Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Nat Commun ; 14(1): 8248, 2023 Dec 12.
Article in English | MEDLINE | ID: mdl-38086790

ABSTRACT

The Mitochondrial Complex I Assembly (MCIA) complex is essential for the biogenesis of respiratory Complex I (CI), the first enzyme in the respiratory chain, which has been linked to Alzheimer's disease (AD) pathogenesis. However, how MCIA facilitates CI assembly, and how it is linked with AD pathogenesis, is poorly understood. Here we report the structural basis of the complex formation between the MCIA subunits ECSIT and ACAD9. ECSIT binding induces a major conformational change in the FAD-binding loop of ACAD9, releasing the FAD cofactor and converting ACAD9 from a fatty acid ß-oxidation (FAO) enzyme to a CI assembly factor. We provide evidence that ECSIT phosphorylation downregulates its association with ACAD9 and is reduced in neuronal cells upon exposure to amyloid-ß (Aß) oligomers. These findings advance our understanding of the MCIA complex assembly and suggest a possible role for ECSIT in the reprogramming of bioenergetic pathways linked to Aß toxicity, a hallmark of AD.


Subject(s)
Alzheimer Disease , Electron Transport Complex I , Humans , Oxidation-Reduction , Electron Transport Complex I/metabolism , Energy Metabolism , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism
2.
Nat Commun ; 14(1): 5732, 2023 09 15.
Article in English | MEDLINE | ID: mdl-37714861

ABSTRACT

Human Respiratory Syncytial Virus (HRSV) is a prevalent cause of severe respiratory infections in children and the elderly. The helical HRSV nucleocapsid is a template for the viral RNA synthesis and a scaffold for the virion assembly. This cryo-electron microscopy analysis reveals the non-canonical arrangement of the HRSV nucleocapsid helix, composed of 16 nucleoproteins per asymmetric unit, and the resulting systematic variations in the RNA accessibility. We demonstrate that this unique helical symmetry originates from longitudinal interactions by the C-terminal arm of the HRSV nucleoprotein. We explore the polymorphism of the nucleocapsid-like assemblies, report five structures of the full-length particles and two alternative arrangements formed by a C-terminally truncated nucleoprotein mutant, and demonstrate the functional importance of the identified longitudinal interfaces. We put all these findings in the context of the HRSV RNA synthesis machinery and delineate the structural basis for its further investigation.


Subject(s)
Respiratory Syncytial Virus, Human , Child , Aged , Humans , Respiratory Syncytial Virus, Human/genetics , Cryoelectron Microscopy , Nucleocapsid/genetics , RNA, Viral/genetics , Nucleoproteins/genetics
3.
Int J Biol Macromol ; 252: 126345, 2023 Dec 01.
Article in English | MEDLINE | ID: mdl-37619685

ABSTRACT

Low-density lipoprotein (LDL) plays a crucial role in cholesterol metabolism. Responsible for cholesterol transport from the liver to the organs, LDL accumulation in the arteries is a primary cause of cardiovascular diseases, such as atherosclerosis. This work focuses on the fundamental question of the LDL molecular structure, as well as the topology and molecular motions of apolipoprotein B-100 (apo B-100), which is addressed by single-particle cryo-electron microscopy (cryo-EM) and high-speed atomic force microscopy (HS-AFM). Our results suggest a revised model of the LDL core organization with respect to the cholesterol ester (CE) arrangement. In addition, a high-density region close to the flattened poles could be identified, likely enriched in free cholesterol. The most remarkable new details are two protrusions on the LDL surface, attributed to the protein apo B-100. HS-AFM adds the dimension of time and reveals for the first time a highly dynamic direct description of LDL, where we could follow large domain fluctuations of the protrusions in real time. To tackle the inherent flexibility and heterogeneity of LDL, the cryo-EM maps are further assessed by 3D variability analysis. Our study gives a detailed explanation how to approach the intrinsic flexibility of a complex system comprising lipids and protein.


