State Department of Health, Albany, NY 12201, United States
| | - Michael Marko
- Wadsworth Center, New York State Department of Health, Albany, NY 12201, United States
| | - Haixin Sui
- Wadsworth Center, New York State Department of Health, Albany, NY 12201, United States; Department of Biomedical Sciences, School of Public Health, University at Albany, Albany, NY 12201, United States.
| |
Collapse
|
420
|
Morrone SR, Matyszewski M, Yu X, Delannoy M, Egelman EH, Sohn J. Assembly-driven activation of the AIM2 foreign-dsDNA sensor provides a polymerization template for downstream ASC. Nat Commun 2015. [PMID: 26197926 PMCID: PMC4525163 DOI: 10.1038/ncomms8827] [Citation(s) in RCA: 118] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
AIM2 recognizes foreign dsDNA and assembles into the inflammasome, a filamentous supramolecular signalling platform required to launch innate immune responses. We show here that the pyrin domain of AIM2 (AIM2PYD) drives both filament formation and dsDNA binding. In addition, the dsDNA-binding domain of AIM2 also oligomerizes and assists in filament formation. The ability to oligomerize is critical for binding dsDNA, and in turn permits the size of dsDNA to regulate the assembly of the AIM2 polymers. The AIM2PYD oligomers define the filamentous structure, and the helical symmetry of the AIM2PYD filament is consistent with the filament assembled by the PYD of the downstream adaptor ASC. Our results suggest that the role of AIM2PYD is not autoinhibitory, but generating a structural template by coupling ligand binding and oligomerization is a key signal transduction mechanism in the AIM2 inflammasome. The AIM2 inflammasome complex is essential for defence against a number of human pathogens but how it assembles upon recognition of foreign DNA remains incompletely understood. Here Morrone et al. suggest the AIM2 pyrin domain acts in both DNA binding and filament assembly to generate a structural template for complex assembly.
Collapse
Affiliation(s)
- Seamus R Morrone
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine Baltimore, Maryland 21205, USA
| | - Mariusz Matyszewski
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine Baltimore, Maryland 21205, USA
| | - Xiong Yu
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine Charlottesville, Virginia 22908, USA
| | - Michael Delannoy
- Microscope Core Facilities, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Edward H Egelman
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine Charlottesville, Virginia 22908, USA
| | - Jungsan Sohn
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine Baltimore, Maryland 21205, USA
| |
Collapse
|
421
|
Unique double-ring structure of the peroxisomal Pex1/Pex6 ATPase complex revealed by cryo-electron microscopy. Proc Natl Acad Sci U S A 2015; 112:E4017-25. [PMID: 26170309 DOI: 10.1073/pnas.1500257112] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Members of the AAA family of ATPases assemble into hexameric double rings and perform vital functions, yet their molecular mechanisms remain poorly understood. Here, we report structures of the Pex1/Pex6 complex; mutations in these proteins frequently cause peroxisomal diseases. The structures were determined in the presence of different nucleotides by cryo-electron microscopy. Models were generated using a computational approach that combines Monte Carlo placement of structurally homologous domains into density maps with energy minimization and refinement protocols. Pex1 and Pex6 alternate in an unprecedented hexameric double ring. Each protein has two N-terminal domains, N1 and N2, structurally related to the single N domains in p97 and N-ethylmaleimide sensitive factor (NSF); N1 of Pex1 is mobile, but the others are packed against the double ring. The N-terminal ATPase domains are inactive, forming a symmetric D1 ring, whereas the C-terminal domains are active, likely in different nucleotide states, and form an asymmetric D2 ring. These results suggest how subunit activity is coordinated and indicate striking similarities between Pex1/Pex6 and p97, supporting the hypothesis that the Pex1/Pex6 complex has a role in peroxisomal protein import analogous to p97 in ER-associated protein degradation.
Collapse
|
422
|
Kang CH, Lee SY, Park JH, Lee Y, Jung HS, Chi YH, Jung YJ, Chae HB, Shin MR, Kim WY, Yun D, Lee SY. Stress‐driven structural and functional switching of Ypt1p from a GTPase to a molecular chaperone mediates thermo tolerance in
Saccharomyces cerevisiae. FASEB J 2015; 29:4424-34. [DOI: 10.1096/fj.15-270140] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 06/30/2015] [Indexed: 11/11/2022]
Affiliation(s)
- Chang Ho Kang
- Division of Applied Life Science and Plant Molecular Biology and Biotechnology Research CenterGyeongsang National UniversityJinjuSouth Korea
| | - Sun Yong Lee
- Division of Applied Life Science and Plant Molecular Biology and Biotechnology Research CenterGyeongsang National UniversityJinjuSouth Korea
| | - Joung Hun Park
- Division of Applied Life Science and Plant Molecular Biology and Biotechnology Research CenterGyeongsang National UniversityJinjuSouth Korea
| | - Yuno Lee
- Division of Applied Life Science and Plant Molecular Biology and Biotechnology Research CenterGyeongsang National UniversityJinjuSouth Korea
| | - Hyun Suk Jung
- Division of Electron Microscopic ResearchKorea Basic Science InstituteDaejeonSouth Korea
- Department of BiochemistryCollege of Natural Sciences, Kangwon National UniversityChuncheonSouth Korea
| | - Yong Hun Chi
- Division of Applied Life Science and Plant Molecular Biology and Biotechnology Research CenterGyeongsang National UniversityJinjuSouth Korea
| | - Young Jun Jung
- Division of Applied Life Science and Plant Molecular Biology and Biotechnology Research CenterGyeongsang National UniversityJinjuSouth Korea
| | - Ho Byoung Chae
- Division of Applied Life Science and Plant Molecular Biology and Biotechnology Research CenterGyeongsang National UniversityJinjuSouth Korea
| | - Mi Rim Shin
- Division of Applied Life Science and Plant Molecular Biology and Biotechnology Research CenterGyeongsang National UniversityJinjuSouth Korea
| | - Woe Yeon Kim
- Division of Applied Life Science and Plant Molecular Biology and Biotechnology Research CenterGyeongsang National UniversityJinjuSouth Korea
| | - Dae‐Jin Yun
- Division of Applied Life Science and Plant Molecular Biology and Biotechnology Research CenterGyeongsang National UniversityJinjuSouth Korea
| | - Sang Yeol Lee
- Division of Applied Life Science and Plant Molecular Biology and Biotechnology Research CenterGyeongsang National UniversityJinjuSouth Korea
| |
Collapse
|
423
|
The molecular basis for flexibility in the flexible filamentous plant viruses. Nat Struct Mol Biol 2015; 22:642-4. [PMID: 26167882 PMCID: PMC4527879 DOI: 10.1038/nsmb.3054] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 06/03/2015] [Indexed: 11/08/2022]
Abstract
Flexible filamentous plant viruses cause more than half the viral crop damage in the world but are also potentially useful for biotechnology. Structural studies began more than 75 years ago but have failed, owing to the virion's extreme flexibility. We have used cryo-EM to generate an atomic model for bamboo mosaic virus, which reveals flexible N- and C-terminal extensions that allow deformation while still maintaining structural integrity.
Collapse
|
424
|
da Fonseca PC, Morris EP. Cryo-EM reveals the conformation of a substrate analogue in the human 20S proteasome core. Nat Commun 2015; 6:7573. [PMID: 26133119 PMCID: PMC4506541 DOI: 10.1038/ncomms8573] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 05/20/2015] [Indexed: 11/08/2022] Open
Abstract
The proteasome is a highly regulated protease complex fundamental for cell homeostasis and controlled cell cycle progression. It functions by removing a wide range of specifically tagged proteins, including key cellular regulators. Here we present the structure of the human 20S proteasome core bound to a substrate analogue inhibitor molecule, determined by electron cryo-microscopy (cryo-EM) and single-particle analysis at a resolution of around 3.5 Å. Our map allows the building of protein coordinates as well as defining the location and conformation of the inhibitor at the different active sites. These results open new prospects to tackle the proteasome functional mechanisms. Moreover, they also further demonstrate that cryo-EM is emerging as a realistic approach for general structural studies of protein-ligand interactions.
