1351
|
Amenitsch H, Benetti F, Ramos A, Legname G, Requena JR. SAXS structural study of PrP(Sc) reveals ~11 nm diameter of basic double intertwined fibers. Prion 2013; 7:496-500. [PMID: 24247356 DOI: 10.4161/pri.27190] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
A sample of purified Syrian hamster PrP27-30 prion fibers was analyzed by synchrotron small-angle X-ray scattering (SAXS). The SAXS pattern obtained was fitted to a model based on infinitely long cylinders with a log-normal intensity distribution, a hard-sphere structure factor and a general Porod term for larger aggregates. The diameter calculated for the cylinders determined from the fit was 11.0 ± 0.2 nm. This measurement offers an estimation of the diameter of PrP(Sc) fibers in suspension, i.e., free of errors derived from estimations based on 2D projections in transmission electron microscopy images, subjected to further possible distortions from the negative stain. This diameter, which corresponds to a maximum diameter of approximately 5.5 nm for each of the two intertwined protofilaments making up the fibers, rules out the possibility that PrP(Sc) conforms to a stack of in-register, single-rung flat PrP(Sc) monomers; rather, PrP(Sc) subunits must necessarily coil, most likely several times, into themselves.
Collapse
Affiliation(s)
- Heinz Amenitsch
- Institute of Inorganic Chemistry; Graz University of Technology; Graz, Austria
| | - Federico Benetti
- Laboratory of Prion Biology; Department of Neuroscience; Scuola Internazionale Superiore di Studi Avanzati (SISSA); Trieste, Italy
| | - Adriana Ramos
- CIMUS Biomedical Research Institute & Department of Medicine; University of Santiago de Compostela-IDIS; Santiago de Compostela, Spain
| | - Giuseppe Legname
- Laboratory of Prion Biology; Department of Neuroscience; Scuola Internazionale Superiore di Studi Avanzati (SISSA); Trieste, Italy
| | - Jesús R Requena
- CIMUS Biomedical Research Institute & Department of Medicine; University of Santiago de Compostela-IDIS; Santiago de Compostela, Spain
| |
Collapse
|
1352
|
Andresen K, Jimenez-Useche I, Howell SC, Yuan C, Qiu X. Solution scattering and FRET studies on nucleosomes reveal DNA unwrapping effects of H3 and H4 tail removal. PLoS One 2013; 8:e78587. [PMID: 24265699 PMCID: PMC3827064 DOI: 10.1371/journal.pone.0078587] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Accepted: 09/13/2013] [Indexed: 11/18/2022] Open
Abstract
Using a combination of small-angle X-ray scattering (SAXS) and fluorescence resonance energy transfer (FRET) measurements we have determined the role of the H3 and H4 histone tails, independently, in stabilizing the nucleosome DNA terminal ends from unwrapping from the nucleosome core. We have performed solution scattering experiments on recombinant wild-type, H3 and H4 tail-removed mutants and fit all scattering data with predictions from PDB models and compared these experiments to complementary DNA-end FRET experiments. Based on these combined SAXS and FRET studies, we find that while all nucleosomes exhibited DNA unwrapping, the extent of this unwrapping is increased for nucleosomes with the H3 tails removed but, surprisingly, decreased in nucleosomes with the H4 tails removed. Studies of salt concentration effects show a minimum amount of DNA unwrapping for all complexes around 50-100mM of monovalent ions. These data exhibit opposite roles for the positively-charged nucleosome tails, with the ability to decrease access (in the case of the H3 histone) or increase access (in the case of the H4 histone) to the DNA surrounding the nucleosome. In the range of salt concentrations studied (0-200mM KCl), the data point to the H4 tail-removed mutant at physiological (50-100mM) monovalent salt concentration as the mononucleosome with the least amount of DNA unwrapping.
Collapse
Affiliation(s)
- Kurt Andresen
- Department of Physics, Gettysburg College, Gettysburg, Pennsylvania, United States of America
- * E-mail:
| | - Isabel Jimenez-Useche
- School of Chemical Engineering, Purdue University, West Lafayette, Indiana, United States of America
| | - Steven C. Howell
- Department of Physics, George Washington University, Washington, District of Columbia, United States of America
| | - Chongli Yuan
- School of Chemical Engineering, Purdue University, West Lafayette, Indiana, United States of America
| | - Xiangyun Qiu
- Department of Physics, George Washington University, Washington, District of Columbia, United States of America
| |
Collapse
|
1353
|
Wang W, Hou H, Du Q, Zhang W, Liu G, Shtykova EV, Xu J, Liu P, Dong Y. Solution small angle X-ray scattering (SAXS) studies of RecQ from Deinococcus radiodurans and its complexes with junction DNA substrates. J Biol Chem 2013; 288:32414-32423. [PMID: 24068706 PMCID: PMC3820876 DOI: 10.1074/jbc.m113.502112] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Revised: 09/14/2013] [Indexed: 11/06/2022] Open
Abstract
RecQ helicases, essential enzymes for maintaining genome integrity, possess the capability to participate in a wide variety of DNA metabolisms. They can initiate the homologous recombination repair pathway by unwinding damaged dsDNA and suppress hyper-recombination by promoting Holliday junction (HJ) migration. To learn how DrRecQ participates in the homologous recombination repair pathway, solution structures of Deinococcus radiodurans RecQ (DrRecQ) and its complexes with DNA substrates were investigated by small angle x-ray scattering. We found that the catalytic core and the most N-terminal HRDC (helicase and RNase D C-terminal) domain (HRDC1) undergo a conformational change to a compact state upon binding to a junction DNA. Furthermore, models of DrRecQ in complexes with two kinds of junction DNA (fork junction and HJ) were built based on the small angle x-ray scattering data, and together with the EMSA results, possible binding sites were proposed. It is demonstrated that two DrRecQ molecules bind to the opposite arms of HJ. This architecture is similar to the RuvAB complex and is hypothesized to be highly conserved in the other HJ migration proteins. This work provides us new clues to understand the roles DrRecQ plays in the RecFOR pathway.
Collapse
Affiliation(s)
- Wenjia Wang
- From the Beijing Synchrotron Radiation Facility, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China
| | - Haifeng Hou
- From the Beijing Synchrotron Radiation Facility, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China
| | - Qian Du
- the Department of Plant Sciences, College of Agriculture and Nature Resources, University of Connecticut, Storrs, Connecticut 06269
| | - Wen Zhang
- the Department of Physiology, The University of Hong Kong, Hong Kong SAR 999077, China
| | - Guangfeng Liu
- From the Beijing Synchrotron Radiation Facility, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China
| | - Eleonora V Shtykova
- the Institute of Crystallography, Russian Academy of Sciences, Moscow 117333, Russia
| | - Jianhua Xu
- From the Beijing Synchrotron Radiation Facility, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China
| | - Peng Liu
- From the Beijing Synchrotron Radiation Facility, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China,.
| | - Yuhui Dong
- From the Beijing Synchrotron Radiation Facility, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China,.
| |
Collapse
|
1354
|
The Salmonella enterica ZinT structure, zinc affinity and interaction with the high-affinity uptake protein ZnuA provide insight into the management of periplasmic zinc. Biochim Biophys Acta Gen Subj 2013; 1840:535-44. [PMID: 24128931 DOI: 10.1016/j.bbagen.2013.10.010] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Revised: 09/17/2013] [Accepted: 10/07/2013] [Indexed: 11/22/2022]
Abstract
BACKGROUND In Gram-negative bacteria the ZnuABC transporter ensures adequate zinc import in Zn(II)-poor environments, like those encountered by pathogens within the infected host. Recently, the metal-binding protein ZinT was suggested to operate as an accessory component of ZnuABC in periplasmic zinc recruitment. Since ZinT is known to form a ZinT-ZnuA complex in the presence of Zn(II) it was proposed to transfer Zn(II) to ZnuA. The present work was undertaken to test this claim. METHODS ZinT and its structural relationship with ZnuA have been characterized by multiple biophysical techniques (X-ray crystallography, SAXS, analytical ultracentrifugation, fluorescence spectroscopy). RESULTS The metal-free and metal-bound crystal structures of Salmonella enterica ZinT show one Zn(II) binding site and limited structural changes upon metal removal. Spectroscopic titrations with Zn(II) yield a KD value of 22±2nM for ZinT, while those with ZnuA point to one high affinity (KD<20nM) and one low affinity Zn(II) binding site (KD in the micromolar range). Sedimentation velocity experiments established that Zn(II)-bound ZinT interacts with ZnuA, whereas apo-ZinT does not. The model of the ZinT-ZnuA complex derived from small angle X-ray scattering experiments points to a disposition that favors metal transfer as the metal binding cavities of the two proteins face each other. CONCLUSIONS ZinT acts as a Zn(II)-buffering protein that delivers Zn(II) to ZnuA. GENERAL SIGNIFICANCE Knowledge of the ZinT-ZnuA relationship is crucial for understanding bacterial Zn(II) uptake.
Collapse
|
1355
|
Petoukhov MV, Billas IML, Takacs M, Graewert MA, Moras D, Svergun DI. Reconstruction of quaternary structure from X-ray scattering by equilibrium mixtures of biological macromolecules. Biochemistry 2013; 52:6844-55. [PMID: 24000896 DOI: 10.1021/bi400731u] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
A recent renaissance in small-angle X-ray scattering (SAXS) made this technique a major tool for the low-resolution structural characterization of biological macromolecules in solution. The major limitation of existing methods for reconstructing 3D models from SAXS is imposed by the requirement of solute monodispersity. We present a novel approach that couples low-resolution 3D SAXS reconstruction with composition analysis of mixtures. The approach is applicable to polydisperse and difficult to purify systems, including weakly associated oligomers and transient complexes. Ab initio shape analysis is possible for symmetric homo-oligomers, whereas rigid body modeling is applied also to dissociating complexes when atomic structures of the individual subunits are available. In both approaches, the sample is considered as an equilibrium mixture of intact complexes/oligomers with their dissociation products or free subunits. The algorithms provide the 3D low-resolution model (for ab initio modeling, also the shape of the monomer) and the volume fractions of the bound and free state(s). The simultaneous fitting of multiple scattering data sets collected under different conditions allows one to restrain the modeling further. The possibilities of the approach are illustrated in simulated and experimental SAXS data from protein oligomers and multisubunit complexes including nucleoproteins. Using this approach, new structural insights are provided in the association behavior and conformations of estrogen-related receptors ERRα and ERRγ. The possibility of 3D modeling from the scattering by mixtures significantly widens the range of applicability of SAXS and opens novel avenues in the analysis of oligomeric mixtures and assembly/dissociation processes.
Collapse
Affiliation(s)
- Maxim V Petoukhov
- European Molecular Biology Laboratory, Hamburg Outstation , Notkestrasse 85, Hamburg 22607, Germany
| | | | | | | | | | | |
Collapse
|
1356
|
Møller M, Nielsen SS, Ramachandran S, Li Y, Tria G, Streicher W, Petoukhov MV, Cerione RA, Gillilan RE, Vestergaard B. Small angle X-ray scattering studies of mitochondrial glutaminase C reveal extended flexible regions, and link oligomeric state with enzyme activity. PLoS One 2013; 8:e74783. [PMID: 24098668 PMCID: PMC3787022 DOI: 10.1371/journal.pone.0074783] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Accepted: 08/07/2013] [Indexed: 11/18/2022] Open
Abstract
Glutaminase C is a key metabolic enzyme, which is unregulated in many cancer systems and believed to play a central role in the Warburg effect, whereby cancer cells undergo changes to an altered metabolic profile. A long-standing hypothesis links enzymatic activity to the protein oligomeric state, hence the study of the solution behavior in general and the oligomer state in particular of glutaminase C is important for the understanding of the mechanism of protein activation and inhibition. In this report, this is extensively investigated in correlation to enzyme concentration or phosphate level, using a high-throughput microfluidic-mixing chip for the SAXS data collection, and we confirm that the oligomeric state correlates with activity. The in-depth solution behavior analysis further reveals the structural behavior of flexible regions of the protein in the dimeric, tetrameric and octameric state and investigates the C-terminal influence on the enzyme solution behavior. Our data enable SAXS-based rigid body modeling of the full-length tetramer states, thereby presenting the first ever experimentally derived structural model of mitochondrial glutaminase C including the N- and C-termini of the enzyme.