Subject(s)
Cholesterol , Lipoproteins, LDL , Lipoproteins, LDL/metabolism , Cryoelectron Microscopy , Apolipoprotein B-100 , Microscopy, Atomic Force/methods
4.
Nat Struct Mol Biol ; 30(1): 81-90, 2023 01.
Article in English | MEDLINE | ID: mdl-36604498

ABSTRACT

The endosomal sorting complex required for transport (ESCRT) is a highly conserved protein machinery that drives a divers set of physiological and pathological membrane remodeling processes. However, the structural basis of ESCRT-III polymers stabilizing, constricting and cleaving negatively curved membranes is yet unknown. Here we present cryo-EM structures of membrane-coated CHMP2A-CHMP3 filaments from Homo sapiens of two different diameters at 3.3 and 3.6 Å resolution. The structures reveal helical filaments assembled by CHMP2A-CHMP3 heterodimers in the open ESCRT-III conformation, which generates a partially positive charged membrane interaction surface, positions short N-terminal motifs for membrane interaction and the C-terminal VPS4 target sequence toward the tube interior. Inter-filament interactions are electrostatic, which may facilitate filament sliding upon VPS4-mediated polymer remodeling. Fluorescence microscopy as well as high-speed atomic force microscopy imaging corroborate that VPS4 can constrict and cleave CHMP2A-CHMP3 membrane tubes. We therefore conclude that CHMP2A-CHMP3-VPS4 act as a minimal membrane fission machinery.


Subject(s)
Endosomal Sorting Complexes Required for Transport , Polymers , Humans , Endosomal Sorting Complexes Required for Transport/chemistry , Polymers/metabolism , Carrier Proteins/metabolism , Protein Transport
5.
Nat Struct Mol Biol ; 30(3): 273-285, 2023 03.
Article in English | MEDLINE | ID: mdl-36702972

ABSTRACT

Target of rapamycin complex 1 (TORC1) is a protein kinase controlling cell homeostasis and growth in response to nutrients and stresses. In Saccharomyces cerevisiae, glucose depletion triggers a redistribution of TORC1 from a dispersed localization over the vacuole surface into a large, inactive condensate called TOROID (TORC1 organized in inhibited domains). However, the mechanisms governing this transition have been unclear. Here, we show that acute depletion and repletion of EGO complex (EGOC) activity is sufficient to control TOROID distribution, independently of other nutrient-signaling pathways. The 3.9-Å-resolution structure of TORC1 from TOROID cryo-EM data together with interrogation of key interactions in vivo provide structural insights into TORC1-TORC1' and TORC1-EGOC interaction interfaces. These data support a model in which glucose-dependent activation of EGOC triggers binding to TORC1 at an interface required for TOROID assembly, preventing TORC1 polymerization and promoting release of active TORC1.


Subject(s)
Saccharomyces cerevisiae Proteins , Mechanistic Target of Rapamycin Complex 1/chemistry , Mechanistic Target of Rapamycin Complex 1/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Polymerization , Transcription Factors/metabolism , Saccharomyces cerevisiae/metabolism , Glucose/metabolism
6.
Nature ; 612(7938): 162-169, 2022 12.
Article in English | MEDLINE | ID: mdl-36418402

ABSTRACT

The poly-ADP-ribosyltransferase tankyrase (TNKS, TNKS2) controls a wide range of disease-relevant cellular processes, including WNT-ß-catenin signalling, telomere length maintenance, Hippo signalling, DNA damage repair and glucose homeostasis1,2. This has incentivized the development of tankyrase inhibitors. Notwithstanding, our knowledge of the mechanisms that control tankyrase activity has remained limited. Both catalytic and non-catalytic functions of tankyrase depend on its filamentous polymerization3-5. Here we report the cryo-electron microscopy reconstruction of a filament formed by a minimal active unit of tankyrase, comprising the polymerizing sterile alpha motif (SAM) domain and its adjacent catalytic domain. The SAM domain forms a novel antiparallel double helix, positioning the protruding catalytic domains for recurring head-to-head and tail-to-tail interactions. The head interactions are highly conserved among tankyrases and induce an allosteric switch in the active site within the catalytic domain to promote catalysis. Although the tail interactions have a limited effect on catalysis, they are essential to tankyrase function in WNT-ß-catenin signalling. This work reveals a novel SAM domain polymerization mode, illustrates how supramolecular assembly controls catalytic and non-catalytic functions, provides important structural insights into the regulation of a non-DNA-dependent poly-ADP-ribosyltransferase and will guide future efforts to modulate tankyrase and decipher its contribution to disease mechanisms.