Collapse
Affiliation(s)
| | - Edward P. Morris
- Division of Structural Biology, The Institute of Cancer Research, London SW3 3RP, UK
| |
Collapse
|
425
|
Lu A, Li Y, Yin Q, Ruan J, Yu X, Egelman E, Wu H. Plasticity in PYD assembly revealed by cryo-EM structure of the PYD filament of AIM2. Cell Discov 2015; 1. [PMID: 26583071 PMCID: PMC4646227 DOI: 10.1038/celldisc.2015.13] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Absent in melanoma 2 (AIM2) is an essential cytosolic double-stranded DNA receptor that assembles with the adaptor, apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC), and caspase-1 to form the AIM2 inflammasome, which leads to proteolytic maturation of cytokines and pyroptotic cell death. AIM2 contains an N-terminal Pyrin domain (PYD) that interacts with ASC through PYD/PYD interactions and nucleates ASCPYD filament formation. To elucidate the molecular basis of AIM2-induced ASCPYD polymerization, we generated AIM2PYD filaments fused to green fluorescent protein (GFP) and determined its cryo-electron microscopic (cryo-EM) structure. The map showed distinct definition of helices, allowing fitting of the crystal structure. Surprisingly, the GFP-AIM2PYD filament is a 1-start helix with helical parameters distinct from those of the 3-start ASCPYD filament. However, despite the apparent symmetry difference, helical net and detailed interface analyses reveal minimal changes in subunit packing. GFP-AIM2PYD nucleated ASCPYD filament formation in comparable efficiency as untagged AIM2PYD, suggesting assembly plasticity in both AIM2PYD and ASCPYD. The DNA-binding domain of AIM2 is able to form AIM2/DNA filaments, within which the AIM2PYD is brought into proximity to template ASCPYD filament assembly. Because ASC is able to interact with many PYD-containing receptors for the formation of inflammasomes, the observed structural plasticity may be critically important for this versatility in the PYD/PYD interactions.
Collapse
Affiliation(s)
- Alvin Lu
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA ; Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
| | - Yang Li
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA ; Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
| | - Qian Yin
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA ; Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
| | - Jianbin Ruan
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA ; Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
| | - Xiong Yu
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, USA
| | - Edward Egelman
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, USA
| | - Hao Wu
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA ; Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
| |
Collapse
|
426
|
Ciniawsky S, Grimm I, Saffian D, Girzalsky W, Erdmann R, Wendler P. Molecular snapshots of the Pex1/6 AAA+ complex in action. Nat Commun 2015; 6:7331. [PMID: 26066397 PMCID: PMC4490564 DOI: 10.1038/ncomms8331] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 04/28/2015] [Indexed: 01/23/2023] Open
Abstract
The peroxisomal proteins Pex1 and Pex6 form a heterohexameric type II AAA+ ATPase complex, which fuels essential protein transport across peroxisomal membranes. Mutations in either ATPase in humans can lead to severe peroxisomal disorders and early death. We present an extensive structural and biochemical analysis of the yeast Pex1/6 complex. The heterohexamer forms a trimer of Pex1/6 dimers with a triangular geometry that is atypical for AAA+ complexes. While the C-terminal nucleotide-binding domains (D2) of Pex6 constitute the main ATPase activity of the complex, both D2 harbour essential substrate-binding motifs. ATP hydrolysis results in a pumping motion of the complex, suggesting that Pex1/6 function involves substrate translocation through its central channel. Mutation of the Walker B motif in one D2 domain leads to ATP hydrolysis in the neighbouring domain, giving structural insights into inter-domain communication of these unique heterohexameric AAA+ assemblies. Pex1 and Pex6 form a heterohexameric AAA+ ATPase complex with triangular geometry at the peroxisome membrane. Here the authors use electron microscopy to show that the complex undergoes conformational changes upon ATP hydrolysis, and demonstrate inter-domain communication between neighbouring nucleotide-binding domains.
Collapse
Affiliation(s)
- Susanne Ciniawsky
- Gene Center Munich, Ludwig-Maximilians-Universität München, Feodor-Lynen-Strasse 25, Munich 81377, Germany
| | - Immanuel Grimm
- Institute of Biochemistry and Pathobiochemistry, Department of Systems Biochemistry, Faculty of Medicine, Ruhr-Universität Bochum, Bochum 44801, Germany
| | - Delia Saffian
- Institute of Biochemistry and Pathobiochemistry, Department of Systems Biochemistry, Faculty of Medicine, Ruhr-Universität Bochum, Bochum 44801, Germany
| | - Wolfgang Girzalsky
- Institute of Biochemistry and Pathobiochemistry, Department of Systems Biochemistry, Faculty of Medicine, Ruhr-Universität Bochum, Bochum 44801, Germany
| | - Ralf Erdmann
- Institute of Biochemistry and Pathobiochemistry, Department of Systems Biochemistry, Faculty of Medicine, Ruhr-Universität Bochum, Bochum 44801, Germany
| | - Petra Wendler
- Gene Center Munich, Ludwig-Maximilians-Universität München, Feodor-Lynen-Strasse 25, Munich 81377, Germany
| |
Collapse
|
427
|
Visualization and quality assessment of the contrast transfer function estimation. J Struct Biol 2015; 192:222-34. [PMID: 26080023 DOI: 10.1016/j.jsb.2015.06.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Revised: 04/20/2015] [Accepted: 06/11/2015] [Indexed: 11/20/2022]
Abstract
The contrast transfer function (CTF) describes an undesirable distortion of image data from a transmission electron microscope. Many users of full-featured processing packages are often new to electron microscopy and are unfamiliar with the CTF concept. Here we present a common graphical output to clearly demonstrate the CTF fit quality independent of estimation software. Separately, many software programs exist to estimate the four CTF parameters, but their results are difficult to compare across multiple runs and it is all but impossible to select the best parameters to use for further processing. A new measurement is presented based on the correlation falloff of the calculated CTF oscillations against the normalized oscillating signal of the data, called the CTF resolution. It was devised to provide a robust numerical quality metric of every CTF estimation for high-throughput screening of micrographs and to select the best parameters for each micrograph. These new CTF visualizations and quantitative measures will help users better assess the quality of their CTF parameters and provide a mechanism to choose the best CTF tool for their data.
Collapse
|
428
|
Nishimura M, Fujii T, Hiyoshi H, Makino F, Inoue H, Motooka D, Kodama T, Ohkubo T, Kobayashi Y, Nakamura S, Namba K, Iida T. A repeat unit of Vibrio diarrheal T3S effector subverts cytoskeletal actin homeostasis via binding to interstrand region of actin filaments. Sci Rep 2015; 5:10870. [PMID: 26039684 PMCID: PMC4650670 DOI: 10.1038/srep10870] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 05/05/2015] [Indexed: 11/09/2022] Open
Abstract
A novel bacterial type III secretion effector, VopV, from the enteric pathogen Vibrio parahaemolyticus has been identified as a key factor in pathogenicity due to its interaction with cytoskeletal actin. One of the repeat units in the long repetitive region of VopV, named VopVrep1, functions as an actin-binding module. Despite its importance in pathogenesis, the manner in which the effector binds to actin and the subsequent effects on actin dynamics remain unclear. Here, we report the molecular basis of the VopVrep1/actin interaction. VopVrep1 exists as an unstructured protein in solution but potently and specifically binds filamentous actin (F-actin) and not globular actin (G-actin). The F-actin/VopVrep1 complex was directly visualized at 9.6-Å resolution using electron cryomicroscopy (cryoEM) and helical image reconstitution. The density map revealed the binding site of VopVrep1 at the interface between two actin strands, which is close to the binding site of the bicyclic heptapeptide toxin phalloidin. Consistent with this observation, VopVrep1 alone prevented the depolymerization of F-actin. Overall, VopVrep1 demonstrated unique characteristics in comparison to known actin-binding proteins, but was relatively similar to phalloidin. The phalloidin-like behavior, targeting the interstrand region of actin filaments to stabilize the filament structure, likely contributes to the pathogenicity of V. parahaemolyticus.