Collapse
Affiliation(s)
- Magda Møller
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Søren S. Nielsen
- Department of Structural Biophysics, University of Copenhagen, Copenhagen, Denmark
- Cornell High Energy Synchrotron Source (CHESS) and Macromolecular Diffraction Facility at CHESS (MacCHESS), Cornell University, Ithaca, New York, United States of America
| | - Sekar Ramachandran
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York, United States of America
| | - Yuxing Li
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York, United States of America
| | - Giancarlo Tria
- European Molecular Biology Laboratory, Hamburg Outstation c/o DESY, Hamburg, Germany
- Center for Bioinformatics, University of Hamburg, Hamburg, Germany
| | - Werner Streicher
- Protein Function and Interactions, Novo Nordisk Foundation Center for Protein Research, Copenhagen, Denmark
| | - Maxim V. Petoukhov
- European Molecular Biology Laboratory, Hamburg Outstation c/o DESY, Hamburg, Germany
| | - Richard A. Cerione
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York, United States of America
- Department of Molecular Medicine, Cornell University, Ithaca, New York, United States of America
| | - Richard E. Gillilan
- Cornell High Energy Synchrotron Source (CHESS) and Macromolecular Diffraction Facility at CHESS (MacCHESS), Cornell University, Ithaca, New York, United States of America
| | - Bente Vestergaard
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
1357
|
Pauw BR. Everything SAXS: small-angle scattering pattern collection and correction. JOURNAL OF PHYSICS. CONDENSED MATTER : AN INSTITUTE OF PHYSICS JOURNAL 2013; 25:383201. [PMID: 23988669 DOI: 10.1088/0953-8984/25/38/383201] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
For obtaining reliable nanostructural details of large amounts of sample--and if it is applicable--small-angle scattering (SAS) is a prime technique to use. It promises to obtain bulk-scale, statistically sound information on the morphological details of the nanostructure, and has thus led to many a researcher investing their time in it over the last eight decades of development. Due to pressure from scientists requesting more details on increasingly complex nanostructures, as well as the ever improving instrumentation leaving less margin for ambiguity, small-angle scattering methodologies have been evolving at a high pace over the past few decades. As the quality of any results can only be as good as the data that go into these methodologies, the improvements in data collection and all imaginable data correction steps are reviewed here. This work is intended to provide a comprehensive overview of all data corrections, to aid the small-angle scatterer to decide which are relevant for their measurement and how these corrections are performed. Clear mathematical descriptions of the corrections are provided where feasible. Furthermore, as no quality data exist without a decent estimate of their precision, the error estimation and propagation through all these steps are provided alongside the corrections. With these data corrections, the collected small-angle scattering pattern can be made of the highest standard, allowing for authoritative nanostructural characterization through its analysis. A brief background of small-angle scattering, the instrumentation developments over the years, and pitfalls that may be encountered upon data interpretation are provided as well.
Collapse
Affiliation(s)
- Brian Richard Pauw
- International Center for Young Scientists, National Institute for Materials Science, 1-2-1 Sengen, 305-0047, Tsukuba, Japan.
| |
Collapse
|
1358
|
Sander B, Tria G, Shkumatov AV, Kim EY, Grossmann JG, Tessmer I, Svergun DI, Schindelin H. Structural characterization of gephyrin by AFM and SAXS reveals a mixture of compact and extended states. ACTA CRYSTALLOGRAPHICA SECTION D: BIOLOGICAL CRYSTALLOGRAPHY 2013; 69:2050-60. [DOI: 10.1107/s0907444913018714] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Accepted: 07/05/2013] [Indexed: 11/10/2022]
Abstract
Gephyrin is a trimeric protein involved in the final steps of molybdenum-cofactor (Moco) biosynthesis and in the clustering of inhibitory glycine and GABAAreceptors at postsynaptic specializations. Each protomer consists of stably folded domains (referred to as the G and E domains) located at either terminus and connected by a proteolytically sensitive linker of ∼150 residues. Both terminal domains can oligomerize in their isolated forms; however, in the context of the full-length protein only the G-domain trimer is permanently present, whereas E-domain dimerization is prevented. Atomic force microscopy (AFM) and small-angle X-ray scattering (SAXS) reveal a high degree of flexibility in the structure of gephyrin. The results imply an equilibrium between compact and extended conformational states in solution, with a preference for compact states. CD spectroscopy suggests that a partial compaction is achieved by interactions of the linker with the G and E domains. Taken together, the data provide a rationale for the role of the linker in the overall structure and the conformational dynamics of gephyrin.
Collapse
|
1359
|
Cerofolini L, Fields GB, Fragai M, Geraldes CFGC, Luchinat C, Parigi G, Ravera E, Svergun DI, Teixeira JMC. Examination of matrix metalloproteinase-1 in solution: a preference for the pre-collagenolysis state. J Biol Chem 2013; 288:30659-30671. [PMID: 24025334 DOI: 10.1074/jbc.m113.477240] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Catalysis of collagen degradation by matrix metalloproteinase 1 (MMP-1) has been proposed to critically rely on flexibility between the catalytic (CAT) and hemopexin-like (HPX) domains. A rigorous assessment of the most readily accessed conformations in solution is required to explain the onset of substrate recognition and collagenolysis. The present study utilized paramagnetic NMR spectroscopy and small angle x-ray scattering (SAXS) to calculate the maximum occurrence (MO) of MMP-1 conformations. The MMP-1 conformations with large MO values (up to 47%) are restricted into a relatively small conformational region. All conformations with high MO values differ largely from the closed MMP-1 structures obtained by x-ray crystallography. The MO of the latter is ~20%, which represents the upper limit for the presence of this conformation in the ensemble sampled by the protein in solution. In all the high MO conformations, the CAT and HPX domains are not in tight contact, and the residues of the HPX domain reported to be responsible for the binding to the collagen triple-helix are solvent exposed. Thus, overall analysis of the highest MO conformations indicated that MMP-1 in solution was poised to interact with collagen and then could readily proceed along the steps of collagenolysis.
Collapse
Affiliation(s)
| | - Gregg B Fields
- the Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida 34987,.
| | - Marco Fragai
- From the CERM and; the Department of Chemistry "U. Schiff," University of Florence, Via Luigi Sacconi 6, 50019 Sesto Fiorentino (FI), Italy
| | - Carlos F G C Geraldes
- the Center for Neuroscience and Cell Biology and; the Department of Life Sciences, Faculty of Science and Technology, University of Coimbra, P.O. Box 3046, 3001-401 Coimbra, Portugal, and
| | - Claudio Luchinat
- From the CERM and; the Department of Chemistry "U. Schiff," University of Florence, Via Luigi Sacconi 6, 50019 Sesto Fiorentino (FI), Italy,.
| | - Giacomo Parigi
- From the CERM and; the Department of Chemistry "U. Schiff," University of Florence, Via Luigi Sacconi 6, 50019 Sesto Fiorentino (FI), Italy
| | - Enrico Ravera
- From the CERM and; the Department of Chemistry "U. Schiff," University of Florence, Via Luigi Sacconi 6, 50019 Sesto Fiorentino (FI), Italy
| | - Dmitri I Svergun
- the EMBL, c/o DESY, Notkestrasse 85, Geb. 25 A, 22603 Hamburg, Germany
| | - João M C Teixeira
- From the CERM and; the Center for Neuroscience and Cell Biology and; the Department of Life Sciences, Faculty of Science and Technology, University of Coimbra, P.O. Box 3046, 3001-401 Coimbra, Portugal, and
| |
Collapse
|
1360
|
Koch C, Tria G, Fielding AJ, Brodhun F, Valerius O, Feussner K, Braus GH, Svergun DI, Bennati M, Feussner I. A structural model of PpoA derived from SAXS-analysis—Implications for substrate conversion. Biochim Biophys Acta Mol Cell Biol Lipids 2013; 1831:1449-57. [DOI: 10.1016/j.bbalip.2013.06.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Revised: 05/29/2013] [Accepted: 06/07/2013] [Indexed: 10/26/2022]
|
1361
|
Jain R, Petri M, Kirschbaum S, Feindt H, Steltenkamp S, Sonnenkalb S, Becker S, Griesinger C, Menzel A, Burg TP, Techert S. X-ray scattering experiments with high-flux X-ray source coupled rapid mixing microchannel device and their potential for high-flux neutron scattering investigations. THE EUROPEAN PHYSICAL JOURNAL. E, SOFT MATTER 2013; 36:109. [PMID: 24092048 DOI: 10.1140/epje/i2013-13109-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Revised: 12/13/2012] [Accepted: 07/26/2013] [Indexed: 06/02/2023]
Abstract
Small-angle X-ray scattering provides global, shape-sensitive structural information about macromolecules in solution. Its extension to time dimension in the form of time-resolved SAXS investigations and combination with other time-resolved biophysical methods contributes immensely to the study of protein dynamics. TR-SAXS can also provide unique information about the global structures of transient intermediates during protein dynamics. An experimental set-up with low protein consumption is essential for an extensive use of TR-SAXS experiments on protein dynamics. In this direction, a newly developed 20-microchannel microfluidic continuous-flow mixer was combined with SAXS. With this set-up, we demonstrate ubiquitin unfolding dynamics after rapid mixing with the chaotropic agent Guanidinium-HCl within milliseconds using only ∼ 40 nanoliters of the protein sample per scattering image. It is suggested that, in the future, this new TR-SAXS platform will help to increase the use of time-resolved small-angle X-ray scattering, wide-angle X-ray scattering and neutron scattering experiments for studying protein dynamics in the early millisecond regime. The potential research field for this set-up includes protein folding, protein misfolding, aggregation in amyloidogenic diseases, function of intrinsically disordered proteins and various protein-ligand interactions.