Subject(s)
Biocatalysis , Cryoelectron Microscopy , Polymerization , Tankyrases , beta Catenin , Tankyrases/chemistry , Tankyrases/metabolism , Tankyrases/ultrastructure , Enzyme Activation , Catalytic Domain , Wnt Signaling Pathway , Amino Acid Motifs
7.
Commun Biol ; 5(1): 317, 2022 04 05.
Article in English | MEDLINE | ID: mdl-35383285

ABSTRACT

Bacterial homologous lysine and arginine decarboxylases play major roles in the acid stress response, physiology, antibiotic resistance and virulence. The Escherichia coli enzymes are considered as their archetypes. Whereas acid stress triggers polymerisation of the E. coli lysine decarboxylase LdcI, such behaviour has not been observed for the arginine decarboxylase Adc. Here we show that the Adc from a multidrug-resistant human pathogen Providencia stuartii massively polymerises into filaments whose cryo-EM structure reveals pronounced differences between Adc and LdcI assembly mechanisms. While the structural determinants of Adc polymerisation are conserved only in certain Providencia and Burkholderia species, acid stress-induced polymerisation of LdcI appears general for enterobacteria. Analysis of the expression, activity and oligomerisation of the P. stuartii Adc further highlights the distinct properties of this unusual protein and lays a platform for future investigation of the role of supramolecular assembly in the superfamily or arginine and lysine decarboxylases.


Subject(s)
Carboxy-Lyases , Providencia , Carboxy-Lyases/genetics , Carboxy-Lyases/metabolism , Escherichia coli/metabolism , Providencia/enzymology
8.
Proc Natl Acad Sci U S A ; 118(2)2021 01 12.
Article in English | MEDLINE | ID: mdl-33372137

ABSTRACT

Pathogenic and commensal bacteria often have to resist the harsh acidity of the host stomach. The inducible lysine decarboxylase LdcI buffers the cytosol and the local extracellular environment to ensure enterobacterial survival at low pH. Here, we investigate the acid stress-response regulation of Escherichia coli LdcI by combining biochemical and biophysical characterization with negative stain and cryoelectron microscopy (cryo-EM) and wide-field and superresolution fluorescence imaging. Due to deleterious effects of fluorescent protein fusions on native LdcI decamers, we opt for three-dimensional localization of nanobody-labeled endogenous wild-type LdcI in acid-stressed E. coli cells and show that it organizes into distinct patches at the cell periphery. Consistent with recent hypotheses that in vivo clustering of metabolic enzymes often reflects their polymerization as a means of stimulus-induced regulation, we show that LdcI assembles into filaments in vitro at physiologically relevant low pH. We solve the structures of these filaments and of the LdcI decamer formed at neutral pH by cryo-EM and reveal the molecular determinants of LdcI polymerization, confirmed by mutational analysis. Finally, we propose a model for LdcI function inside the enterobacterial cell, providing a structural and mechanistic basis for further investigation of the role of its supramolecular organization in the acid stress response.


Subject(s)
Carboxy-Lyases/metabolism , Microscopy, Fluorescence/methods , Stress, Physiological/physiology , Adenosine Triphosphatases/metabolism , Amino Acid Sequence/genetics , Carboxy-Lyases/physiology , Cryoelectron Microscopy/methods , Crystallography, X-Ray/methods , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Hydrogen-Ion Concentration , Models, Molecular , Protein Binding/genetics , Protein Multimerization/genetics
9.
Nat Commun ; 11(1): 743, 2020 02 06.
Article in English | MEDLINE | ID: mdl-32029744