Collapse
Affiliation(s)
- Mitsuhiro Nishimura
- 1] Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan [2] Osaka University of Pharmaceutical Sciences, 4-20-1 Nasahara, Takatsuki, Osaka, 569-1094, Japan
| | - Takashi Fujii
- 1] Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan [2] Riken Quantitative Biology Center, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Hirotaka Hiyoshi
- Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Fumiaki Makino
- Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Hajime Inoue
- Osaka University of Pharmaceutical Sciences, 4-20-1 Nasahara, Takatsuki, Osaka, 569-1094, Japan
| | - Daisuke Motooka
- 1] Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan [2] Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka Suita, Osaka 565-0871, Japan
| | - Toshio Kodama
- Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Tadayasu Ohkubo
- Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka Suita, Osaka 565-0871, Japan
| | - Yuji Kobayashi
- Osaka University of Pharmaceutical Sciences, 4-20-1 Nasahara, Takatsuki, Osaka, 569-1094, Japan
| | - Shota Nakamura
- Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Keiichi Namba
- 1] Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan [2] Riken Quantitative Biology Center, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Tetsuya Iida
- Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| |
Collapse
|
429
|
Chang L, Liu X, Li Y, Liu CC, Yang F, Zhao J, Sui SF. Structural organization of an intact phycobilisome and its association with photosystem II. Cell Res 2015; 25:726-37. [PMID: 25998682 DOI: 10.1038/cr.2015.59] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2014] [Revised: 11/24/2014] [Accepted: 01/29/2015] [Indexed: 11/09/2022] Open
Abstract
Phycobilisomes (PBSs) are light-harvesting antennae that transfer energy to photosynthetic reaction centers in cyanobacteria and red algae. PBSs are supermolecular complexes composed of phycobiliproteins (PBPs) that bear chromophores for energy absorption and linker proteins. Although the structures of some individual components have been determined using crystallography, the three-dimensional structure of an entire PBS complex, which is critical for understanding the energy transfer mechanism, remains unknown. Here, we report the structures of an intact PBS and a PBS in complex with photosystem II (PSII) from Anabaena sp. strain PCC 7120 using single-particle electron microscopy in combination with biochemical and molecular analyses. In the PBS structure, all PBP trimers and the conserved linker protein domains were unambiguously located, and the global distribution of all chromophores was determined. We provide evidence that ApcE and ApcF are critical for the formation of a protrusion at the bottom of PBS, which plays an important role in mediating PBS interaction with PSII. Our results provide insights into the molecular architecture of an intact PBS at different assembly levels and provide the basis for understanding how the light energy absorbed by PBS is transferred to PSII.
Collapse
Affiliation(s)
- Leifu Chang
- 1] State Key Laboratory of Biomembrane and Membrane Biotechnology, Tsinghua University, Beijing 100084, China [2] Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China [3] Current address: MRC Laboratory of Molecular Biology, Cambridge, CB2 0QH, UK
| | - Xianwei Liu
- State Key Laboratory of Protein and Plant Genetic Engineering, College of Life Sciences, Peking University, Beijing 100871, China
| | - Yanbing Li
- State Key Laboratory of Protein and Plant Genetic Engineering, College of Life Sciences, Peking University, Beijing 100871, China
| | - Cui-Cui Liu
- 1] State Key Laboratory of Biomembrane and Membrane Biotechnology, Tsinghua University, Beijing 100084, China [2] Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Fan Yang
- 1] State Key Laboratory of Biomembrane and Membrane Biotechnology, Tsinghua University, Beijing 100084, China [2] Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Jindong Zhao
- 1] State Key Laboratory of Protein and Plant Genetic Engineering, College of Life Sciences, Peking University, Beijing 100871, China [2] Key Laboratory of Phycology of CAS, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei 430072, China
| | - Sen-Fang Sui
- 1] State Key Laboratory of Biomembrane and Membrane Biotechnology, Tsinghua University, Beijing 100084, China [2] Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| |
Collapse
|
430
|
Gold VAM, Salzer R, Averhoff B, Kühlbrandt W. Structure of a type IV pilus machinery in the open and closed state. eLife 2015; 4. [PMID: 25997099 PMCID: PMC4463427 DOI: 10.7554/elife.07380] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 05/20/2015] [Indexed: 01/20/2023] Open
Abstract
Proteins of the secretin family form large macromolecular complexes, which assemble in the outer membrane of Gram-negative bacteria. Secretins are major components of type II and III secretion systems and are linked to extrusion of type IV pili (T4P) and to DNA uptake. By electron cryo-tomography of whole Thermus thermophilus cells, we determined the in situ structure of a T4P molecular machine in the open and the closed state. Comparison reveals a major conformational change whereby the N-terminal domains of the central secretin PilQ shift by ∼30 Å, and two periplasmic gates open to make way for pilus extrusion. Furthermore, we determine the structure of the assembled pilus. DOI:http://dx.doi.org/10.7554/eLife.07380.001 Gram-negative bacteria can cause serious diseases in humans, such as cholera and bacterial meningitis. These bacteria are surrounded by two membranes: an inner membrane and an outer membrane. Proteins called secretins are components of several large molecular complexes that are embedded within the outer membrane. Some secretin-containing complexes form pores in the bacterial membranes and allow molecules to pass in or out of the cell. Some secretins also form part of the machinery that allow Gram-negative bacteria to grow fibre-like structures called type IV pili. These pili help bacteria that cause infections to move and stick to host cells, where they can also trigger massive changes in the host cells' architecture. Multiple copies of a secretin protein called PilQ form a channel in the outer membrane of the bacteria that allows a type IV pilus to grow out of the surface of the cell. The pilus can then hook the bacteria onto surfaces and other cells. There is evidence to suggest the type IV pilus machinery is involved in the uptake of DNA from other bacteria, an important but poorly understood process that has contributed to the spread of multi-drug resistance. Now, Gold et al. have used a cutting-edge technique called ‘electron cryo-tomography’ to analyse the three-dimensional structure of the machinery that builds the type IV pili in the membranes of a bacterium called Thermus thermophilus. This analysis revealed that, similar to many other channel complexes, the PilQ channel can be ‘open’ or ‘closed’. When pili are absent, the channel is closed, but the channel opens when pili are present. Further analysis also revealed the structure of an assembled pilus. Next, Gold et al. studied the open state of the type IV pilus in more detail and observed that a region of each of the PilQ proteins moves a considerable distance to make way for the pilus to enter the central pore. These results will pave the way for future studies of type IV pili and other secretin-containing complexes and underpin efforts to investigate new drug targets to combat bacterial infections. DOI:http://dx.doi.org/10.7554/eLife.07380.002
Collapse
Affiliation(s)
- Vicki A M Gold
- Department of Structural Biology, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
| | - Ralf Salzer
- Molecular Microbiology and Bioenergetics, Institute of Molecular Biosciences, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Beate Averhoff
- Molecular Microbiology and Bioenergetics, Institute of Molecular Biosciences, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Werner Kühlbrandt
- Department of Structural Biology, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
| |
Collapse
|
431
|
Structural rearrangements in the phage head-to-tail interface during assembly and infection. Proc Natl Acad Sci U S A 2015; 112:7009-14. [PMID: 25991862 DOI: 10.1073/pnas.1504039112] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Many icosahedral viruses use a specialized portal vertex to control genome encapsidation and release from the viral capsid. In tailed bacteriophages, the portal system is connected to a tail structure that provides the pipeline for genome delivery to the host cell. We report the first, to our knowledge, subnanometer structures of the complete portal-phage tail interface that mimic the states before and after DNA release during phage infection. They uncover structural rearrangements associated with intimate protein-DNA interactions. The portal protein gp6 of bacteriophage SPP1 undergoes a concerted reorganization of the structural elements of its central channel during interaction with DNA. A network of protein-protein interactions primes consecutive binding of proteins gp15 and gp16 to extend and close the channel. This critical step that prevents genome leakage from the capsid is achieved by a previously unidentified allosteric mechanism: gp16 binding to two different regions of gp15 drives correct positioning and folding of an inner gp16 loop to interact with equivalent loops of the other gp16 subunits. Together, these loops build a plug that closes the channel. Gp16 then fastens the tail to yield the infectious virion. The gatekeeper system opens for viral genome exit at the beginning of infection but recloses afterward, suggesting a molecular diaphragm-like mechanism to control DNA efflux. The mechanisms described here, controlling the essential steps of phage genome movements during virus assembly and infection, are likely to be conserved among long-tailed phages, the largest group of viruses in the Biosphere.
Collapse
|
432
|
Structural analyses of the chromatin remodelling enzymes INO80-C and SWR-C. Nat Commun 2015; 6:7108. [PMID: 25964121 PMCID: PMC4431590 DOI: 10.1038/ncomms8108] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Accepted: 04/01/2015] [Indexed: 01/11/2023] Open
Abstract
INO80-C and SWR-C are conserved members of a subfamily of ATP-dependent chromatin remodeling enzymes that function in transcription and genome-maintenance pathways. A crucial role for these enzymes is to control chromosomal distribution of the H2A.Z histone variant. Here we use electron microscopy (EM) and two-dimensional (2D) class averaging to demonstrate that these remodeling enzymes have similar overall architectures. Each enzyme is characterized by a dynamic ‘tail’ domain and a compact ‘head’ that contains Rvb1/Rvb2 subunits organized as hexameric rings. EM class averages and mass spectrometry support the existence of single heterohexameric rings in both SWR-C and INO80-C. EM studies define the position of the Arp8/Arp4/Act1 module within INO80-C, and we find that this module enhances nucleosome binding affinity but is largely dispensable for remodeling activities. In contrast, the Ies6/Arp5 module is essential for INO80-C remodeling, and furthermore this module controls conformational changes that may couple nucleosome binding to remodeling.