Collapse
Affiliation(s)
- R Jain
- Structural Dynamics of (Bio)chemical Systems, MPI-BPC, Am Fassberg 11, 37077, Goettingen, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1362
|
Pham CLL, Kirby N, Wood K, Ryan T, Roberts B, Sokolova A, Barnham KJ, Masters CL, Knott RB, Cappai R, Curtain CC, Rekas A. Guanidine hydrochloride denaturation of dopamine-induced α-synuclein oligomers: A small-angle X-ray scattering study. Proteins 2013; 82:10-21. [DOI: 10.1002/prot.24332] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Revised: 05/10/2013] [Accepted: 05/19/2013] [Indexed: 01/18/2023]
Affiliation(s)
- Chi L. L. Pham
- Department of Pathology and Bio21 Molecular Science and Technology Institute; The University of Melbourne; Victoria 3010 Australia
| | - Nigel Kirby
- SAXS/WAXS Beamline, The Australian Synchrotron; Clayton Victoria 3168 Australia
| | - Kathleen Wood
- Australian Nuclear Science and Technology Organisation (ANSTO); Kirrawee New South Wales 2232 Australia
| | - Timothy Ryan
- The University of Melbourne, Florey Institute of Neuroscience and Mental Health; Victoria 3010 Australia
| | - Blaine Roberts
- The University of Melbourne, Florey Institute of Neuroscience and Mental Health; Victoria 3010 Australia
| | - Anna Sokolova
- Australian Nuclear Science and Technology Organisation (ANSTO); Kirrawee New South Wales 2232 Australia
| | - Kevin J. Barnham
- The University of Melbourne, Florey Institute of Neuroscience and Mental Health; Victoria 3010 Australia
| | - Colin L. Masters
- The University of Melbourne, Florey Institute of Neuroscience and Mental Health; Victoria 3010 Australia
| | - Robert B. Knott
- Australian Nuclear Science and Technology Organisation (ANSTO); Kirrawee New South Wales 2232 Australia
| | - Roberto Cappai
- Department of Pathology and Bio21 Molecular Science and Technology Institute; The University of Melbourne; Victoria 3010 Australia
| | - Cyril C. Curtain
- Department of Pathology and Bio21 Molecular Science and Technology Institute; The University of Melbourne; Victoria 3010 Australia
- The University of Melbourne, Florey Institute of Neuroscience and Mental Health; Victoria 3010 Australia
| | - Agata Rekas
- Australian Nuclear Science and Technology Organisation (ANSTO); Kirrawee New South Wales 2232 Australia
| |
Collapse
|
1363
|
Yan R, Konarev PV, Iannuzzi C, Adinolfi S, Roche B, Kelly G, Simon L, Martin SR, Py B, Barras F, Svergun DI, Pastore A. Ferredoxin competes with bacterial frataxin in binding to the desulfurase IscS. J Biol Chem 2013; 288:24777-87. [PMID: 23839945 PMCID: PMC3750173 DOI: 10.1074/jbc.m113.480327] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Revised: 07/03/2013] [Indexed: 11/06/2022] Open
Abstract
The bacterial iron-sulfur cluster (isc) operon is an essential machine that is highly conserved from bacteria to primates and responsible for iron-sulfur cluster biogenesis. Among its components are the genes for the desulfurase IscS that provides sulfur for cluster formation, and a specialized ferredoxin (Fdx) whose role is still unknown. Preliminary evidence suggests that IscS and Fdx interact but nothing is known about the binding site and the role of the interaction. Here, we have characterized the interaction using a combination of biophysical tools and mutagenesis. By modeling the Fdx·IscS complex based on experimental restraints we show that Fdx competes for the binding site of CyaY, the bacterial ortholog of frataxin and sits in a cavity close to the enzyme active site. By in vivo mutagenesis in bacteria we prove the importance of the surface of interaction for cluster formation. Our data provide the first structural insights into the role of Fdx in cluster assembly.
Collapse
Affiliation(s)
- Robert Yan
- From the MRC National Institute for Medical Research, The Ridgeway, London NW7 1AA, United Kingdom
| | - Petr V. Konarev
- the European Molecular Biology Laboratory, EMBL c/o DESY, Notkestrasse 85, Hamburg D-22603, Germany, and
| | - Clara Iannuzzi
- From the MRC National Institute for Medical Research, The Ridgeway, London NW7 1AA, United Kingdom
| | - Salvatore Adinolfi
- From the MRC National Institute for Medical Research, The Ridgeway, London NW7 1AA, United Kingdom
| | | | - Geoff Kelly
- From the MRC National Institute for Medical Research, The Ridgeway, London NW7 1AA, United Kingdom
| | - Léa Simon
- From the MRC National Institute for Medical Research, The Ridgeway, London NW7 1AA, United Kingdom
| | - Stephen R. Martin
- From the MRC National Institute for Medical Research, The Ridgeway, London NW7 1AA, United Kingdom
| | - Béatrice Py
- the Aix-Marseille Université and
- Laboratoire de Chimie Bactérienne, Institut de Microbiologie de la Méditerranée, UMR 7283, CNRS, 31 Chemin Joseph Aiguier, 13009 Marseille, France
| | - Frédéric Barras
- the Aix-Marseille Université and
- Laboratoire de Chimie Bactérienne, Institut de Microbiologie de la Méditerranée, UMR 7283, CNRS, 31 Chemin Joseph Aiguier, 13009 Marseille, France
| | - Dmitri I. Svergun
- the European Molecular Biology Laboratory, EMBL c/o DESY, Notkestrasse 85, Hamburg D-22603, Germany, and
| | - Annalisa Pastore
- From the MRC National Institute for Medical Research, The Ridgeway, London NW7 1AA, United Kingdom
| |
Collapse
|
1364
|
Structure-informed design of an enzymatically inactive vaccine component for group A Streptococcus. mBio 2013; 4:mBio.00509-13. [PMID: 23919999 PMCID: PMC3735194 DOI: 10.1128/mbio.00509-13] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Streptococcus pyogenes (group A Streptococcus [GAS]) causes ~700 million human infections/year, resulting in >500,000 deaths. There is no commercial GAS vaccine available. The GAS surface protein arginine deiminase (ADI) protects mice against a lethal challenge. ADI is an enzyme that converts arginine to citrulline and ammonia. Administration of a GAS vaccine preparation containing wild-type ADI, a protein with inherent enzymatic activity, may present a safety risk. In an approach intended to maximize the vaccine safety of GAS ADI, X-ray crystallography and structural immunogenic epitope mapping were used to inform vaccine design. This study aimed to knock out ADI enzyme activity without disrupting the three-dimensional structure or the recognition of immunogenic epitopes. We determined the crystal structure of ADI at 2.5 Å resolution and used it to select a number of amino acid residues for mutagenesis to alanine (D166, E220, H275, D277, and C401). Each mutant protein displayed abrogated activity, and three of the mutant proteins (those with the D166A, H275A, and D277A mutations) possessed a secondary structure and oligomerization state equivalent to those of the wild type, produced high-titer antisera, and avoided disruption of B-cell epitopes of ADI. In addition, antisera raised against the D166A and D277A mutant proteins bound to the GAS cell surface. The inactivated D166A and D277A mutant ADIs are ideal for inclusion in a GAS vaccine preparation. There is no human ortholog of ADI, and we confirm that despite limited structural similarity in the active-site region to human peptidyl ADI 4 (PAD4), ADI does not functionally mimic PAD4 and antiserum raised against GAS ADI does not recognize human PAD4. We present an example of structural biology informing human vaccine design. We previously showed that the administration of the enzyme arginine deiminase (ADI) to mice protected the mice against infection with multiple GAS serotypes. In this study, we determined the structure of GAS ADI and used this information to improve the vaccine safety of GAS ADI. Catalytically inactive mutant forms of ADI retained structure, recognition by antisera, and immunogenic epitopes, rendering them ideal for inclusion in GAS vaccine preparations. This example of structural biology informing vaccine design may underpin the formulation of a safe and efficacious GAS vaccine.
Collapse
|
1365
|
Dian C, Bernaudat F, Langer K, Oliva MF, Fornerod M, Schoehn G, Müller CW, Petosa C. Structure of a truncation mutant of the nuclear export factor CRM1 provides insights into the auto-inhibitory role of its C-terminal helix. Structure 2013; 21:1338-49. [PMID: 23850454 DOI: 10.1016/j.str.2013.06.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2013] [Revised: 06/12/2013] [Accepted: 06/13/2013] [Indexed: 11/23/2022]
Abstract
Chromosome region maintenance 1/exportin1/Xpo1 (CRM1) associates with the GTPase Ran to mediate the nuclear export of proteins bearing a leucine-rich nuclear export signal (NES). CRM1 consists of helical hairpin HEAT repeats and a C-terminal helical extension (C-extension) that inhibits the binding of NES-bearing cargos. We report the crystal structure and small-angle X-ray scattering analysis of a human CRM1 mutant with enhanced NES-binding activity due to deletion of the C-extension. We show that loss of the C-extension leads to a repositioning of CRM1's C-terminal repeats and to a more extended overall conformation. Normal mode analysis predicts reduced rigidity for the deletion mutant, consistent with an observed decrease in thermal stability. Point mutations that destabilize the C-extension shift CRM1 to the more extended conformation, reduce thermal stability, and enhance NES-binding activity. These findings suggest that an important mechanism by which the C-extension regulates CRM1's cargo-binding affinity is by modulating the conformation and flexibility of its HEAT repeats.
Collapse
Affiliation(s)
- Cyril Dian
- Université de Grenoble Alpes, Institut de Biologie Structurale, 38027 Grenoble, France
| | | | | | | | | | | | | | | |
Collapse
|
1366
|
Modern X-ray scattering studies of complex biological systems. Curr Opin Biotechnol 2013; 24:716-23. [DOI: 10.1016/j.copbio.2013.01.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Revised: 01/07/2013] [Accepted: 01/08/2013] [Indexed: 11/16/2022]
|
1367
|
Taltynov O, Demeulemeester J, Christ F, De Houwer S, Tsirkone VG, Gerard M, Weeks SD, Strelkov SV, Debyser Z. Interaction of transportin-SR2 with Ras-related nuclear protein (Ran) GTPase. J Biol Chem 2013; 288:25603-25613. [PMID: 23878195 DOI: 10.1074/jbc.m113.484345] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The human immunodeficiency virus type 1 (HIV-1) and other lentiviruses are capable of infecting non-dividing cells and, therefore, need to be imported into the nucleus before integration into the host cell chromatin. Transportin-SR2 (TRN-SR2, Transportin-3, TNPO3) is a cellular karyopherin implicated in nuclear import of HIV-1. A model in which TRN-SR2 imports the viral preintegration complex into the nucleus is supported by direct interaction between TRN-SR2 and HIV-1 integrase (IN). Residues in the C-terminal domain of HIV-1 IN that mediate binding to TRN-SR2 were recently delineated. As for most nuclear import cargoes, the driving force behind HIV-1 preintegration complex import is likely a gradient of the GDP- and GTP-bound forms of Ran, a small GTPase. In this study we offer biochemical and structural characterization of the interaction between TRN-SR2 and Ran. By size exclusion chromatography we demonstrate stable complex formation of TRN-SR2 and RanGTP in solution. Consistent with the behavior of normal nuclear import cargoes, HIV-1 IN is released from the complex with TRN-SR2 by RanGTP. Although in concentrated solutions TRN-SR2 by itself was predominantly present as a dimer, the TRN-SR2-RanGTP complex was significantly more compact. Further analysis supported a model wherein one monomer of TRN-SR2 is bound to one monomer of RanGTP. Finally, we present a homology model of the TRN-SR2-RanGTP complex that is in excellent agreement with the experimental small angle x-ray scattering data.
Collapse
Affiliation(s)
- Oliver Taltynov
- From the Laboratory for Molecular Virology and Gene Therapy and
| | | | - Frauke Christ
- From the Laboratory for Molecular Virology and Gene Therapy and
| | | | - Vicky G Tsirkone
- Laboratory for Biocrystallography, KU Leuven, B-3000 Leuven, Belgium
| | - Melanie Gerard
- From the Laboratory for Molecular Virology and Gene Therapy and
| | - Stephen D Weeks
- Laboratory for Biocrystallography, KU Leuven, B-3000 Leuven, Belgium
| | - Sergei V Strelkov
- Laboratory for Biocrystallography, KU Leuven, B-3000 Leuven, Belgium
| | - Zeger Debyser
- From the Laboratory for Molecular Virology and Gene Therapy and.
| |
Collapse
|
1368
|
Keown JR, Griffin MDW, Mertens HDT, Pearce FG. Small oligomers of ribulose-bisphosphate carboxylase/oxygenase (Rubisco) activase are required for biological activity. J Biol Chem 2013; 288:20607-15. [PMID: 23720775 PMCID: PMC3711324 DOI: 10.1074/jbc.m113.466383] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Revised: 05/27/2013] [Indexed: 12/24/2022] Open
Abstract
Ribulose-bisphosphate carboxylase/oxygenase (Rubisco) activase uses the energy from ATP hydrolysis to remove tight binding inhibitors from Rubisco, thus playing a key role in regulating photosynthesis in plants. Although several structures have recently added much needed structural information for different Rubisco activase enzymes, the arrangement of these subunits in solution remains unclear. In this study, we use a variety of techniques to show that Rubisco activase forms a wide range of structures in solution, ranging from monomers to much higher order species, and that the distribution of these species is highly dependent on protein concentration. The data support a model in which Rubisco activase forms an open spiraling structure rather than a closed hexameric structure. At protein concentrations of 1 μM, corresponding to the maximal activity of the enzyme, Rubisco activase has an oligomeric state of 2-4 subunits. We propose a model in which Rubisco activase requires at least 1 neighboring subunit for hydrolysis of ATP.