ABSTRACT

Motile bacteria sense chemical gradients with transmembrane receptors organised in supramolecular signalling arrays. Understanding stimulus detection and transmission at the molecular level requires precise structural characterisation of the array building block known as a core signalling unit. Here we introduce an Escherichia coli strain that forms small minicells possessing extended and highly ordered chemosensory arrays. We use cryo-electron tomography and subtomogram averaging to provide a three-dimensional map of a complete core signalling unit, with visible densities corresponding to the HAMP and periplasmic domains. This map, combined with previously determined high resolution structures and molecular dynamics simulations, yields a molecular model of the transmembrane core signalling unit and enables spatial localisation of its individual domains. Our work thus offers a solid structural basis for the interpretation of a wide range of existing data and the design of further experiments to elucidate signalling mechanisms within the core signalling unit and larger array.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli/chemistry , Methyl-Accepting Chemotaxis Proteins/chemistry , Cryoelectron Microscopy , Electron Microscope Tomography , Escherichia coli/genetics , Escherichia coli/ultrastructure , Escherichia coli Proteins/genetics , Escherichia coli Proteins/ultrastructure , Histidine Kinase/chemistry , Histidine Kinase/genetics , Histidine Kinase/ultrastructure , Methyl-Accepting Chemotaxis Proteins/genetics , Methyl-Accepting Chemotaxis Proteins/ultrastructure , Models, Molecular , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Multiprotein Complexes/ultrastructure
10.
PLoS Biol ; 17(11): e3000472, 2019 11.
Article in English | MEDLINE | ID: mdl-31714936

ABSTRACT

With the rapid improvement of cryo-electron microscopy (cryo-EM) resolution, new computational tools are needed to assist and improve upon atomic model building and refinement options. This communication demonstrates that microscopists can now collaborate with the players of the computer game Foldit to generate high-quality de novo structural models. This development could greatly speed the generation of excellent cryo-EM structures when used in addition to current methods.


Subject(s)
Citizen Science , Cryoelectron Microscopy/trends , Video Games , Algorithms , Biochemistry , Cryoelectron Microscopy/methods , Models, Molecular
12.
Nat Microbiol ; 4(11): 1885-1894, 2019 11.
Article in English | MEDLINE | ID: mdl-31384001

ABSTRACT

Contractile injection systems are sophisticated multiprotein nanomachines that puncture target cell membranes. Although the number of atomic-resolution insights into contractile bacteriophage tails, bacterial type six secretion systems and R-pyocins is rapidly increasing, structural information on the contraction of bacterial phage-like protein-translocation structures directed towards eukaryotic hosts is scarce. Here, we characterize the antifeeding prophage AFP from Serratia entomophila by cryo-electron microscopy. We present the high-resolution structure of the entire AFP particle in the extended state, trace 11 protein chains de novo from the apical cap to the needle tip, describe localization variants and perform specific structural comparisons with related systems. We analyse inter-subunit interactions and highlight their universal conservation within contractile injection systems while revealing the specificities of AFP. Furthermore, we provide the structure of the AFP sheath-baseplate complex in a contracted state. This study reveals atomic details of interaction networks that accompany and define the contraction mechanism of toxin-delivery tailocins, offering a comprehensive framework for understanding their mode of action and for their possible adaptation as biocontrol agents.


Subject(s)
Prophages/physiology , Serratia/virology , Type VI Secretion Systems/chemistry , Cryoelectron Microscopy , Prophages/chemistry , Protein Conformation , Type VI Secretion Systems/metabolism , Viral Proteins/chemistry , Viral Proteins/metabolism
13.
Proc Natl Acad Sci U S A ; 116(10): 4256-4264, 2019 03 05.
Article in English | MEDLINE | ID: mdl-30787192

ABSTRACT

Assembly of paramyxoviral nucleocapsids on the RNA genome is an essential step in the viral cycle. The structural basis of this process has remained obscure due to the inability to control encapsidation. We used a recently developed approach to assemble measles virus nucleocapsid-like particles on specific sequences of RNA hexamers (poly-Adenine and viral genomic 5') in vitro, and determined their cryoelectron microscopy maps to 3.3-Å resolution. The structures unambiguously determine 5' and 3' binding sites and thereby the binding-register of viral genomic RNA within nucleocapsids. This observation reveals that the 3' end of the genome is largely exposed in fully assembled measles nucleocapsids. In particular, the final three nucleotides of the genome are rendered accessible to the RNA-dependent RNA polymerase complex, possibly enabling efficient RNA processing. The structures also reveal local and global conformational changes in the nucleoprotein upon assembly, in particular involving helix α6 and helix α13 that form edges of the RNA binding groove. Disorder is observed in the bound RNA, localized at one of the two backbone conformational switch sites. The high-resolution structure allowed us to identify putative nucleobase interaction sites in the RNA-binding groove, whose impact on assembly kinetics was measured using real-time NMR. Mutation of one of these sites, R195, whose sidechain stabilizes both backbone and base of a bound nucleic acid, is thereby shown to be essential for nucleocapsid-like particle assembly.