Collapse
|
433
|
Rajagopal P, Tse E, Borst AJ, Delbecq SP, Shi L, Southworth DR, Klevit RE. A conserved histidine modulates HSPB5 structure to trigger chaperone activity in response to stress-related acidosis. eLife 2015; 4. [PMID: 25962097 PMCID: PMC4456606 DOI: 10.7554/elife.07304] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 05/08/2015] [Indexed: 11/13/2022] Open
Abstract
Small heat shock proteins (sHSPs) are essential ‘holdase’ chaperones that form large assemblies and respond dynamically to pH and temperature stresses to protect client proteins from aggregation. While the alpha-crystallin domain (ACD) dimer of sHSPs is the universal building block, how the ACD transmits structural changes in response to stress to promote holdase activity is unknown. We found that the dimer interface of HSPB5 is destabilized over physiological pHs and a conserved histidine (His-104) controls interface stability and oligomer structure in response to acidosis. Destabilization by pH or His-104 mutation shifts the ACD from dimer to monomer but also results in a large expansion of HSPB5 oligomer states. Remarkably, His-104 mutant-destabilized oligomers are efficient holdases that reorganize into structurally distinct client–bound complexes. Our data support a model for sHSP function wherein cell stress triggers small perturbations that alter the ACD building blocks to unleash a cryptic mode of chaperone action. DOI:http://dx.doi.org/10.7554/eLife.07304.001 Proteins are composed of one or more long chain-like molecules that must fold into complex three-dimensional shapes in order to work properly. Incorrectly folded proteins cannot function and often aggregate into toxic states that are associated with a number of neurological diseases including Alzheimer's, Huntington's, and Parkinson's. Elevated temperatures, increased acidity, and other stressful conditions in the cell can hinder the folding process and may cause existing proteins to unfold and aggregate. However, when cells experience these stresses, certain proteins—known as small heat shock proteins (or sHSPs for short)—act as ‘holdase chaperones’ to protect cells from protein misfolding. HSPB5 is one such chaperone that binds to and stabilizes other proteins (called ‘clients’) to prevent their aggregation. The core structure of HSPB5 and other similar chaperone proteins is well known. But, it is not clear how chaperones sense stressful conditions and respond to increase their activity to help stabilize client proteins. Now, Rajagopal et al. have identified a single amino acid in HSPB5 that is sensitive to pH changes. When the environment inside a cell becomes more acidic, this amino acid (a histidine) triggers changes in HSPB5's structure that enhance the chaperone's activity. This histidine was then replaced with another amino acid in an attempt to lock HSPB5 into a low-pH state that mimics an active HSPB5 chaperone inside a stressed cell. Further experiments revealed that this mutant HSPB5 is a super-active holdase chaperone, and that it dramatically changes its structure to bind to a client protein in the holdase state. From this, Rajagopal et al. propose a model to explain how cellular stress triggers small changes in HSPB5 that propagate through the chaperone in a response mechanism that increases its activity. Future studies will investigate whether inherited mutations in HSPB5 and other similar chaperones—which are associated with cardiac, muscle, and nerve disorders—exert their effect by disrupting this response mechanism. DOI:http://dx.doi.org/10.7554/eLife.07304.002
Collapse
Affiliation(s)
- Ponni Rajagopal
- Department of Biochemistry, University of Washington, Seattle, United States
| | - Eric Tse
- Department of Biological Chemistry, Life Sciences Institute, University of Michigan, Ann Arbor, United States
| | - Andrew J Borst
- Department of Biochemistry, University of Washington, Seattle, United States
| | - Scott P Delbecq
- Department of Biochemistry, University of Washington, Seattle, United States
| | - Lei Shi
- Department of Biochemistry, University of Washington, Seattle, United States
| | - Daniel R Southworth
- Department of Biological Chemistry, Life Sciences Institute, University of Michigan, Ann Arbor, United States
| | - Rachel E Klevit
- Department of Biochemistry, University of Washington, Seattle, United States
| |
Collapse
|
434
|
Cianfrocco MA, Leschziner AE. Low cost, high performance processing of single particle cryo-electron microscopy data in the cloud. eLife 2015; 4:e06664. [PMID: 25955969 PMCID: PMC4440898 DOI: 10.7554/elife.06664] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2015] [Accepted: 05/01/2015] [Indexed: 01/27/2023] Open
Abstract
The advent of a new generation of electron microscopes and direct electron detectors has realized the potential of single particle cryo-electron microscopy (cryo-EM) as a technique to generate high-resolution structures. Calculating these structures requires high performance computing clusters, a resource that may be limiting to many likely cryo-EM users. To address this limitation and facilitate the spread of cryo-EM, we developed a publicly available 'off-the-shelf' computing environment on Amazon's elastic cloud computing infrastructure. This environment provides users with single particle cryo-EM software packages and the ability to create computing clusters with 16-480+ CPUs. We tested our computing environment using a publicly available 80S yeast ribosome dataset and estimate that laboratories could determine high-resolution cryo-EM structures for $50 to $1500 per structure within a timeframe comparable to local clusters. Our analysis shows that Amazon's cloud computing environment may offer a viable computing environment for cryo-EM.
Collapse
Affiliation(s)
- Michael A Cianfrocco
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, United States
- Department of Cell Biology, Harvard Medical School, Boston, United States
| | - Andres E Leschziner
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, United States
| |
Collapse
|
435
|
Behrmann E, Loerke J, Budkevich TV, Yamamoto K, Schmidt A, Penczek PA, Vos MR, Bürger J, Mielke T, Scheerer P, Spahn CMT. Structural snapshots of actively translating human ribosomes. Cell 2015; 161:845-57. [PMID: 25957688 PMCID: PMC4432480 DOI: 10.1016/j.cell.2015.03.052] [Citation(s) in RCA: 154] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Revised: 01/05/2015] [Accepted: 02/27/2015] [Indexed: 10/23/2022]
Abstract
Macromolecular machines, such as the ribosome, undergo large-scale conformational changes during their functional cycles. Although their mode of action is often compared to that of mechanical machines, a crucial difference is that, at the molecular dimension, thermodynamic effects dominate functional cycles, with proteins fluctuating stochastically between functional states defined by energetic minima on an energy landscape. Here, we have used cryo-electron microscopy to image ex-vivo-derived human polysomes as a source of actively translating ribosomes. Multiparticle refinement and 3D variability analysis allowed us to visualize a variety of native translation intermediates. Significantly populated states include not only elongation cycle intermediates in pre- and post-translocational states, but also eEF1A-containing decoding and termination/recycling complexes. Focusing on the post-translocational state, we extended this assessment to the single-residue level, uncovering striking details of ribosome-ligand interactions and identifying both static and functionally important dynamic elements.
Collapse
Affiliation(s)
- Elmar Behrmann
- Institut für Medizinische Physik und Biophysik, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Justus Loerke
- Institut für Medizinische Physik und Biophysik, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Tatyana V Budkevich
- Institut für Medizinische Physik und Biophysik, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Kaori Yamamoto
- Institut für Medizinische Physik und Biophysik, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Andrea Schmidt
- Institut für Medizinische Physik und Biophysik, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany; Institut für Medizinische Physik und Biophysik, AG Protein X-Ray Crystallography, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Pawel A Penczek
- Department of Biochemistry and Molecular Biology, The University of Texas Medical School, 6431 Fannin MSB 6.220, Houston, TX 77054, USA
| | - Matthijn R Vos
- FEI Company, Nanoport Europe, Achtseweg Noord 5, 5651 GG Eindhoven, the Netherlands
| | - Jörg Bürger
- Institut für Medizinische Physik und Biophysik, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Thorsten Mielke
- Institut für Medizinische Physik und Biophysik, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany; Max-Planck Institut für Molekulare Genetik, Ihnestraße 63-73, 14195 Berlin, Germany
| | - Patrick Scheerer
- Institut für Medizinische Physik und Biophysik, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany; Institut für Medizinische Physik und Biophysik, AG Protein X-Ray Crystallography, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Christian M T Spahn
- Institut für Medizinische Physik und Biophysik, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany.