Collapse
Affiliation(s)
- Jeremy R. Keown
- From the Biomolecular Interactions Centre and School of Biological Sciences, University of Canterbury, Private Bag 4800, Christchurch 8140, New Zealand
| | - Michael D. W. Griffin
- the Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville Victoria 3010, Australia, and
| | - Haydyn D. T. Mertens
- the Australian Synchrotron, 800 Blackburn Road, Clayton Victoria 3168, Australia
| | - F. Grant Pearce
- From the Biomolecular Interactions Centre and School of Biological Sciences, University of Canterbury, Private Bag 4800, Christchurch 8140, New Zealand
| |
Collapse
|
1369
|
Graewert MA, Svergun DI. Impact and progress in small and wide angle X-ray scattering (SAXS and WAXS). Curr Opin Struct Biol 2013; 23:748-54. [PMID: 23835228 DOI: 10.1016/j.sbi.2013.06.007] [Citation(s) in RCA: 128] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Accepted: 06/12/2013] [Indexed: 10/26/2022]
Abstract
The advances made in small and wide angle X-ray scattering over the past decades have had a large impact on structural biology. Many new insights into challenging biological probes including large and transient complexes, flexible macromolecules as well as other exciting objects of various sizes were gained with this low resolution technique. Here, we review the recent developments in the experimental setups and in software for data collection and analysis, specifically for hybrid approaches. These progresses have allowed scientists to address a number of intriguing questions which could not be answered with other structural methods alone.
Collapse
Affiliation(s)
- Melissa A Graewert
- European Molecular Biology Laboratory, Hamburg Unit, EMBL c/o DESY, Notkestraße 85, Hamburg 22603, Germany
| | | |
Collapse
|
1370
|
Samygina VR, Sokolov AV, Bourenkov G, Petoukhov MV, Pulina MO, Zakharova ET, Vasilyev VB, Bartunik H, Svergun DI. Ceruloplasmin: macromolecular assemblies with iron-containing acute phase proteins. PLoS One 2013; 8:e67145. [PMID: 23843990 PMCID: PMC3700992 DOI: 10.1371/journal.pone.0067145] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Accepted: 05/15/2013] [Indexed: 11/19/2022] Open
Abstract
Copper-containing ferroxidase ceruloplasmin (Cp) forms binary and ternary complexes with cationic proteins lactoferrin (Lf) and myeloperoxidase (Mpo) during inflammation. We present an X-ray crystal structure of a 2Cp-Mpo complex at 4.7 Å resolution. This structure allows one to identify major protein-protein interaction areas and provides an explanation for a competitive inhibition of Mpo by Cp and for the activation of p-phenylenediamine oxidation by Mpo. Small angle X-ray scattering was employed to construct low-resolution models of the Cp-Lf complex and, for the first time, of the ternary 2Cp-2Lf-Mpo complex in solution. The SAXS-based model of Cp-Lf supports the predicted 1:1 stoichiometry of the complex and demonstrates that both lobes of Lf contact domains 1 and 6 of Cp. The 2Cp-2Lf-Mpo SAXS model reveals the absence of interaction between Mpo and Lf in the ternary complex, so Cp can serve as a mediator of protein interactions in complex architecture. Mpo protects antioxidant properties of Cp by isolating its sensitive loop from proteases. The latter is important for incorporation of Fe(3+) into Lf, which activates ferroxidase activity of Cp and precludes oxidation of Cp substrates. Our models provide the structural basis for possible regulatory role of these complexes in preventing iron-induced oxidative damage.
Collapse
Affiliation(s)
- Valeriya R. Samygina
- Institute of Crystallography RAS, Moscow, Russia
- Structural Biology Unit, CICbioGUNE, Derio, Spain
| | | | | | | | - Maria O. Pulina
- Institute of Experimental Medicine NWB RAMS, St.Petersburg, Russia
| | | | | | - Hans Bartunik
- Research Unit for Structural Molecular Biology, Max-Planck Institute, Hamburg, Germany
| | | |
Collapse
|
1371
|
Myllykoski M, Raasakka A, Lehtimäki M, Han H, Kursula I, Kursula P. Crystallographic analysis of the reaction cycle of 2',3'-cyclic nucleotide 3'-phosphodiesterase, a unique member of the 2H phosphoesterase family. J Mol Biol 2013; 425:4307-22. [PMID: 23831225 PMCID: PMC7094350 DOI: 10.1016/j.jmb.2013.06.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Revised: 06/12/2013] [Accepted: 06/13/2013] [Indexed: 11/26/2022]
Abstract
2H phosphoesterases catalyze reactions on nucleotide substrates and contain two conserved histidine residues in the active site. Very limited information is currently available on the details of the active site and substrate/product binding during the catalytic cycle of these enzymes. We performed a comprehensive X-ray crystallographic study of mouse 2′,3′-cyclic nucleotide 3′-phosphodiesterase (CNPase), a membrane-associated enzyme present at high levels in the tetrapod myelin sheath. We determined crystal structures of the CNPase phosphodiesterase domain complexed with substrate, product, and phosphorothioate analogues. The data provide detailed information on the CNPase reaction mechanism, including substrate binding mode and coordination of the nucleophilic water molecule. Linked to the reaction, an open/close motion of the β5–α7 loop is observed. The role of the N terminus of helix α7—unique for CNPase in the 2H family—during the reaction indicates that 2H phosphoesterases differ in their respective reaction mechanisms despite the conserved catalytic residues. Furthermore, based on small-angle X-ray scattering, we present a model for the full-length enzyme, indicating that the two domains of CNPase form an elongated molecule. Finally, based on our structural data and a comprehensive bioinformatics study, we discuss the conservation of CNPase in various organisms. A detailed structural analysis of the CNPase catalytic cycle was carried out. Complexes with substrates, products, and analogues highlight roles for a nearby helix and loop in the reaction mechanism. The full-length CNPase adopts an elongated conformation in solution. CNPase is a unique member of the 2H family, and the results will help understand its physiological significance.
Collapse
Affiliation(s)
- Matti Myllykoski
- Department of Biochemistry, University of Oulu, FIN-90014 Oulu, Finland; Biocenter Oulu, University of Oulu, FIN-90014 Oulu, Finland
| | | | | | | | | | | |
Collapse
|
1372
|
Groothuizen FS, Fish A, Petoukhov MV, Reumer A, Manelyte L, Winterwerp HHK, Marinus MG, Lebbink JHG, Svergun DI, Friedhoff P, Sixma TK. Using stable MutS dimers and tetramers to quantitatively analyze DNA mismatch recognition and sliding clamp formation. Nucleic Acids Res 2013; 41:8166-81. [PMID: 23821665 PMCID: PMC3783165 DOI: 10.1093/nar/gkt582] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The process of DNA mismatch repair is initiated when MutS recognizes mismatched DNA bases and starts the repair cascade. The Escherichia coli MutS protein exists in an equilibrium between dimers and tetramers, which has compromised biophysical analysis. To uncouple these states, we have generated stable dimers and tetramers, respectively. These proteins allowed kinetic analysis of DNA recognition and structural analysis of the full-length protein by X-ray crystallography and small angle X-ray scattering. Our structural data reveal that the tetramerization domains are flexible with respect to the body of the protein, resulting in mostly extended structures. Tetrameric MutS has a slow dissociation from DNA, which can be due to occasional bending over and binding DNA in its two binding sites. In contrast, the dimer dissociation is faster, primarily dependent on a combination of the type of mismatch and the flanking sequence. In the presence of ATP, we could distinguish two kinetic groups: DNA sequences where MutS forms sliding clamps and those where sliding clamps are not formed efficiently. Interestingly, this inability to undergo a conformational change rather than mismatch affinity is correlated with mismatch repair.
Collapse
Affiliation(s)
- Flora S Groothuizen
- Division of Biochemistry and CancerGenomiCs.nl, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands, European Molecular Biology Laboratory, Hamburg Outstation, EMBL c/o DESY, Notkestrasse 85, 22607 Hamburg, Germany, Institute for Biochemistry, Justus Liebig University, Heinrich-Buff Ring 58, D-35392, Giessen, Germany, Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, 364 Plantation Street, Worcester, MA 01605, USA, Department of Cell Biology and Genetics, Cancer Genomics Center, Erasmus Medical Center, PO Box 2040, 3000 CA Rotterdam, the Netherlands and Department of Radiation Oncology, Erasmus Medical Center, PO Box 2040, 3000 CA Rotterdam, the Netherlands
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1373
|
Seraphim TV, Alves MM, Silva IM, Gomes FER, Silva KP, Murta SMF, Barbosa LRS, Borges JC. Low resolution structural studies indicate that the activator of Hsp90 ATPase 1 (Aha1) of Leishmania braziliensis has an elongated shape which allows its interaction with both N- and M-domains of Hsp90. PLoS One 2013; 8:e66822. [PMID: 23826147 PMCID: PMC3691308 DOI: 10.1371/journal.pone.0066822] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Accepted: 05/13/2013] [Indexed: 11/18/2022] Open
Abstract
The Hsp90 molecular chaperone is essential for protein homeostasis and in the maturation of proteins involved with cell-cycle control. The low ATPase activity of Hsp90 is critical to drive its functional cycle, which is dependent on the Hsp90 cochaperones. The Activator of Hsp90 ATPase-1 (Aha1) is a protein formed by two domains, N- and C-terminal, that stimulates the Hsp90 ATPase activity by several folds. Although the relevance of Aha1 for Hsp90 functions has been proved, as well as its involvement in the desensitization to inhibitors of the Hsp90, the knowledge on its overall structure and behavior in solution is limited. In this work we present the functional and structural characterization of Leishmania braziliensis Aha1 (LbAha1). This protozoan is the causative agent of cutaneous and mucocutaneous leishmaniasis, a neglected disease. The recombinant LbAha1 behaves as an elongated monomer and is organized into two folded domains interconnected by a flexible linker. Functional experiments showed that LbAha1 interacts with L. braziliensis Hsp90 (LbHsp90) with micromolar dissociation constant in a stoichiometry of 2 LbAha1 to 1 LbHsp90 dimer and stimulates 10-fold the LbHsp90 ATPase activity showing positive cooperativity. Furthermore, the LbHsp90::LbAha1 complex is directed by enthalphy and opposed by entropy, probably due to the spatial freedom restrictions imposed by the proteins' interactions. Small-angle X-ray scattering data allowed the reconstruction of low resolution models and rigid body simulations of LbAha1, indicating its mode of action on LbHsp90. Western blot experiments allowed Aha1 identification (as well as Hsp90) in three Leishmania species at two temperatures, suggesting that Aha1 is a cognate protein. All these data shed light on the LbAha1 mechanism of action, showing that it has structural dimensions and flexibility that allow interacting with both N-terminal and middle domains of the LbHsp90.