Subject(s)
Cryoelectron Microscopy/methods , Measles virus/chemistry , Measles virus/metabolism , Nucleocapsid/chemistry , Nucleocapsid/metabolism , Nucleocapsid/ultrastructure , Virus Assembly , Binding Sites , Genome, Viral , Kinetics , Magnetic Resonance Imaging/methods , Models, Molecular , Molecular Conformation , Nucleocapsid Proteins , Nucleoproteins/chemistry , Nucleoproteins/metabolism , Nucleoproteins/ultrastructure , Paramyxoviridae/chemistry , Paramyxoviridae/ultrastructure , RNA, Viral/chemistry , RNA, Viral/metabolism , RNA, Viral/ultrastructure , Viral Proteins/chemistry , Viral Proteins/metabolism , Viral Proteins/ultrastructure
14.
Elife ; 82019 01 14.
Article in English | MEDLINE | ID: mdl-30638449

ABSTRACT

Negative-strand RNA viruses condense their genome into helical nucleocapsids that constitute essential templates for viral replication and transcription. The intrinsic flexibility of nucleocapsids usually prevents their full-length structural characterisation at high resolution. Here, we describe purification of full-length recombinant metastable helical nucleocapsid of Hantaan virus (Hantaviridae family, Bunyavirales order) and determine its structure at 3.3 Å resolution by cryo-electron microscopy. The structure reveals the mechanisms of helical multimerisation via sub-domain exchanges between protomers and highlights nucleotide positions in a continuous positively charged groove compatible with viral genome binding. It uncovers key sites for future structure-based design of antivirals that are currently lacking to counteract life-threatening hantavirus infections. The structure also suggests a model of nucleoprotein-polymerase interaction that would enable replication and transcription solely upon local disruption of the nucleocapsid.


Subject(s)
Cryoelectron Microscopy/methods , Hantaan virus/metabolism , Nucleocapsid/metabolism , RNA, Viral/metabolism , Virus Assembly , Animals , Genome, Viral , HEK293 Cells , Hantaan virus/genetics , Hantaan virus/physiology , Humans , Insecta , Nucleocapsid/chemistry , Protein Conformation
15.
Nat Commun ; 9(1): 4041, 2018 10 02.
Article in English | MEDLINE | ID: mdl-30279415

ABSTRACT

The CARD11-BCL10-MALT1 (CBM) complex triggers the adaptive immune response in lymphocytes and lymphoma cells. CARD11/CARMA1 acts as a molecular seed inducing BCL10 filaments, but the integration of MALT1 and the assembly of a functional CBM complex has remained elusive. Using cryo-EM we solved the helical structure of the BCL10-MALT1 filament. The structural model of the filament core solved at 4.9 Å resolution identified the interface between the N-terminal MALT1 DD and the BCL10 caspase recruitment domain. The C-terminal MALT1 Ig and paracaspase domains protrude from this core to orchestrate binding of mediators and substrates at the filament periphery. Mutagenesis studies support the importance of the identified BCL10-MALT1 interface for CBM complex assembly, MALT1 protease activation and NF-κB signaling in Jurkat and primary CD4 T-cells. Collectively, we present a model for the assembly and architecture of the CBM signaling complex and how it functions as a signaling hub in T-lymphocytes.