| |
Collapse
|
436
|
Zhang X, Zhang L, Tong H, Peng B, Rames MJ, Zhang S, Ren G. 3D Structural Fluctuation of IgG1 Antibody Revealed by Individual Particle Electron Tomography. Sci Rep 2015; 5:9803. [PMID: 25940394 PMCID: PMC4419541 DOI: 10.1038/srep09803] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 03/02/2015] [Indexed: 12/21/2022] Open
Abstract
Commonly used methods for determining protein structure, including X-ray crystallography and single-particle reconstruction, often provide a single and unique three-dimensional (3D) structure. However, in these methods, the protein dynamics and flexibility/fluctuation remain mostly unknown. Here, we utilized advances in electron tomography (ET) to study the antibody flexibility and fluctuation through structural determination of individual antibody particles rather than averaging multiple antibody particles together. Through individual-particle electron tomography (IPET) 3D reconstruction from negatively-stained ET images, we obtained 120 ab-initio 3D density maps at an intermediate resolution (~1-3 nm) from 120 individual IgG1 antibody particles. Using these maps as a constraint, we derived 120 conformations of the antibody via structural flexible docking of the crystal structure to these maps by targeted molecular dynamics simulations. Statistical analysis of the various conformations disclosed the antibody 3D conformational flexibility through the distribution of its domain distances and orientations. This blueprint approach, if extended to other flexible proteins, may serve as a useful methodology towards understanding protein dynamics and functions.
Collapse
Affiliation(s)
- Xing Zhang
- 1] The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA [2] Department of Applied Physics, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Lei Zhang
- The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Huimin Tong
- The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Bo Peng
- The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Matthew J Rames
- The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Shengli Zhang
- Department of Applied Physics, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Gang Ren
- The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| |
Collapse
|
437
|
Cryo-Electron Microscopy Structure of Human Peroxiredoxin-3 Filament Reveals the Assembly of a Putative Chaperone. Structure 2015; 23:912-920. [DOI: 10.1016/j.str.2015.03.019] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Revised: 03/21/2015] [Accepted: 03/23/2015] [Indexed: 01/07/2023]
|
438
|
Fernández-Millán P, Lázaro M, Cansız-Arda Ş, Gerhold JM, Rajala N, Schmitz CA, Silva-Espiña C, Gil D, Bernadó P, Valle M, Spelbrink JN, Solà M. The hexameric structure of the human mitochondrial replicative helicase Twinkle. Nucleic Acids Res 2015; 43:4284-95. [PMID: 25824949 PMCID: PMC4417153 DOI: 10.1093/nar/gkv189] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2015] [Revised: 12/21/2014] [Accepted: 02/23/2015] [Indexed: 01/28/2023] Open
Abstract
The mitochondrial replicative helicase Twinkle is involved in strand separation at the replication fork of mitochondrial DNA (mtDNA). Twinkle malfunction is associated with rare diseases that include late onset mitochondrial myopathies, neuromuscular disorders and fatal infantile mtDNA depletion syndrome. We examined its 3D structure by electron microscopy (EM) and small angle X-ray scattering (SAXS) and built the corresponding atomic models, which gave insight into the first molecular architecture of a full-length SF4 helicase that includes an N-terminal zinc-binding domain (ZBD), an intermediate RNA polymerase domain (RPD) and a RecA-like hexamerization C-terminal domain (CTD). The EM model of Twinkle reveals a hexameric two-layered ring comprising the ZBDs and RPDs in one layer and the CTDs in another. In the hexamer, contacts in trans with adjacent subunits occur between ZBDs and RPDs, and between RPDs and CTDs. The ZBDs show important structural heterogeneity. In solution, the scattering data are compatible with a mixture of extended hexa- and heptameric models in variable conformations. Overall, our structural data show a complex network of dynamic interactions that reconciles with the structural flexibility required for helicase activity.
Collapse
Affiliation(s)
- Pablo Fernández-Millán
- Structural MitoLab; Department of Structural Biology, Molecular Biology Institute Barcelona (IBMB-CSIC), Barcelona, E-08028, Spain
| | - Melisa Lázaro
- Structural Biology Unit. Centre for Cooperative Research in Biosciences, CICbioGUNE, Derio, E-48160, Spain
| | - Şirin Cansız-Arda
- Department of Pediatrics, Nijmegen Centre for Mitochondrial Disorders, Radboud University Medical Centre, Nijmegen, 6525 GA, The Netherlands
| | - Joachim M Gerhold
- Department of Pediatrics, Nijmegen Centre for Mitochondrial Disorders, Radboud University Medical Centre, Nijmegen, 6525 GA, The Netherlands
| | - Nina Rajala
- Mitochondrial DNA Maintenance Group, BioMediTech, University of Tampere, Tampere, FI-33014, Finland
| | - Claus-A Schmitz
- Structural MitoLab; Department of Structural Biology, Molecular Biology Institute Barcelona (IBMB-CSIC), Barcelona, E-08028, Spain
| | - Cristina Silva-Espiña
- Structural MitoLab; Department of Structural Biology, Molecular Biology Institute Barcelona (IBMB-CSIC), Barcelona, E-08028, Spain
| | - David Gil
- Structural Biology Unit. Centre for Cooperative Research in Biosciences, CICbioGUNE, Derio, E-48160, Spain
| | - Pau Bernadó
- Centre de Biochimie Structurale, INSERM-U1054, CNRS UMR-5048, Université de Montpellier I&II. Montpellier, F-34090, France
| | - Mikel Valle
- Structural Biology Unit. Centre for Cooperative Research in Biosciences, CICbioGUNE, Derio, E-48160, Spain
| | - Johannes N Spelbrink
- Department of Pediatrics, Nijmegen Centre for Mitochondrial Disorders, Radboud University Medical Centre, Nijmegen, 6525 GA, The Netherlands Mitochondrial DNA Maintenance Group, BioMediTech, University of Tampere, Tampere, FI-33014, Finland
| | - Maria Solà
- Structural MitoLab; Department of Structural Biology, Molecular Biology Institute Barcelona (IBMB-CSIC), Barcelona, E-08028, Spain
| |
Collapse
|
439
|
Mattaini KR, Brignole EJ, Kini M, Davidson SM, Fiske BP, Drennan CL, Vander Heiden MG. An epitope tag alters phosphoglycerate dehydrogenase structure and impairs ability to support cell proliferation. Cancer Metab 2015; 3:5. [PMID: 25926973 PMCID: PMC4414297 DOI: 10.1186/s40170-015-0131-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 03/31/2015] [Indexed: 11/22/2022] Open
Abstract
Background The gene encoding the serine biosynthesis pathway enzyme PHGDH is located in a region of focal genomic copy number gain in human cancers. Cells with PHGDH amplification are dependent on enzyme expression for proliferation. However, dependence on increased PHGDH expression extends beyond production of serine alone, and further studies of PHGDH function are necessary to elucidate its role in cancer cells. These studies will require a physiologically relevant form of the enzyme for experiments using engineered cell lines and recombinant protein. Results The addition of an N-terminal epitope tag to PHGDH abolished the ability to support proliferation of PHGDH-amplified cells despite retention of some activity to convert 3-PG to PHP. Introducing an R236E mutation into PHGDH eliminates enzyme activity, and this catalytically inactive enzyme cannot support proliferation of PHGDH-dependent cells, arguing that canonical enzyme activity is required. Tagged and untagged PHGDH exhibit the same intracellular localization and ability to produce D-2-hydroxyglutarate (D-2HG), an error product of PHGDH, arguing that neither mislocalization nor loss of D-2HG production explains the inability of epitope-tagged PHGDH to support proliferation. To enable studies of PHGDH function, we report a method to purify recombinant PHGDH and found that untagged enzyme activity was greater than N-terminally tagged enzyme. Analysis of tagged and untagged PHGDH using size exclusion chromatography and electron microscopy found that an N-terminal epitope tag alters enzyme structure. Conclusions Purification of untagged recombinant PHGDH eliminates the need to use an epitope tag for enzyme studies. Furthermore, while tagged PHGDH retains some ability to convert 3PG to PHP, the structural alterations caused by including an epitope tag disrupts the ability of PHGDH to sustain cancer cell proliferation. Electronic supplementary material The online version of this article (doi:10.1186/s40170-015-0131-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Katherine R Mattaini
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139 USA ; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139 USA
| | - Edward J Brignole
- Department of Chemistry Massachusetts Institute of Technology, Cambridge, MA 02139 USA ; Howard Hughes Medical Institute Massachusetts Institute of Technology, Cambridge, MA 02139 USA
| | - Mitali Kini
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139 USA ; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139 USA
| | - Shawn M Davidson
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139 USA ; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139 USA
| | - Brian P Fiske
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139 USA ; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139 USA
| | - Catherine L Drennan
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139 USA ; Department of Chemistry Massachusetts Institute of Technology, Cambridge, MA 02139 USA ; Howard Hughes Medical Institute Massachusetts Institute of Technology, Cambridge, MA 02139 USA
| | - Matthew G Vander Heiden
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139 USA ; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139 USA ; Dana-Farber Cancer Institute, Boston, MA 02215 USA ; Broad Institute, Cambridge, MA 02139 USA
| |
Collapse
|
440
|
Kudryashev M, Wang RYR, Brackmann M, Scherer S, Maier T, Baker D, DiMaio F, Stahlberg H, Egelman EH, Basler M. Structure of the type VI secretion system contractile sheath. Cell 2015; 160:952-962. [PMID: 25723169 DOI: 10.1016/j.cell.2015.01.037] [Citation(s) in RCA: 188] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Revised: 11/17/2014] [Accepted: 01/20/2015] [Indexed: 12/15/2022]
Abstract
Bacteria use rapid contraction of a long sheath of the type VI secretion system (T6SS) to deliver effectors into a target cell. Here, we present an atomic-resolution structure of a native contracted Vibrio cholerae sheath determined by cryo-electron microscopy. The sheath subunits, composed of tightly interacting proteins VipA and VipB, assemble into a six-start helix. The helix is stabilized by a core domain assembled from four β strands donated by one VipA and two VipB molecules. The fold of inner and middle layers is conserved between T6SS and phage sheaths. However, the structure of the outer layer is distinct and suggests a mechanism of interaction of the bacterial sheath with an accessory ATPase, ClpV, that facilitates multiple rounds of effector delivery. Our results provide a mechanistic insight into assembly of contractile nanomachines that bacteria and phages use to translocate macromolecules across membranes.