Collapse
Affiliation(s)
- Thiago V. Seraphim
- Instituto de Química de São Carlos, Universidade de São Paulo - USP, São Carlos, SP, Brazil
| | - Marina M. Alves
- Instituto de Química de São Carlos, Universidade de São Paulo - USP, São Carlos, SP, Brazil
- Centro de Ciências Biológicas e da Saúde, Universidade Federal de São Carlos, São Carlos, SP, Brazil
| | - Indjara M. Silva
- Instituto de Química de São Carlos, Universidade de São Paulo - USP, São Carlos, SP, Brazil
| | - Francisco E. R. Gomes
- Instituto de Química de São Carlos, Universidade de São Paulo - USP, São Carlos, SP, Brazil
| | - Kelly P. Silva
- Instituto de Química de São Carlos, Universidade de São Paulo - USP, São Carlos, SP, Brazil
| | | | - Leandro R. S. Barbosa
- Departamento de Física Geral, Instituto de Física, Universidade de São Paulo - USP, São Paulo, SP, Brazil
| | - Júlio C. Borges
- Instituto de Química de São Carlos, Universidade de São Paulo - USP, São Carlos, SP, Brazil
| |
Collapse
|
1374
|
Majumder A, Gopalakrishna KN, Cheguru P, Gakhar L, Artemyev NO. Interaction of aryl hydrocarbon receptor-interacting protein-like 1 with the farnesyl moiety. J Biol Chem 2013; 288:21320-21328. [PMID: 23737531 DOI: 10.1074/jbc.m113.476242] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Aryl hydrocarbon receptor-interacting protein-like 1 (AIPL1) is a photoreceptor specific chaperone of the visual effector enzyme phosphodiesterase-6 (PDE6). AIPL1 has been shown to bind the farnesylated PDE6A subunit. Mutations in AIPL1 are thought to destabilize PDE6 and thereby cause Leber congenital amaurosis type 4 (LCA4), a severe form of childhood blindness. Here, we examined the solution structure of AIPL1 by small angle x-ray scattering. A structural model of AIPL1 with the best fit to the scattering data features two independent FK506-binding protein (FKBP)-like and tetratricopeptide repeat domains. Guided by the model, we tested the hypothesis that AIPL1 directly binds the farnesyl moiety. Our studies revealed high affinity binding of the farnesylated-Cys probe to the FKBP-like domain of AIPL1, thus uncovering a novel function of this domain. Mutational analysis of the potential farnesyl-binding sites on AIPL1 identified two critical residues, Cys-89 and Leu-147, located in close proximity in the structure model. The L147A mutation and the LCA-linked C89R mutation prevented the binding of the farnesyl-Cys probe to AIPL1. Furthermore, Cys-89 and Leu-147 flank the unique insert region of AIPL1, deletion of which also abolished the farnesyl interaction. Our results suggest that the binding of PDE6A farnesyl is essential to normal function of AIPL1 and its disruption is one of the mechanisms underlying LCA.
Collapse
Affiliation(s)
| | | | | | - Lokesh Gakhar
- Department of Biochemistry,; Protein Crystallography Facility, and
| | - Nikolai O Artemyev
- From the Department of Molecular Physiology and Biophysics,; Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, Iowa 52242.
| |
Collapse
|
1375
|
A Two-State Cooperative Expansion Converts the Procapsid Shell of Bacteriophage T5 into a Highly Stable Capsid Isomorphous to the Final Virion Head. J Mol Biol 2013; 425:1999-2014. [DOI: 10.1016/j.jmb.2013.03.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2012] [Revised: 02/20/2013] [Accepted: 03/03/2013] [Indexed: 11/19/2022]
|
1376
|
Ryan TM, Caine J, Mertens HDT, Kirby N, Nigro J, Breheney K, Waddington LJ, Streltsov VA, Curtain C, Masters CL, Roberts BR. Ammonium hydroxide treatment of Aβ produces an aggregate free solution suitable for biophysical and cell culture characterization. PeerJ 2013; 1:e73. [PMID: 23678397 PMCID: PMC3646356 DOI: 10.7717/peerj.73] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2012] [Accepted: 04/22/2013] [Indexed: 11/20/2022] Open
Abstract
Alzheimer's disease is the leading cause of dementia in the elderly. Pathologically it is characterized by the presence of amyloid plaques and neuronal loss within the brain tissue of affected individuals. It is now widely hypothesised that fibrillar structures represent an inert structure. Biophysical and toxicity assays attempting to characterize the formation of both the fibrillar and the intermediate oligomeric structures of Aβ typically involves preparing samples which are largely monomeric; the most common method by which this is achieved is to use the fluorinated organic solvent 1,1,1,3,3,3-hexafluoro-2-propanol (HFIP). Recent evidence has suggested that this method is not 100% effective in producing an aggregate free solution. We show, using dynamic light scattering, size exclusion chromatography and small angle X-ray scattering that this is indeed the case, with HFIP pretreated Aβ peptide solutions displaying an increased proportion of oligomeric and aggregated material and an increased propensity to aggregate. Furthermore we show that an alternative technique, involving treatment with strong alkali results in a much more homogenous solution that is largely monomeric. These techniques for solubilising and controlling the oligomeric state of Aβ are valuable starting points for future biophysical and toxicity assays.
Collapse
Affiliation(s)
- Timothy M Ryan
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne , Parkville, Victoria , Australia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1377
|
Simultaneous binding of the anti-cancer IgM monoclonal antibody PAT-SM6 to low density lipoproteins and GRP78. PLoS One 2013; 8:e61239. [PMID: 23620733 PMCID: PMC3631193 DOI: 10.1371/journal.pone.0061239] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2013] [Accepted: 03/06/2013] [Indexed: 11/30/2022] Open
Abstract
The tumour-derived monoclonal IgM antibody PAT-SM6 specifically kills malignant cells by an apoptotic mechanism linked to the excessive uptake of plasma lipids. The mechanism is postulated to occur via the multi-point attachment of PAT-SM6 to the unfolded protein response regulator GRP78, located on the surface of tumour cells, coupled to the simultaneous binding of plasma low density lipoprotein (LDL). We prepared and characterised LDL and oxidized LDL using sedimentation velocity and small-angle X-ray scattering (SAXS) analysis. Enzyme-linked immunosorbent (ELISA) techniques indicated apparent dissociation constants of approximately 20 nM for the binding of LDL or oxidized LDL to PAT-SM6. ELISA experiments showed cross competition with LDL inhibiting PAT-SM6 binding to immobilised GRP78, while, in the reverse experiment, GRP78 inhibited PAT-SM6 binding to immobilized LDL. In contrast to the results of the ELISA experiments, sedimentation velocity experiments indicated relatively weak interactions between LDL and PAT-SM6, suggesting immunoabsorbance to the microtiter plate is driven by an avidity-based binding mechanism. The importance of avidity and the multipoint attachment of antigens to PAT-SM6 was further investigated using antigen-coated polystyrene beads. Absorption of GRP78 or LDL to polystyrene microspheres led to an increase in the inhibition of PAT-SM6 binding to microtiter plates coated with GRP78 or LDL, respectively. These results support the hypothesis that the biological action of PAT-SM6 in tumour cell apoptosis depends on the multivalent nature of PAT-SM6 and the ability to interact simultaneously with LDL and multiple GRP78 molecules clustered on the tumour cell surface.
Collapse
|
1378
|
Karaca E, Bonvin AMJJ. On the usefulness of ion-mobility mass spectrometry and SAXS data in scoring docking decoys. ACTA CRYSTALLOGRAPHICA SECTION D: BIOLOGICAL CRYSTALLOGRAPHY 2013; 69:683-94. [DOI: 10.1107/s0907444913007063] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Accepted: 03/13/2013] [Indexed: 12/20/2022]
|
1379
|
Kettner K, Kuettner EB, Otto A, Lilie H, Golbik RP, Sträter N, Kriegel TM. In vivo phosphorylation and in vitro autophosphorylation-inactivation of Kluyveromyces lactis hexokinase KlHxk1. Biochem Biophys Res Commun 2013; 435:313-8. [PMID: 23583397 DOI: 10.1016/j.bbrc.2013.03.121] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Accepted: 03/28/2013] [Indexed: 11/16/2022]
Abstract
The bifunctional hexokinase KlHxk1 is a key component of glucose-dependent signal transduction in Kluyveromyces lactis. KlHxk1 is phosphorylated in vivo and undergoes ATP-dependent autophosphorylation-inactivation in vitro. This study identifies serine-15 as the site of in vivo phosphorylation and serine-157 as the autophosphorylation-inactivation site. X-ray crystallography of the in vivo phosphorylated enzyme indicates the existence of a ring-shaped symmetrical homodimer carrying two phosphoserine-15 residues. In contrast, small-angle X-ray scattering and equilibrium sedimentation analyses reveal the existence of monomeric phosphoserine-15 KlHxk1 in solution. While phosphorylation at serine-15 and concomitant homodimer dissociation are likely to be involved in glucose signalling, mechanism and putative physiological significance of KlHxk1 inactivation by autophosphorylation at serine-157 remain to be established.
Collapse
Affiliation(s)
- Karina Kettner
- Institute of Physiological Chemistry, Carl Gustav Carus Medical Faculty, Technische Universität Dresden, Dresden, Germany.
| | | | | | | | | | | | | |
Collapse
|
1380
|
Blanchet CE, Svergun DI. Small-Angle X-Ray Scattering on Biological Macromolecules and Nanocomposites in Solution. Annu Rev Phys Chem 2013; 64:37-54. [DOI: 10.1146/annurev-physchem-040412-110132] [Citation(s) in RCA: 152] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Clement E. Blanchet
- European Molecular Biology Laboratory (EMBL) Hamburg, 22603 Hamburg, Germany;
| | - Dmitri I. Svergun
- European Molecular Biology Laboratory (EMBL) Hamburg, 22603 Hamburg, Germany;
| |
Collapse
|
1381
|
Söderberg CAG, Rajan S, Shkumatov AV, Gakh O, Schaefer S, Ahlgren EC, Svergun DI, Isaya G, Al-Karadaghi S. The molecular basis of iron-induced oligomerization of frataxin and the role of the ferroxidation reaction in oligomerization. J Biol Chem 2013; 288:8156-8167. [PMID: 23344952 PMCID: PMC3605634 DOI: 10.1074/jbc.m112.442285] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Revised: 01/22/2013] [Indexed: 11/06/2022] Open
Abstract
The role of the mitochondrial protein frataxin in iron storage and detoxification, iron delivery to iron-sulfur cluster biosynthesis, heme biosynthesis, and aconitase repair has been extensively studied during the last decade. However, still no general consensus exists on the details of the mechanism of frataxin function and oligomerization. Here, using small-angle x-ray scattering and x-ray crystallography, we describe the solution structure of the oligomers formed during the iron-dependent assembly of yeast (Yfh1) and Escherichia coli (CyaY) frataxin. At an iron-to-protein ratio of 2, the initially monomeric Yfh1 is converted to a trimeric form in solution. The trimer in turn serves as the assembly unit for higher order oligomers induced at higher iron-to-protein ratios. The x-ray crystallographic structure obtained from iron-soaked crystals demonstrates that iron binds at the trimer-trimer interaction sites, presumably contributing to oligomer stabilization. For the ferroxidation-deficient D79A/D82A variant of Yfh1, iron-dependent oligomerization may still take place, although >50% of the protein is found in the monomeric state at the highest iron-to-protein ratio used. This demonstrates that the ferroxidation reaction controls frataxin assembly and presumably the iron chaperone function of frataxin and its interactions with target proteins. For E. coli CyaY, the assembly unit of higher order oligomers is a tetramer, which could be an effect of the much shorter N-terminal region of this protein. The results show that understanding of the mechanistic features of frataxin function requires detailed knowledge of the interplay between the ferroxidation reaction, iron-induced oligomerization, and the structure of oligomers formed during assembly.