Subject(s)
B-Cell CLL-Lymphoma 10 Protein/ultrastructure , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/ultrastructure , B-Cell CLL-Lymphoma 10 Protein/chemistry , B-Cell CLL-Lymphoma 10 Protein/metabolism , CARD Signaling Adaptor Proteins/metabolism , Cryoelectron Microscopy , Guanylate Cyclase/metabolism , Lymphocyte Activation , Models, Chemical , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/chemistry , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/metabolism , Protein Conformation
16.
Nat Commun ; 9(1): 4043, 2018 10 02.
Article in English | MEDLINE | ID: mdl-30279485

ABSTRACT

Activation of the innate immune pattern recognition receptor NOD2 by the bacterial muramyl-dipeptide peptidoglycan fragment triggers recruitment of the downstream adaptor kinase RIP2, eventually leading to NF-κB activation and proinflammatory cytokine production. Here we show that full-length RIP2 can form long filaments mediated by its caspase recruitment domain (CARD), in common with other innate immune adaptor proteins. We further show that the NOD2 tandem CARDs bind to one end of the RIP2 CARD filament, suggesting a mechanism for polar filament nucleation by activated NOD2. We combine X-ray crystallography, solid-state NMR and high-resolution cryo-electron microscopy to determine the atomic structure of the helical RIP2 CARD filament, which reveals the intermolecular interactions that stabilize the assembly. Using structure-guided mutagenesis, we demonstrate the importance of RIP2 polymerization for the activation of NF-κB signalling by NOD2. Our results could be of use to develop new pharmacological strategies to treat inflammatory diseases characterised by aberrant NOD2 signalling.


Subject(s)
NF-kappa B/metabolism , Nod2 Signaling Adaptor Protein/metabolism , Receptor-Interacting Protein Serine-Threonine Kinase 2/metabolism , Caspase Activation and Recruitment Domain , HEK293 Cells , Humans , Protein Conformation , Receptor-Interacting Protein Serine-Threonine Kinase 2/genetics
17.
Nature ; 550(7675): 265-269, 2017 10 12.
Article in English | MEDLINE | ID: mdl-28976958

ABSTRACT

The target of rapamycin (TOR) is a eukaryotic serine/threonine protein kinase that functions in two distinct complexes, TORC1 and TORC2, to regulate growth and metabolism. GTPases, responding to signals generated by abiotic stressors, nutrients, and, in metazoans, growth factors, play an important but poorly understood role in TORC1 regulation. Here we report that, in budding yeast, glucose withdrawal (which leads to an acute loss of TORC1 kinase activity) triggers a similarly rapid Rag GTPase-dependent redistribution of TORC1 from being semi-uniform around the vacuolar membrane to a single, vacuole-associated cylindrical structure visible by super-resolution optical microscopy. Three-dimensional reconstructions of cryo-electron micrograph images of these purified cylinders demonstrate that TORC1 oligomerizes into a higher-level hollow helical assembly, which we name a TOROID (TORC1 organized in inhibited domain). Fitting of the recently described mammalian TORC1 structure into our helical map reveals that oligomerization leads to steric occlusion of the active site. Guided by the implications from our reconstruction, we present a TOR1 allele that prevents both TOROID formation and TORC1 inactivation in response to glucose withdrawal, demonstrating that oligomerization is necessary for TORC1 inactivation. Our results reveal a novel mechanism by which Rag GTPases regulate TORC1 activity and suggest that the reversible assembly and/or disassembly of higher-level structures may be an underappreciated mechanism for the regulation of protein kinases.


Subject(s)
Cryoelectron Microscopy , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 1/ultrastructure , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/ultrastructure , Alleles , Catalytic Domain , Enzyme Activation , Glucose/deficiency , Glucose/metabolism , Glucose/pharmacology , Mechanistic Target of Rapamycin Complex 1/chemistry , Monomeric GTP-Binding Proteins/deficiency , Monomeric GTP-Binding Proteins/genetics , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics
18.
Sci Rep ; 7(1): 2913, 2017 06 06.
Article in English | MEDLINE | ID: mdl-28588198

ABSTRACT

During a proteolytically-driven maturation process, the orthoretroviral capsid protein (CA) assembles to form the convex shell that surrounds the viral genome. In some orthoretroviruses, including Rous Sarcoma Virus (RSV), CA carries a short and hydrophobic spacer peptide (SP) at its C-terminus early in the maturation process, which is progressively removed as maturation proceeds. In this work, we show that RSV CA assembles in vitro at near-physiological temperatures, forming hexamer tubes that effectively model the mature capsid surface. Tube assembly is strongly influenced by electrostatic effects, and is a nucleated process that remains thermodynamically favored at lower temperatures, but is effectively arrested by the large Gibbs energy barrier associated with nucleation. RSV CA tubes are multi-layered, being formed by nested and concentric tubes of capsid hexamers. However the spacer peptide acts as a layering determinant during tube assembly. If only a minor fraction of CA-SP is present, multi-layered tube formation is blocked, and single-layered tubes predominate. This likely prevents formation of biologically aberrant multi-layered capsids in the virion. The generation of single-layered hexamer tubes facilitated 3D helical image reconstruction from cryo-electron microscopy data, revealing the basic tube architecture.