Collapse
Affiliation(s)
- Mikhail Kudryashev
- Focal Area Infection Biology, Biozentrum, University of Basel, Klingelbergstrasse 50/70, CH-4056 Basel, Switzerland; Center for Cellular Imaging and NanoAnalytics, Biozentrum, University of Basel, Mattenstrasse 26, CH-4058 Basel, Switzerland
| | - Ray Yu-Ruei Wang
- Department of Biochemistry, University of Washington, 1705 NE Pacific Street, Seattle, WA 98195, USA; Graduate Program in Biological Physics, Structure and Design, University of Washington, Box 357350, Seattle, WA 98195, USA
| | - Maximilian Brackmann
- Focal Area Infection Biology, Biozentrum, University of Basel, Klingelbergstrasse 50/70, CH-4056 Basel, Switzerland
| | - Sebastian Scherer
- Center for Cellular Imaging and NanoAnalytics, Biozentrum, University of Basel, Mattenstrasse 26, CH-4058 Basel, Switzerland
| | - Timm Maier
- Focal Area Structural Biology, Biozentrum, University of Basel, Klingelbergstrasse 50/70, CH-4056 Basel, Switzerland
| | - David Baker
- Department of Biochemistry, University of Washington, 1705 NE Pacific Street, Seattle, WA 98195, USA
| | - Frank DiMaio
- Department of Biochemistry, University of Washington, 1705 NE Pacific Street, Seattle, WA 98195, USA
| | - Henning Stahlberg
- Center for Cellular Imaging and NanoAnalytics, Biozentrum, University of Basel, Mattenstrasse 26, CH-4058 Basel, Switzerland
| | - Edward H Egelman
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA 22908, USA.
| | - Marek Basler
- Focal Area Infection Biology, Biozentrum, University of Basel, Klingelbergstrasse 50/70, CH-4056 Basel, Switzerland.
| |
Collapse
|
441
|
Sohmen D, Chiba S, Shimokawa-Chiba N, Innis CA, Berninghausen O, Beckmann R, Ito K, Wilson DN. Structure of the Bacillus subtilis 70S ribosome reveals the basis for species-specific stalling. Nat Commun 2015; 6:6941. [PMID: 25903689 PMCID: PMC4423224 DOI: 10.1038/ncomms7941] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 03/16/2015] [Indexed: 12/23/2022] Open
Abstract
Ribosomal stalling is used to regulate gene expression and can occur in a species-specific manner. Stalling during translation of the MifM leader peptide regulates expression of the downstream membrane protein biogenesis factor YidC2 (YqjG) in Bacillus subtilis, but not in Escherichia coli. In the absence of structures of Gram-positive bacterial ribosomes, a molecular basis for species-specific stalling has remained unclear. Here we present the structure of a Gram-positive B. subtilis MifM-stalled 70S ribosome at 3.5–3.9 Å, revealing a network of interactions between MifM and the ribosomal tunnel, which stabilize a non-productive conformation of the PTC that prevents aminoacyl-tRNA accommodation and thereby induces translational arrest. Complementary genetic analyses identify a single amino acid within ribosomal protein L22 that dictates the species specificity of the stalling event. Such insights expand our understanding of how the synergism between the ribosome and the nascent chain is utilized to modulate the translatome in a species-specific manner. Ribosome stalling regulates gene expression by exposing otherwise inaccessible downstream ribosome-binding sites. Here the authors present a high-resolution Cryo-EM structure of the Bacillus subtilis MifM-stalled 70S ribosome to provide mechanistic insight into species-specific nascent peptide induced translational arrest.
Collapse
Affiliation(s)
- Daniel Sohmen
- Gene Center and Department for Biochemistry, University of Munich, Feodor-Lynen-Street 25, Munich 81377, Germany
| | - Shinobu Chiba
- Faculty of Life Sciences, Kyoto Sangyo University, Motoyama, Kamigamo, Kita-Ku, Kyoto 603-8555, Japan
| | - Naomi Shimokawa-Chiba
- Faculty of Life Sciences, Kyoto Sangyo University, Motoyama, Kamigamo, Kita-Ku, Kyoto 603-8555, Japan
| | - C Axel Innis
- 1] Institut Européen de Chimie et Biologie, Université de Bordeaux, Pessac, France [2] Institut National de la Santé et de la Recherche Médicale (U869), Bordeaux, France
| | - Otto Berninghausen
- Gene Center and Department for Biochemistry, University of Munich, Feodor-Lynen-Street 25, Munich 81377, Germany
| | - Roland Beckmann
- 1] Gene Center and Department for Biochemistry, University of Munich, Feodor-Lynen-Street 25, Munich 81377, Germany [2] Center for integrated Protein Science Munich (CiPSM), University of Munich, Feodor-Lynen-Street 25, Munich 81377, Germany
| | - Koreaki Ito
- Faculty of Life Sciences, Kyoto Sangyo University, Motoyama, Kamigamo, Kita-Ku, Kyoto 603-8555, Japan
| | - Daniel N Wilson
- 1] Gene Center and Department for Biochemistry, University of Munich, Feodor-Lynen-Street 25, Munich 81377, Germany [2] Center for integrated Protein Science Munich (CiPSM), University of Munich, Feodor-Lynen-Street 25, Munich 81377, Germany
| |
Collapse
|
442
|
Egelman EH. Three-dimensional reconstruction of helical polymers. Arch Biochem Biophys 2015; 581:54-8. [PMID: 25912526 DOI: 10.1016/j.abb.2015.04.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 04/06/2015] [Accepted: 04/14/2015] [Indexed: 11/18/2022]
Abstract
The field of three-dimensional electron microscopy began more than 45years ago with a reconstruction of a helical phage tail, and helical polymers continue to be important objects for three-dimensional reconstruction due to the centrality of helical protein and nucleoprotein polymers in all aspects of biology. We are now witnessing a fundamental revolution in this area, made possible by direct electron detectors, which has led to near-atomic resolution for a number of important helical structures. Most importantly, the possibility of achieving such resolution routinely for a vast number of helical samples is within our reach. One of the main problems in helical reconstruction, ambiguities in assigning the helical symmetry, is overcome when one reaches a resolution where secondary structure is clearly visible. However, obstacles still exist due to the intrinsic variability within many helical filaments.