Collapse
Affiliation(s)
- Christopher A G Söderberg
- Center for Molecular Protein Science, Institute for Chemistry and Chemical Engineering, Lund University, P. O. Box 124, SE-221 00 Lund, Sweden
| | - Sreekanth Rajan
- Center for Molecular Protein Science, Institute for Chemistry and Chemical Engineering, Lund University, P. O. Box 124, SE-221 00 Lund, Sweden
| | - Alexander V Shkumatov
- European Molecular Biology Laboratory (EMBL), Hamburg Unit c/o DESY, Notkestrasse 85, D-22603 Hamburg, Germany
| | - Oleksandr Gakh
- Departments of Pediatric and Adolescent Medicine and Biochemistry and Molecular Biology, Mayo Clinic, College of Medicine, Rochester, Minnesota 55905
| | - Susanne Schaefer
- Center for Molecular Protein Science, Institute for Chemistry and Chemical Engineering, Lund University, P. O. Box 124, SE-221 00 Lund, Sweden
| | - Eva-Christina Ahlgren
- Center for Molecular Protein Science, Institute for Chemistry and Chemical Engineering, Lund University, P. O. Box 124, SE-221 00 Lund, Sweden
| | - Dmitri I Svergun
- European Molecular Biology Laboratory (EMBL), Hamburg Unit c/o DESY, Notkestrasse 85, D-22603 Hamburg, Germany
| | - Grazia Isaya
- Departments of Pediatric and Adolescent Medicine and Biochemistry and Molecular Biology, Mayo Clinic, College of Medicine, Rochester, Minnesota 55905.
| | - Salam Al-Karadaghi
- Center for Molecular Protein Science, Institute for Chemistry and Chemical Engineering, Lund University, P. O. Box 124, SE-221 00 Lund, Sweden.
| |
Collapse
|
1382
|
Felix J, Elegheert J, Gutsche I, Shkumatov AV, Wen Y, Bracke N, Pannecoucke E, Vandenberghe I, Devreese B, Svergun DI, Pauwels E, Vergauwen B, Savvides SN. Human IL-34 and CSF-1 establish structurally similar extracellular assemblies with their common hematopoietic receptor. Structure 2013; 21:528-39. [PMID: 23478061 DOI: 10.1016/j.str.2013.01.018] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2012] [Revised: 01/22/2013] [Accepted: 01/28/2013] [Indexed: 12/21/2022]
Abstract
The discovery that hematopoietic human colony stimulating factor-1 receptor (CSF-1R) can be activated by two distinct cognate cytokines, colony stimulating factor-1 (CSF-1) and interleukin-34 (IL-34), created puzzling scenarios for the two possible signaling complexes. We here employ a hybrid structural approach based on small-angle X-ray scattering (SAXS) and negative-stain EM to reveal that bivalent binding of human IL-34 to CSF-1R leads to an extracellular assembly hallmarked by striking similarities to the CSF-1:CSF-1R complex, including homotypic receptor-receptor interactions. Thus, IL-34 and CSF-1 have evolved to exploit the geometric requirements of CSF-1R activation. Our models include N-linked oligomannose glycans derived from a systematic approach resulting in the accurate fitting of glycosylated models to the SAXS data. We further show that the C-terminal region of IL-34 is heavily glycosylated and that it can be proteolytically cleaved from the IL-34:hCSF-1R complex, providing insights into its role in the functional nonredundancy of IL-34 and CSF-1.
Collapse
Affiliation(s)
- Jan Felix
- Unit for Structural Biology, Laboratory for Protein Biochemistry and Biomolecular Engineering (L-ProBE), Ghent University, K.L. Ledeganckstraat 35, 9000 Ghent, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1383
|
Perry JJP, Tainer JA. Developing advanced X-ray scattering methods combined with crystallography and computation. Methods 2013; 59:363-71. [PMID: 23376408 PMCID: PMC3684416 DOI: 10.1016/j.ymeth.2013.01.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Revised: 01/15/2013] [Accepted: 01/18/2013] [Indexed: 01/09/2023] Open
Abstract
The extensive use of small angle X-ray scattering (SAXS) over the last few years is rapidly providing new insights into protein interactions, complex formation and conformational states in solution. This SAXS methodology allows for detailed biophysical quantification of samples of interest. Initial analyses provide a judgment of sample quality, revealing the potential presence of aggregation, the overall extent of folding or disorder, the radius of gyration, maximum particle dimensions and oligomerization state. Structural characterizations include ab initio approaches from SAXS data alone, and when combined with previously determined crystal/NMR, atomistic modeling can further enhance structural solutions and assess validity. This combination can provide definitions of architectures, spatial organizations of protein domains within a complex, including those not determined by crystallography or NMR, as well as defining key conformational states of a protein interaction. SAXS is not generally constrained by macromolecule size, and the rapid collection of data in a 96-well plate format provides methods to screen sample conditions. This includes screening for co-factors, substrates, differing protein or nucleotide partners or small molecule inhibitors, to more fully characterize the variations within assembly states and key conformational changes. Such analyses may be useful for screening constructs and conditions to determine those most likely to promote crystal growth of a complex under study. Moreover, these high throughput structural determinations can be leveraged to define how polymorphisms affect assembly formations and activities. This is in addition to potentially providing architectural characterizations of complexes and interactions for systems biology-based research, and distinctions in assemblies and interactions in comparative genomics. Thus, SAXS combined with crystallography/NMR and computation provides a unique set of tools that should be considered as being part of one's repertoire of biophysical analyses, when conducting characterizations of protein and other macromolecular interactions.
Collapse
Affiliation(s)
- J. Jefferson P. Perry
- Department of Integrative Structural and Computational Biology and Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA USA
- School of Biotechnology, Amrita University at Amritapuri, Kollam, Kerala, India
| | - John A. Tainer
- Department of Integrative Structural and Computational Biology and Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA USA
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA USA
| |
Collapse
|
1384
|
Wang YT, Kuo PH, Chiang CH, Liang JR, Chen YR, Wang S, Shen JCK, Yuan HS. The truncated C-terminal RNA recognition motif of TDP-43 protein plays a key role in forming proteinaceous aggregates. J Biol Chem 2013; 288:9049-57. [PMID: 23372158 PMCID: PMC3610977 DOI: 10.1074/jbc.m112.438564] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
TDP-43 is the major pathological protein identified in the cellular inclusions in amyotrophic lateral sclerosis and frontotemporal lobar degeneration. The pathogenic forms of TDP-43 are processed C-terminal fragments containing a truncated RNA-recognition motif (RRM2) and a glycine-rich region. Although extensive studies have focused on this protein, it remains unclear how the dimeric full-length TDP-43 is folded and assembled and how the processed C-terminal fragments are misfolded and aggregated. Here, using size-exclusion chromatography, pulldown assays, and small angle x-ray scattering, we show that the C-terminal-deleted TDP-43 without the glycine-rich tail is sufficient to form a head-to-head homodimer primarily via its N-terminal domain. The truncated RRM2, as well as two β-strands within the RRM2, form fibrils in vitro with a similar amyloid-negative staining property to those of TDP-43 pathogenic fibrils in diseases. In addition to the glycine-rich region, the truncated RRM2, but not the intact RRM2, plays a key role in forming cytoplasmic inclusions in neuronal cells. Our data thus suggest that the process that disrupts the dimeric structure, such as the proteolytic cleavage of TDP-43 within the RRM2 that removes the N-terminal dimerization domain, may produce unassembled truncated RRM2 fragments with abnormally exposed β-strands, which can oligomerize into high-order inclusions.
Collapse
Affiliation(s)
- Yi-Ting Wang
- Institute of Molecular Biology, Academia Sinica, Taipei 11529, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
1385
|
Meisburger SP, Warkentin M, Chen H, Hopkins JB, Gillilan RE, Pollack L, Thorne RE. Breaking the radiation damage limit with Cryo-SAXS. Biophys J 2013; 104:227-36. [PMID: 23332075 DOI: 10.1016/j.bpj.2012.11.3817] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Revised: 11/27/2012] [Accepted: 11/29/2012] [Indexed: 11/27/2022] Open
Abstract
Small angle x-ray scattering (SAXS) is a versatile and widely used technique for obtaining low-resolution structures of macromolecules and complexes. SAXS experiments measure molecules in solution, without the need for labeling or crystallization. However, radiation damage currently limits the application of SAXS to molecules that can be produced in microgram quantities; for typical proteins, 10-20 μL of solution at 1 mg/mL is required to accumulate adequate signal before irreversible x-ray damage is observed. Here, we show that cryocooled proteins and nucleic acids can withstand doses at least two orders of magnitude larger than room temperature samples. We demonstrate accurate T = 100 K particle envelope reconstructions from sample volumes as small as 15 nL, a factor of 1000 smaller than in current practice. Cryo-SAXS will thus enable structure determination of difficult-to-express proteins and biologically important, highly radiation-sensitive proteins including light-activated switches and metalloenzymes.
Collapse
Affiliation(s)
- Steve P Meisburger
- School of Applied and Engineering Physics, Cornell University, Ithaca, New York, USA
| | | | | | | | | | | | | |
Collapse
|
1386
|
Conrad KS, Bilwes AM, Crane BR. Light-induced subunit dissociation by a light-oxygen-voltage domain photoreceptor from Rhodobacter sphaeroides. Biochemistry 2013; 52:378-91. [PMID: 23252338 DOI: 10.1021/bi3015373] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Light-oxygen-voltage (LOV) domains bind a flavin chromophore to serve as blue light sensors in a wide range of eukaryotic and prokaryotic proteins. LOV domains are associated with a variable effector domain or a separate protein signaling partner to execute a wide variety of functions that include regulation of kinases, generation of anti-sigma factor antagonists, and regulation of circadian clocks. Here we present the crystal structure, photocycle kinetics, association properties, and spectroscopic features of a full-length LOV domain protein from Rhodobacter sphaeroides (RsLOV). RsLOV exhibits N- and C-terminal helical extensions that form an unusual helical bundle at its dimer interface with some resemblance to the helical transducer of sensory rhodopsin II. The blue light-induced conformational changes of RsLOV revealed from a comparison of light- and dark-state crystal structures support a shared signaling mechanism of LOV domain proteins that originates with the light-induced formation of a flavin-cysteinyl photoadduct. Adduct formation disrupts hydrogen bonding in the active site and propagates structural changes through the LOV domain core to the N- and C-terminal extensions. Single-residue variants in the active site and dimer interface of RsLOV alter photoadduct lifetimes and induce structural changes that perturb the oligomeric state. Size exclusion chromatography, multiangle light scattering, small-angle X-ray scattering, and cross-linking studies indicate that RsLOV dimerizes in the dark but, upon light excitation, dissociates into monomers. This light-induced switch in oligomeric state may prove to be useful for engineering molecular associations in controlled cellular settings.