Subject(s)
Capsid Proteins/metabolism , Rous sarcoma virus/physiology , Virus Assembly , Capsid Proteins/chemistry , Capsid Proteins/genetics , Capsid Proteins/ultrastructure , Imaging, Three-Dimensional , In Vitro Techniques , Models, Molecular , Osmolar Concentration , Protein Binding , Protein Conformation , Protein Multimerization , Proteolysis , Static Electricity , Temperature
19.
J Am Chem Soc ; 139(5): 2006-2013, 2017 02 08.
Article in English | MEDLINE | ID: mdl-28094514

ABSTRACT

The orthoretroviral capsid protein (CA) assembles into polymorphic capsids, whose architecture, assembly, and stability are still being investigated. The N-terminal and C-terminal domains of CA (NTD and CTD, respectively) engage in both homotypic and heterotypic interactions to create the capsid. Hexameric turrets formed by the NTD decorate the majority of the capsid surface. We report nearly complete solid-state NMR (ssNMR) resonance assignments of Rous sarcoma virus (RSV) CA, assembled into hexamer tubes that mimic the authentic capsid. The ssNMR assignments show that, upon assembly, large conformational changes occur in loops connecting helices, as well as the short 310 helix initiating the CTD. The interdomain linker becomes statically disordered. Combining constraints from ssNMR and cryo-electron microscopy (cryo-EM), we establish an atomic resolution model of the RSV CA tubular assembly using molecular dynamics flexible fitting (MDFF) simulations. On the basis of comparison of this MDFF model with an earlier-derived crystallographic model for the planar assembly, the induction of curvature into the RSV CA hexamer lattice arises predominantly from reconfiguration of the NTD-CTD and CTD trimer interfaces. The CTD dimer and CTD trimer interfaces are also intrinsically variable. Hence, deformation of the CA hexamer lattice results from the variable displacement of the CTDs that surround each hexameric turret. Pervasive H-bonding is found at all interdomain interfaces, which may contribute to their malleability. Finally, we find helices at the interfaces of HIV and RSV CA assemblies have very different contact angles, which may reflect differences in the capsid assembly pathway for these viruses.


Subject(s)
Capsid Proteins/chemistry , Rous sarcoma virus/chemistry , Crystallography, X-Ray , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular , Particle Size , Protein Conformation , Surface Properties
20.
EMBO Rep ; 18(2): 264-279, 2017 02.
Article in English | MEDLINE | ID: mdl-27974378

ABSTRACT

The highly conserved eukaryotic Elongator complex performs specific chemical modifications on wobble base uridines of tRNAs, which are essential for proteome stability and homeostasis. The complex is formed by six individual subunits (Elp1-6) that are all equally important for its tRNA modification activity. However, its overall architecture and the detailed reaction mechanism remain elusive. Here, we report the structures of the fully assembled yeast Elongator and the Elp123 sub-complex solved by an integrative structure determination approach showing that two copies of the Elp1, Elp2, and Elp3 subunits form a two-lobed scaffold, which binds Elp456 asymmetrically. Our topological models are consistent with previous studies on individual subunits and further validated by complementary biochemical analyses. Our study provides a structural framework on how the tRNA modification activity is carried out by Elongator.


Subject(s)
Fungal Proteins/chemistry , Models, Molecular , Multiprotein Complexes/chemistry , Fungal Proteins/genetics , Fungal Proteins/metabolism , Multiprotein Complexes/metabolism , Multiprotein Complexes/ultrastructure , Mutation , Protein Binding , Protein Conformation , Protein Multimerization , Protein Subunits/chemistry , Protein Subunits/metabolism , Protein Transport , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...