Collapse
Affiliation(s)
- Edward H Egelman
- Department of Biochemistry and Molecular Genetics, University of Virginia, Box 800733, Charlottesville, VA 22908, USA.
| |
Collapse
|
443
|
Cryo-EM structure of the tetracycline resistance protein TetM in complex with a translating ribosome at 3.9-Å resolution. Proc Natl Acad Sci U S A 2015; 112:5401-6. [PMID: 25870267 DOI: 10.1073/pnas.1501775112] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Ribosome protection proteins (RPPs) confer resistance to tetracycline by binding to the ribosome and chasing the drug from its binding site. Current models for RPP action are derived from 7.2- to 16-Å resolution structures of RPPs bound to vacant or nontranslating ribosomes. Here we present a cryo-electron microscopy reconstruction of the RPP TetM in complex with a translating ribosome at 3.9-Å resolution. The structure reveals the contacts of TetM with the ribosome, including interaction between the conserved and functionally critical C-terminal extension of TetM with a unique splayed conformation of nucleotides A1492 and A1493 at the decoding center of the small subunit. The resolution enables us to unambiguously model the side chains of the amino acid residues comprising loop III in domain IV of TetM, revealing that the tyrosine residues Y506 and Y507 are not responsible for drug-release as suggested previously but rather for intrafactor contacts that appear to stabilize the conformation of loop III. Instead, Pro509 at the tip of loop III is located directly within the tetracycline binding site where it interacts with nucleotide C1054 of the 16S rRNA, such that RPP action uses Pro509, rather than Y506/Y507, to directly dislodge and release tetracycline from the ribosome.
Collapse
|
444
|
van der Sluis EO, Bauerschmitt H, Becker T, Mielke T, Frauenfeld J, Berninghausen O, Neupert W, Herrmann JM, Beckmann R. Parallel Structural Evolution of Mitochondrial Ribosomes and OXPHOS Complexes. Genome Biol Evol 2015; 7:1235-51. [PMID: 25861818 PMCID: PMC4453056 DOI: 10.1093/gbe/evv061] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/06/2015] [Indexed: 01/06/2023] Open
Abstract
The five macromolecular complexes that jointly mediate oxidative phosphorylation (OXPHOS) in mitochondria consist of many more subunits than those of bacteria, yet, it remains unclear by which evolutionary mechanism(s) these novel subunits were recruited. Even less well understood is the structural evolution of mitochondrial ribosomes (mitoribosomes): while it was long thought that their exceptionally high protein content would physically compensate for their uniquely low amount of ribosomal RNA (rRNA), this hypothesis has been refuted by structural studies. Here, we present a cryo-electron microscopy structure of the 73S mitoribosome from Neurospora crassa, together with genomic and proteomic analyses of mitoribosome composition across the eukaryotic domain. Surprisingly, our findings reveal that both structurally and compositionally, mitoribosomes have evolved very similarly to mitochondrial OXPHOS complexes via two distinct phases: A constructive phase that mainly acted early in eukaryote evolution, resulting in the recruitment of altogether approximately 75 novel subunits, and a reductive phase that acted during metazoan evolution, resulting in gradual length-reduction of mitochondrially encoded rRNAs and OXPHOS proteins. Both phases can be well explained by the accumulation of (slightly) deleterious mutations and deletions, respectively, in mitochondrially encoded rRNAs and OXPHOS proteins. We argue that the main role of the newly recruited (nuclear encoded) ribosomal- and OXPHOS proteins is to provide structural compensation to the mutationally destabilized mitochondrially encoded components. While the newly recruited proteins probably provide a selective advantage owing to their compensatory nature, and while their presence may have opened evolutionary pathways toward novel mitochondrion-specific functions, we emphasize that the initial events that resulted in their recruitment was nonadaptive in nature. Our framework is supported by population genetic studies, and it can explain the complete structural evolution of mitochondrial ribosomes and OXPHOS complexes, as well as many observed functions of individual proteins.
Collapse
Affiliation(s)
- Eli O van der Sluis
- Gene Center and Center for integrated Protein Science Munich (CiPSM), Department of Biochemistry, University of Munich, Germany
| | | | - Thomas Becker
- Gene Center and Center for integrated Protein Science Munich (CiPSM), Department of Biochemistry, University of Munich, Germany
| | - Thorsten Mielke
- Max Planck Institute for Molecular Genetics, UltraStrukturNetzwerk, Berlin, Germany Institut für Medizinische Physik und Biophysik, Charité, Berlin, Germany
| | - Jens Frauenfeld
- Gene Center and Center for integrated Protein Science Munich (CiPSM), Department of Biochemistry, University of Munich, Germany Present address: Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Otto Berninghausen
- Gene Center and Center for integrated Protein Science Munich (CiPSM), Department of Biochemistry, University of Munich, Germany
| | - Walter Neupert
- Max Planck Institute of Biochemistry, Martinsried, Germany
| | | | - Roland Beckmann
- Gene Center and Center for integrated Protein Science Munich (CiPSM), Department of Biochemistry, University of Munich, Germany
| |
Collapse
|
445
|
Rosenberg OS, Dovala D, Li X, Connolly L, Bendebury A, Finer-Moore J, Holton J, Cheng Y, Stroud RM, Cox JS. Substrates Control Multimerization and Activation of the Multi-Domain ATPase Motor of Type VII Secretion. Cell 2015; 161:501-512. [PMID: 25865481 DOI: 10.1016/j.cell.2015.03.040] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 11/10/2014] [Accepted: 02/11/2015] [Indexed: 01/13/2023]
Abstract
Mycobacterium tuberculosis and Staphylococcus aureus secrete virulence factors via type VII protein secretion (T7S), a system that intriguingly requires all of its secretion substrates for activity. To gain insights into T7S function, we used structural approaches to guide studies of the putative translocase EccC, a unique enzyme with three ATPase domains, and its secretion substrate EsxB. The crystal structure of EccC revealed that the ATPase domains are joined by linker/pocket interactions that modulate its enzymatic activity. EsxB binds via its signal sequence to an empty pocket on the C-terminal ATPase domain, which is accompanied by an increase in ATPase activity. Surprisingly, substrate binding does not activate EccC allosterically but, rather, by stimulating its multimerization. Thus, the EsxB substrate is also an integral T7S component, illuminating a mechanism that helps to explain interdependence of substrates, and suggests a model in which binding of substrates modulates their coordinate release from the bacterium.
Collapse
Affiliation(s)
- Oren S Rosenberg
- Division of Infectious Diseases, Department of Medicine, UCSF Medical Center, University of California, San Francisco, San Francisco, CA 94143-0654, USA
| | - Dustin Dovala
- Department of Microbiology and Immunology, Program in Microbial Pathogenesis and Host Defense, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Xueming Li
- School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Lynn Connolly
- Division of Infectious Diseases, Department of Medicine, UCSF Medical Center, University of California, San Francisco, San Francisco, CA 94143-0654, USA; Achaogen, Inc., South San Francisco, CA 94080, USA
| | - Anastasia Bendebury
- Department of Microbiology and Immunology, Program in Microbial Pathogenesis and Host Defense, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Janet Finer-Moore
- Department of Biophysics and Biochemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - James Holton
- Department of Biophysics and Biochemistry, University of California, San Francisco, San Francisco, CA 94158, USA; Lawrence Berkeley National Laboratory, MS6-2100, Berkeley, CA 94720, USA
| | - Yifan Cheng
- Department of Biophysics and Biochemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Robert M Stroud
- Department of Biophysics and Biochemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Jeffery S Cox
- Department of Microbiology and Immunology, Program in Microbial Pathogenesis and Host Defense, University of California, San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
446
|
Structure of the TRPA1 ion channel suggests regulatory mechanisms. Nature 2015; 520:511-7. [PMID: 25855297 PMCID: PMC4409540 DOI: 10.1038/nature14367] [Citation(s) in RCA: 460] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 03/04/2015] [Indexed: 02/08/2023]
Abstract
The TRPA1 ion channel (a.k.a the ‘wasabi receptor’) is a detector of noxious chemical agents encountered in our environment or produced endogenously during tissue injury or drug metabolism. These include a broad class of electrophiles that activate the channel through covalent protein modification. TRPA1 antagonists hold potential for treating neurogenic inflammatory conditions provoked or exacerbated by irritant exposure. Despite compelling reasons to understand TRPA1 function, structural mechanisms underlying channel regulation remain obscure. Here, we use single-particle electron cryo-microscopy to determine the structure of full-length human TRPA1 to ~4Å resolution in the presence of pharmacophores, including a potent antagonist. A number of unexpected features are revealed, including an extensive coiled-coil assembly domain stabilized by polyphosphate co-factors and a highly integrated nexus that converges on an unpredicted TRP-like allosteric domain. These findings provide novel insights into mechanisms of TRPA1 regulation, and establish a blueprint for structure-based design of analgesic and anti-inflammatory agents.