Collapse
Affiliation(s)
- Karen S Conrad
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, USA
| | | | | |
Collapse
|
1387
|
Bhaumik A, Luchinat C, Parigi G, Ravera E, Rinaldelli M. NMR crystallography on paramagnetic systems: solved and open issues. CrystEngComm 2013. [DOI: 10.1039/c3ce41485j] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
1388
|
Makou E, Mertens HD, Maciejewski M, Soares DC, Matis I, Schmidt CQ, Herbert AP, Svergun DI, Barlow PN. Solution structure of CCP modules 10-12 illuminates functional architecture of the complement regulator, factor H. J Mol Biol 2012; 424:295-312. [PMID: 23017427 PMCID: PMC4068365 DOI: 10.1016/j.jmb.2012.09.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Revised: 09/17/2012] [Accepted: 09/18/2012] [Indexed: 01/08/2023]
Abstract
The 155-kDa plasma glycoprotein factor H (FH), which consists of 20 complement control protein (CCP) modules, protects self-tissue but not foreign organisms from damage by the complement cascade. Protection is achieved by selective engagement of FH, via CCPs 1-4, CCPs 6-8 and CCPs 19-20, with polyanion-rich host surfaces that bear covalently attached, activation-specific, fragments of complement component C3. The role of intervening CCPs 9-18 in this process is obscured by lack of structural knowledge. We have concatenated new high-resolution solution structures of overlapping recombinant CCP pairs, 10-11 and 11-12, to form a three-dimensional structure of CCPs 10-12 and validated it by small-angle X-ray scattering of the recombinant triple-module fragment. Superimposing CCP 12 of this 10-12 structure with CCP 12 from the previously solved CCP 12-13 structure yielded an S-shaped structure for CCPs 10-13 in which modules are tilted by 80-110° with respect to immediate neighbors, but the bend between CCPs 10 and 11 is counter to the arc traced by CCPs 11-13. Including this four-CCP structure in interpretation of scattering data for the longer recombinant segments, CCPs 10-15 and 8-15, implied flexible attachment of CCPs 8 and 9 to CCP 10 but compact and intimate arrangements of CCP 14 with CCPs 12, 13 and 15. Taken together with difficulties in recombinant production of module pairs 13-14 and 14-15, the aberrant structure of CCP 13 and the variability of 13-14 linker sequences among orthologues, a structural dependency of CCP 14 on its neighbors is suggested; this has implications for the FH mechanism.
Collapse
Key Words
- ccp, complement control protein
- cr1, complement receptor type 1
- daf, decay accelerating factor
- fh, factor h
- eom, ensemble optimization method
- hsqc, heteronuclear single quantum coherence
- mcp, membrane cofactor protein
- noe, nuclear overhauser enhancement
- saxs, small-angle x-ray scattering
- tocsy, total correlated spectroscopy
- protein nmr
- protein domains
- complement system
- small-angle x-ray scattering
- regulators of complement activation
Collapse
Affiliation(s)
- Elisavet Makou
- Schools of Chemistry and Biological Sciences, University of Edinburgh, West Mains Road, Edinburgh EH9 3JJ, UK
| | - Haydyn D.T. Mertens
- European Molecular Biology Laboratory Hamburg Outstation, c/o Deutsches Elektronen‐Synchrotron, Notkestrasse 85, 22603 Hamburg, Germany
| | - Mateusz Maciejewski
- Schools of Chemistry and Biological Sciences, University of Edinburgh, West Mains Road, Edinburgh EH9 3JJ, UK
| | - Dinesh C. Soares
- Medical Genetics Section, Molecular Medicine Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh Western General Hospital, Crewe Road South, Edinburgh EH4 2XU, UK
| | - Ilias Matis
- Schools of Chemistry and Biological Sciences, University of Edinburgh, West Mains Road, Edinburgh EH9 3JJ, UK
| | - Christoph Q. Schmidt
- Schools of Chemistry and Biological Sciences, University of Edinburgh, West Mains Road, Edinburgh EH9 3JJ, UK
| | - Andrew P. Herbert
- Schools of Chemistry and Biological Sciences, University of Edinburgh, West Mains Road, Edinburgh EH9 3JJ, UK
| | - Dmitri I. Svergun
- European Molecular Biology Laboratory Hamburg Outstation, c/o Deutsches Elektronen‐Synchrotron, Notkestrasse 85, 22603 Hamburg, Germany
| | - Paul N. Barlow
- Schools of Chemistry and Biological Sciences, University of Edinburgh, West Mains Road, Edinburgh EH9 3JJ, UK
| |
Collapse
|
1389
|
Cossu F, Milani M, Vachette P, Malvezzi F, Grassi S, Lecis D, Delia D, Drago C, Seneci P, Bolognesi M, Mastrangelo E. Structural insight into inhibitor of apoptosis proteins recognition by a potent divalent smac-mimetic. PLoS One 2012; 7:e49527. [PMID: 23166698 PMCID: PMC3499469 DOI: 10.1371/journal.pone.0049527] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Accepted: 10/10/2012] [Indexed: 11/18/2022] Open
Abstract
Genetic alterations enhancing cell survival and suppressing apoptosis are hallmarks of cancer that significantly reduce the efficacy of chemotherapy or radiotherapy. The Inhibitor of Apoptosis Protein (IAP) family hosts conserved proteins in the apoptotic pathway whose over-expression, frequently found in tumours, potentiates survival and resistance to anticancer agents. In humans, IAPs comprise eight members hosting one or more structural Baculoviral IAP Repeat (BIR) domains. Cellular IAPs (cIAP1 and 2) indirectly inhibit caspase-8 activation, and regulate both the canonical and the non-canonical NF-κB signaling pathways. In contrast to cIAPs, XIAP (X chromosome-linked Inhibitor of Apoptosis Protein) inhibits directly the effector caspases-3 and -7 through its BIR2 domain, and initiator caspase-9 through its BIR3 domain; molecular docking studies suggested that Smac/DIABLO antagonizes XIAP by simultaneously targeting both BIR2 and BIR3 domains. Here we report analytical gel filtration, crystallographic and SAXS experiments on cIAP1-BIR3, XIAP-BIR3 and XIAP-BIR2BIR3 domains, alone and in the presence of compound 9a, a divalent homodimeric Smac mimetic. 9a is shown to bind two BIR domains inter- (in the case of two BIR3) and intra-molecularly (in the case of XIAP-BIR2BIR3), with higher affinity for cIAP1-BIR3, relative to XIAP-BIR3. Despite the different crystal lattice packing, 9a maintains a right handed helical conformation in both cIAP1-BIR3 and XIAP-BIR3 crystals, that is likely conserved in solution as shown by SAXS data. Our structural results demonstrate that the 9a linker length, its conformational degrees of freedom and its hydrophobicity, warrant an overall compact structure with optimal solvent exposure of its two active moieties for IAPs binding. Our results show that 9a is a good candidate for pre-clinical and clinical studies, worth of further investigations in the field of cancer therapy.
Collapse
Affiliation(s)
- Federica Cossu
- Department of Biosciences, University of Milano, Milano, Italy
| | - Mario Milani
- Department of Biosciences, University of Milano, Milano, Italy
- CNR-Istituto di Biofisica, Università degli Studi di Milano, Milano, Italy
| | - Patrice Vachette
- Institut de Biochimie et de Biophysique Moléculaire et Cellulaire, UMR8619 CNRS, Université Paris-Sud, IFR115, Orsay, France
| | | | - Serena Grassi
- Department of Biosciences, University of Milano, Milano, Italy
| | | | | | - Carmelo Drago
- Centro Interdisciplinare Studi bio-molecolari e applicazioni Industriali (CISI), University of Milano, Milano, Italy
| | - Pierfausto Seneci
- Centro Interdisciplinare Studi bio-molecolari e applicazioni Industriali (CISI), University of Milano, Milano, Italy
- Department of Organic and Industrial Chemistry, University of Milano, Milano, Italy
| | - Martino Bolognesi
- Department of Biosciences, University of Milano, Milano, Italy
- CNR-Istituto di Biofisica, Università degli Studi di Milano, Milano, Italy
| | - Eloise Mastrangelo
- Department of Biosciences, University of Milano, Milano, Italy
- CNR-Istituto di Biofisica, Università degli Studi di Milano, Milano, Italy
- * E-mail:
| |
Collapse
|
1390
|
Petoukhov MV, Svergun DI. Applications of small-angle X-ray scattering to biomacromolecular solutions. Int J Biochem Cell Biol 2012; 45:429-37. [PMID: 23142499 DOI: 10.1016/j.biocel.2012.10.017] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2012] [Revised: 10/25/2012] [Accepted: 10/30/2012] [Indexed: 01/10/2023]
Abstract
Small-angle scattering of X-rays (SAXS) is an established method for low-resolution structural characterization of biological macromolecules in solution. Being complementary to the high resolution methods (X-ray crystallography and NMR), SAXS is often used in combination with them. The technique provides overall three-dimensional structures using ab initio reconstructions and hybrid modeling, and allows one to quantitatively characterize equilibrium mixtures as well as flexible systems. Recent progress in SAXS instrumentation, most notably, high brilliance synchrotron sources, has paved the way for high throughput automated SAXS studies allowing screening of external conditions (pH, temperature, ligand binding etc.). The modern approaches for SAXS data analysis are presented in this review including rapid characterization of macromolecular solutions in terms of low-resolution shapes, validation of high-resolution models in close-to-native conditions, quaternary structure analysis of complexes and quantitative description of the oligomeric composition in mixtures. Practical aspects of SAXS as a standalone tool and its combinations with other structural, biophysical or bioinformatics methods are reviewed. The capabilities of the technique are illustrated by a selection of recent applications for the studies of biological molecules. Future perspectives on SAXS and its potential impact to structural molecular biology are discussed.
Collapse
Affiliation(s)
- Maxim V Petoukhov
- European Molecular Biology Laboratory, Hamburg Outstation, Notkestrasse 85, 22607 Hamburg, Germany
| | | |
Collapse
|
1391
|
Müller MP, Shkumatov AV, Oesterlin LK, Schoebel S, Goody PR, Goody RS, Itzen A. Characterization of enzymes from Legionella pneumophila involved in reversible adenylylation of Rab1 protein. J Biol Chem 2012; 287:35036-35046. [PMID: 22872634 PMCID: PMC3471704 DOI: 10.1074/jbc.m112.396861] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2012] [Revised: 08/01/2012] [Indexed: 12/19/2022] Open
Abstract
After the pathogenic bacterium Legionella pneumophila is phagocytosed, it injects more than 250 different proteins into the cytoplasm of host cells to evade lysosomal digestion and to replicate inside the host cell. Among these secreted proteins is the protein DrrA/SidM, which has been shown to modify Rab1b, a main regulator of vesicular trafficking in eukaryotic cells, by transfer of adenosine monophosphate (AMP) to Tyr(77). In addition, Legionella provides the protein SidD that hydrolytically reverses the covalent modification, suggesting a tight spatial and temporal control of Rab1 function by Legionella during infection. Small angle x-ray scattering experiments of DrrA allowed us to validate a tentative complex model built by combining available crystallographic data. We have established the effects of adenylylation on Rab1 interactions and properties in a quantitative way. In addition, we have characterized the kinetics of DrrA-catalyzed adenylylation as well as SidD-catalyzed deadenylylation toward Rab1 and have determined the nucleotide specificities of both enzymes. This study enhances our knowledge of proteins subverting Rab1 function at the Legionella-containing vacuole.