Collapse
|
447
|
Wang RYR, Kudryashev M, Li X, Egelman EH, Basler M, Cheng Y, Baker D, DiMaio F. De novo protein structure determination from near-atomic-resolution cryo-EM maps. Nat Methods 2015; 12:335-8. [PMID: 25707029 PMCID: PMC4435692 DOI: 10.1038/nmeth.3287] [Citation(s) in RCA: 138] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 12/31/2014] [Indexed: 12/12/2022]
Abstract
We present a de novo model-building approach that combines predicted backbone conformations with side-chain fit to density to accurately assign sequence into density maps. This method yielded accurate models for six of nine experimental maps at 3.3- to 4.8-Å resolution and produced a nearly complete model for an unsolved map containing a 660-residue heterodimeric protein. This method should enable rapid and reliable protein structure determination from near-atomic-resolution cryo-electron microscopy (cryo-EM) maps.
Collapse
Affiliation(s)
- Ray Yu-Ruei Wang
- Graduate program in Biological Physics, Structure and Design, University of Washington, Seattle, WA 98195, USA
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Mikhail Kudryashev
- Focal Area Infection Biology, Biozentrum, University of Basel, Switzerland
- Center for Cellular Imaging and NanoAnalytics, Biozentrum, University of Basel, Switzerland
| | - Xueming Li
- The Keck Advanced Microscopy Laboratory, Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA
| | - Edward H. Egelman
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA 22908-0733, USA
| | - Marek Basler
- Focal Area Infection Biology, Biozentrum, University of Basel, Switzerland
| | - Yifan Cheng
- The Keck Advanced Microscopy Laboratory, Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA
| | - David Baker
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA
| | - Frank DiMaio
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
448
|
Skruzny M, Desfosses A, Prinz S, Dodonova S, Gieras A, Uetrecht C, Jakobi A, Abella M, Hagen W, Schulz J, Meijers R, Rybin V, Briggs J, Sachse C, Kaksonen M. An Organized Co-assembly of Clathrin Adaptors Is Essential for Endocytosis. Dev Cell 2015; 33:150-62. [DOI: 10.1016/j.devcel.2015.02.023] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Revised: 11/29/2014] [Accepted: 02/25/2015] [Indexed: 10/23/2022]
|
449
|
Tracz DM, Gilmour MW, Mabon P, Beniac DR, Hoang L, Kibsey P, Van Domselaar G, Tabor H, Westmacott GR, Corbett CR, Bernard KA. Tatumella saanichensis sp. nov., isolated from a cystic fibrosis patient. Int J Syst Evol Microbiol 2015; 65:1959-1966. [PMID: 25807976 DOI: 10.1099/ijs.0.000207] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Polyphasic taxonomic analysis was performed on a clinical isolate (NML 06-3099T) from a cystic fibrosis patient, including whole-genome sequencing, proteomics, phenotypic testing, electron microscopy, chemotaxonomy and a clinical investigation. Comparative whole-genome sequence analysis and multilocus sequence analysis (MLSA) between Tatumella ptyseos ATCC 33301T and clinical isolate NML 06-3099T suggested that the clinical isolate was closely related to, but distinct from, the species T. ptyseos. By 16S rRNA gene sequencing, the clinical isolate shared 98.7 % sequence identity with T. ptyseos ATCC 33301T. A concatenate of six MLSA loci (totalling 4500 bp) revealed < 93.9 % identity between T. ptyseos ATCC 33301T, other members of the genus and the clinical isolate. A whole-genome sequence comparison between NML 06-3099T and ATCC 33301T determined that the average nucleotide identity was 76.24 %. The overall DNA G+C content of NML 06-3099T was 51.27 %, consistent with members of the genus Tatumella. By matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) MS analysis, NML 06-3099T had a genus-level match, but not a species-level match, to T. ptyseos. By shotgun proteomics, T. ptyseos ATCC 33301T and NML 06-3099T were found to have unique proteomes. The two strains had similar morphologies and multiple fimbriae, as observed by transmission electron microscopy, but were distinguishable by phenotypic testing. Cellular fatty acids found were typical for members of the Enterobacteriaceae. NML 06-3099T was susceptible to commonly used antibiotics. Based on these data, NML 06-3099T represents a novel species in the genus Tatumella, for which the name Tatumella saanichensis sp. nov. is proposed (type strain NML 06-3099T = CCUG 55408T = DSM 19846T).
Collapse
Affiliation(s)
- Dobryan M Tracz
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, Manitoba R3E 3R2, Canada
| | - Matthew W Gilmour
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, Manitoba R3E 3R2, Canada.,Department of Medical Microbiology and Infectious Diseases, Winnipeg, University of Manitoba, Manitoba, Canada
| | - Philip Mabon
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, Manitoba R3E 3R2, Canada
| | - Daniel R Beniac
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, Manitoba R3E 3R2, Canada
| | - Linda Hoang
- Laboratory Services, British Columbia Centre for Disease Control, Provincial Health Services Authority, 655 12th Avenue W., Vancouver, British Columbia, V5Z 4R4, Canada
| | - Pamela Kibsey
- Victoria General Hospital, 1 Hospital Way, Victoria, British Columbia, V8Z 6R5, Canada
| | - Gary Van Domselaar
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, Manitoba R3E 3R2, Canada.,Department of Medical Microbiology and Infectious Diseases, Winnipeg, University of Manitoba, Manitoba, Canada
| | - Helen Tabor
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, Manitoba R3E 3R2, Canada
| | - Garrett R Westmacott
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, Manitoba R3E 3R2, Canada
| | - Cindi R Corbett
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, Manitoba R3E 3R2, Canada.,Department of Medical Microbiology and Infectious Diseases, Winnipeg, University of Manitoba, Manitoba, Canada
| | - Kathryn A Bernard
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, Manitoba R3E 3R2, Canada.,Department of Medical Microbiology and Infectious Diseases, Winnipeg, University of Manitoba, Manitoba, Canada
| |
Collapse
|
450
|
Gaik M, Flemming D, von Appen A, Kastritis P, Mücke N, Fischer J, Stelter P, Ori A, Bui KH, Baßler J, Barbar E, Beck M, Hurt E. Structural basis for assembly and function of the Nup82 complex in the nuclear pore scaffold. ACTA ACUST UNITED AC 2015; 208:283-97. [PMID: 25646085 PMCID: PMC4315244 DOI: 10.1083/jcb.201411003] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The yeast Nup82 complex forms an unusual asymmetric structure with a dimeric array of subunits that mediate its anchorage to the NPC scaffold and its concomitant interaction with the soluble nucleocytoplasmic transport machinery. Nuclear pore complexes (NPCs) are huge assemblies formed from ∼30 different nucleoporins, typically organized in subcomplexes. One module, the conserved Nup82 complex at the cytoplasmic face of NPCs, is crucial to terminate mRNA export. To gain insight into the structure, assembly, and function of the cytoplasmic pore filaments, we reconstituted in yeast the Nup82–Nup159–Nsp1–Dyn2 complex, which was suitable for biochemical, biophysical, and electron microscopy analyses. Our integrative approach revealed that the yeast Nup82 complex forms an unusual asymmetric structure with a dimeric array of subunits. Based on all these data, we developed a three-dimensional structural model of the Nup82 complex that depicts how this module might be anchored to the NPC scaffold and concomitantly can interact with the soluble nucleocytoplasmic transport machinery.
Collapse
Affiliation(s)
- Monika Gaik
- Biochemistry Center of Heidelberg University, D-69120 Heidelberg, Germany
| | - Dirk Flemming
- Biochemistry Center of Heidelberg University, D-69120 Heidelberg, Germany
| | - Alexander von Appen
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, D-69117 Heidelberg, Germany
| | - Panagiotis Kastritis
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, D-69117 Heidelberg, Germany
| | - Norbert Mücke
- Division of Biophysics of Macromolecules, German Center Research Center, D-69120 Heidelberg, Germany
| | - Jessica Fischer
- Biochemistry Center of Heidelberg University, D-69120 Heidelberg, Germany
| | - Philipp Stelter
- Biochemistry Center of Heidelberg University, D-69120 Heidelberg, Germany
| | - Alessandro Ori
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, D-69117 Heidelberg, Germany
| | - Khanh Huy Bui
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, D-69117 Heidelberg, Germany
| | - Jochen Baßler
- Biochemistry Center of Heidelberg University, D-69120 Heidelberg, Germany
| | - Elisar Barbar
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR 97331
| | - Martin Beck
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, D-69117 Heidelberg, Germany
| | - Ed Hurt
- Biochemistry Center of Heidelberg University, D-69120 Heidelberg, Germany
| |
Collapse
|