Collapse
Affiliation(s)
- Matthias P Müller
- Department of Physical Biochemistry, Max Planck Institute of Molecular Physiology, Otto-Hahn-Strasse 11, 44227 Dortmund
| | - Alexander V Shkumatov
- European Molecular Biology Laboratory, Hamburg Outstation, EMBL c/o DESY, Notkestrasse 85, Geb 25 A, 22603 Hamburg
| | - Lena K Oesterlin
- Department of Physical Biochemistry, Max Planck Institute of Molecular Physiology, Otto-Hahn-Strasse 11, 44227 Dortmund
| | - Stefan Schoebel
- Department of Physical Biochemistry, Max Planck Institute of Molecular Physiology, Otto-Hahn-Strasse 11, 44227 Dortmund
| | - Philip R Goody
- Department of Physical Biochemistry, Max Planck Institute of Molecular Physiology, Otto-Hahn-Strasse 11, 44227 Dortmund
| | - Roger S Goody
- Department of Physical Biochemistry, Max Planck Institute of Molecular Physiology, Otto-Hahn-Strasse 11, 44227 Dortmund
| | - Aymelt Itzen
- Department of Physical Biochemistry, Max Planck Institute of Molecular Physiology, Otto-Hahn-Strasse 11, 44227 Dortmund; Center for Integrated Protein Science Munich (CIPSM), Chemistry Department, Technische Universität München, Lichtenbergstrasse 4, 85747 Garching, Germany.
| |
Collapse
|
1392
|
Cell-free protein synthesis of membrane (1,3)-β-d-glucan (curdlan) synthase: co-translational insertion in liposomes and reconstitution in nanodiscs. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2012; 1828:743-57. [PMID: 23063656 DOI: 10.1016/j.bbamem.2012.10.003] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2012] [Revised: 09/25/2012] [Accepted: 10/04/2012] [Indexed: 01/07/2023]
Abstract
A membrane-embedded curdlan synthase (CrdS) from Agrobacterium is believed to catalyse a repetitive addition of glucosyl residues from UDP-glucose to produce the (1,3)-β-d-glucan (curdlan) polymer. We report wheat germ cell-free protein synthesis (WG-CFPS) of full-length CrdS containing a 6xHis affinity tag and either Factor Xa or Tobacco Etch Virus proteolytic sites, using a variety of hydrophobic membrane-mimicking environments. Full-length CrdS was synthesised with no variations in primary structure, following analysis of tryptic fragments by MALDI-TOF/TOF Mass Spectrometry. Preparative scale WG-CFPS in dialysis mode with Brij-58 yielded CrdS in mg/ml quantities. Analysis of structural and functional properties of CrdS during protein synthesis showed that CrdS was co-translationally inserted in DMPC liposomes during WG-CFPS, and these liposomes could be purified in a single step by density gradient floatation. Incorporated CrdS exhibited a random orientation topology. Following affinity purification of CrdS, the protein was reconstituted in nanodiscs with Escherichia coli lipids or POPC and a membrane scaffold protein MSP1E3D1. CrdS nanodiscs were characterised by small-angle X-ray scattering using synchrotron radiation and the data obtained were consistent with insertion of CrdS into bilayers. We found CrdS synthesised in the presence of the Ac-AAAAAAD surfactant peptide or co-translationally inserted in liposomes made from E. coli lipids to be catalytically competent. Conversely, CrdS synthesised with only Brij-58 was inactive. Our findings pave the way for future structural studies of this industrially important catalytic membrane protein.
Collapse
|
1393
|
Crystal structure of ATV(ORF273), a new fold for a thermo- and acido-stable protein from the Acidianus two-tailed virus. PLoS One 2012; 7:e45847. [PMID: 23056221 PMCID: PMC3466262 DOI: 10.1371/journal.pone.0045847] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Accepted: 08/23/2012] [Indexed: 11/19/2022] Open
Abstract
Acidianus two-tailed virus (ATV) infects crenarchaea of the genus Acidianus living in terrestrial thermal springs at extremely high temperatures and low pH. ATV is a member of the Bicaudaviridae virus family and undergoes extra-cellular development of two tails, a process that is unique in the viral world. To understand this intriguing phenomenon, we have undertaken structural studies of ATV virion proteins and here we present the crystal structure of one of these proteins, ATV. ATV forms tetramers in solution and a molecular envelope is provided for the tetramer, computed from small-angle X-ray scattering (SAXS) data. The crystal structure has properties typical of hyperthermostable proteins, including a relatively high number of salt bridges. However, the protein also exhibits flexible loops and surface pockets. Remarkably, ATV displays a new protein fold, consistent with the absence of homologues of this protein in public sequence databases.
Collapse
|
1394
|
Allosteric competitive inactivation of hematopoietic CSF-1 signaling by the viral decoy receptor BARF1. Nat Struct Mol Biol 2012; 19:938-47. [PMID: 22902366 DOI: 10.1038/nsmb.2367] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2012] [Accepted: 07/19/2012] [Indexed: 12/24/2022]
Abstract
Hematopoietic human colony-stimulating factor 1 (hCSF-1) is essential for innate and adaptive immunity against viral and microbial infections and cancer. The human pathogen Epstein-Barr virus secretes the lytic-cycle protein BARF1 that neutralizes hCSF-1 to achieve immunomodulation. Here we show that BARF1 binds the dimer interface of hCSF-1 with picomolar affinity, away from the cognate receptor-binding site, to establish a long-lived complex featuring three hCSF-1 at the periphery of the BARF1 toroid. BARF1 locks dimeric hCSF-1 into an inactive conformation, rendering it unable to signal via its cognate receptor on human monocytes. This reveals a new functional role for hCSF-1 cooperativity in signaling. We propose a new viral strategy paradigm featuring an allosteric decoy receptor of the competitive type, which couples efficient sequestration and inactivation of the host growth factor to abrogate cooperative assembly of the cognate signaling complex.
Collapse
|
1395
|
Mertens HDT, Kjaergaard M, Mysling S, Gårdsvoll H, Jørgensen TJD, Svergun DI, Ploug M. A flexible multidomain structure drives the function of the urokinase-type plasminogen activator receptor (uPAR). J Biol Chem 2012; 287:34304-15. [PMID: 22896701 DOI: 10.1074/jbc.m112.398404] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The urokinase-type plasminogen activator receptor (uPAR) provides a rendezvous between proteolytic degradation of the extracellular matrix and integrin-mediated adhesion to vitronectin. These processes are, however, tightly linked because the high affinity binding of urokinase regulates the binding of uPAR to matrix-embedded vitronectin. Although crystal structures exist to define the corresponding static bi- and trimolecular receptor complexes, it is evident that the dynamic property of uPAR plays a decisive role in its function. In the present study, we combine small angle x-ray scattering, hydrogen-deuterium exchange, and surface plasmon resonance to develop a structural model describing the allosteric regulation of uPAR. We show that the flexibility of its N-terminal domain provides the key for understanding this allosteric mechanism. Importantly, our model has direct implications for understanding uPAR-assisted cell adhesion and migration as well as for translational research, including targeted intervention therapy and non-invasive tumor imaging in vivo.
Collapse
Affiliation(s)
- Haydyn D T Mertens
- Finsen Laboratory, Rigshospitalet and Biotech Research and Innovation Centre (BRIC), Copenhagen Biocenter, Ole Maaløes Vej 5, DK-2200 Copenhagen N, Denmark
| | | | | | | | | | | | | |
Collapse
|
1396
|
Taylor JEN, Chow JYH, Jeffries CM, Kwan AH, Duff AP, Hamilton WA, Trewhella J. Calmodulin binds a highly extended HIV-1 MA protein that refolds upon its release. Biophys J 2012; 103:541-549. [PMID: 22947870 PMCID: PMC3414894 DOI: 10.1016/j.bpj.2012.06.042] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2012] [Revised: 06/15/2012] [Accepted: 06/28/2012] [Indexed: 10/28/2022] Open
Abstract
Calmodulin (CaM) expression is upregulated upon HIV-1 infection and interacts with proteins involved in viral processing, including the multifunctional HIV-1 MA protein. We present here the results of studies utilizing small-angle neutron scattering with contrast variation that, when considered in the light of earlier fluorescence and NMR data, show CaM binds MA in an extended open-clamp conformation via interactions with two tryptophans that are widely spaced in sequence and space. The interaction requires a disruption of the MA tertiary fold such that MA becomes highly extended in a long snakelike conformation. The CaM-MA interface is extensive, covering ~70% of the length of the MA such that regions known to be important in MA interactions with critical binding partners would be impacted. The CaM conformation is semiextended and as such is distinct from the classical CaM-collapse about short α-helical targets. NMR data show that upon dissociation of the CaM-MA complex, either by the removal of Ca2+ or increasing ionic strength, MA reforms its native tertiary contacts. Thus, we observe a high level of structural plasticity in MA that may facilitate regulation of its activities via intracellular Ca2+-signaling during viral processing.
Collapse
Affiliation(s)
- James E N Taylor
- Bragg Institute, Australian Nuclear Science and Technology Organisation, New South Wales, Australia; School of Molecular Bioscience, The University of Sydney, New South Wales, Australia
| | - John Y H Chow
- School of Molecular Bioscience, The University of Sydney, New South Wales, Australia
| | - Cy M Jeffries
- School of Molecular Bioscience, The University of Sydney, New South Wales, Australia
| | - Ann H Kwan
- School of Molecular Bioscience, The University of Sydney, New South Wales, Australia
| | - Anthony P Duff
- Bragg Institute, Australian Nuclear Science and Technology Organisation, New South Wales, Australia
| | - William A Hamilton
- Bragg Institute, Australian Nuclear Science and Technology Organisation, New South Wales, Australia
| | - Jill Trewhella
- School of Molecular Bioscience, The University of Sydney, New South Wales, Australia.
| |
Collapse
|
1397
|
Schneidman-Duhovny D, Kim SJ, Sali A. Integrative structural modeling with small angle X-ray scattering profiles. BMC STRUCTURAL BIOLOGY 2012; 12:17. [PMID: 22800408 PMCID: PMC3427135 DOI: 10.1186/1472-6807-12-17] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Accepted: 07/16/2012] [Indexed: 01/24/2023]
Abstract
Recent technological advances enabled high-throughput collection of Small Angle X-ray Scattering (SAXS) profiles of biological macromolecules. Thus, computational methods for integrating SAXS profiles into structural modeling are needed more than ever. Here, we review specifically the use of SAXS profiles for the structural modeling of proteins, nucleic acids, and their complexes. First, the approaches for computing theoretical SAXS profiles from structures are presented. Second, computational methods for predicting protein structures, dynamics of proteins in solution, and assembly structures are covered. Third, we discuss the use of SAXS profiles in integrative structure modeling approaches that depend simultaneously on several data types.
Collapse
Affiliation(s)
- Dina Schneidman-Duhovny
- Department of Bioengineering and Therapeutic Sciences, University of California at San Francisco, San Francisco, USA.
| | | | | |
Collapse
|
1398
|
Ligand-binding properties and conformational dynamics of autolysin repeat domains in staphylococcal cell wall recognition. J Bacteriol 2012; 194:3789-802. [PMID: 22609916 DOI: 10.1128/jb.00331-12] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The bifunctional major autolysin Atl plays a key role in staphylococcal cell separation. Processing of Atl yields catalytically active amidase (AM) and glucosaminidase (GL) domains that are each fused to repeating units. The two repeats of AM (R1 and R2) target the enzyme to the septum, where it cleaves murein between dividing cells. We have determined the crystal structure of R2, which reveals that each repeat folds into two half-open β-barrel subunits. We further demonstrate that lipoteichoic acid serves as a receptor for the repeats and that this interaction depends on conserved surfaces in each subunit. Small-angle X-ray scattering of the mature amidase reveals the presence of flexible linkers separating the AM, R1, and R2 units. Different levels of flexibility for each linker provide mechanistic insights into the conformational dynamics of the full-length protein and the roles of its components in cell wall association and catalysis. Our analysis supports a model in which the repeats direct the catalytic AM domain to the septum, where it can optimally perform the final step of cell division.
Collapse
|