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

Publication year range
1.
Mol Cell ; 61(1): 125-37, 2016 Jan 07.
Article in English | MEDLINE | ID: mdl-26711008

ABSTRACT

Influenza virus polymerase transcribes or replicates the segmented RNA genome (vRNA) into respectively viral mRNA or full-length copies and initiates RNA synthesis by binding the conserved 3' and 5' vRNA ends (the promoter). In recent structures of promoter-bound polymerase, the cap-binding and endonuclease domains are configured for cap snatching, which generates capped transcription primers. Here, we present a FluB polymerase structure with a bound complementary cRNA 5' end that exhibits a major rearrangement of the subdomains within the C-terminal two-thirds of PB2 (PB2-C). Notably, the PB2 nuclear localization signal (NLS)-containing domain translocates ∼90 Što bind to the endonuclease domain. FluA PB2-C alone and RNA-free FluC polymerase are similarly arranged. Biophysical and cap-dependent endonuclease assays show that in solution the polymerase explores different conformational distributions depending on which RNA is bound. The inherent flexibility of the polymerase allows it to adopt alternative conformations that are likely important during polymerase maturation into active progeny RNPs.


Subject(s)
Gammainfluenzavirus/enzymology , Influenza A Virus, H5N1 Subtype/enzymology , Influenza B virus/enzymology , RNA-Dependent RNA Polymerase/metabolism , Viral Proteins/metabolism , Amino Acid Sequence , Crystallography, X-Ray , Humans , Influenza A Virus, H5N1 Subtype/genetics , Influenza B virus/genetics , Gammainfluenzavirus/genetics , Lasers , Mass Spectrometry , Models, Molecular , Molecular Sequence Data , Nuclear Localization Signals/metabolism , Protein Interaction Domains and Motifs , RNA, Viral/metabolism , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/genetics , Ribonucleoproteins/metabolism , Scattering, Small Angle , Structure-Activity Relationship , Viral Proteins/chemistry , Viral Proteins/genetics
2.
Angew Chem Int Ed Engl ; 60(9): 4689-4697, 2021 02 23.
Article in English | MEDLINE | ID: mdl-33320993

ABSTRACT

Fatty acid ß-oxidation (FAO) and oxidative phosphorylation (OXPHOS) are mitochondrial redox processes that generate ATP. The biogenesis of the respiratory Complex I, a 1 MDa multiprotein complex that is responsible for initiating OXPHOS, is mediated by assembly factors including the mitochondrial complex I assembly (MCIA) complex. However, the organisation and the role of the MCIA complex are still unclear. Here we show that ECSIT functions as the bridging node of the MCIA core complex. Furthermore, cryo-electron microscopy together with biochemical and biophysical experiments reveal that the C-terminal domain of ECSIT directly binds to the vestigial dehydrogenase domain of the FAO enzyme ACAD9 and induces its deflavination, switching ACAD9 from its role in FAO to an MCIA factor. These findings provide the structural basis for the MCIA complex architecture and suggest a unique molecular mechanism for coordinating the regulation of the FAO and OXPHOS pathways to ensure an efficient energy production.


Subject(s)
Electron Transport Complex I/chemistry , Flavin-Adenine Dinucleotide/metabolism , Mitochondria/metabolism , Acyl-CoA Dehydrogenases/genetics , Acyl-CoA Dehydrogenases/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Cryoelectron Microscopy , Electron Transport Complex I/metabolism , Energy Metabolism , Flavin-Adenine Dinucleotide/chemistry , Humans , Oxidative Phosphorylation , Protein Interaction Domains and Motifs , Protein Structure, Tertiary , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification
3.
J Biol Chem ; 292(16): 6468-6477, 2017 04 21.
Article in English | MEDLINE | ID: mdl-28249940

ABSTRACT

Disrupted in Schizophrenia 1 (DISC1) is a scaffolding protein of significant importance for neurodevelopment and a prominent candidate protein in the pathology of major mental illness. DISC1 modulates a number of critical neuronal signaling pathways through protein-protein interactions; however, the mechanism by which this occurs and how DISC1 causes mental illness is unclear, partly because knowledge of the structure of DISC1 is lacking. A lack of homology with known proteins has hindered attempts to define its domain composition. Here, we employed the high-throughput Expression of Soluble Proteins by Random Incremental Truncation (ESPRIT) technique to identify discretely folded regions of human DISC1 via solubility assessment of tens of thousands of fragments of recombinant DISC1. We identified four novel structured regions, named D, I, S, and C, at amino acids 257-383, 539-655, 635-738, and 691-836, respectively. One region (D) is located in a DISC1 section previously predicted to be unstructured. All regions encompass coiled-coil or α-helical structures, and three are involved in DISC1 oligomerization. Crucially, three of these domains would be lost or disrupted by a chromosomal translocation event after amino acid 597, which has been strongly linked to major mental illness. Furthermore, we observed that a known illness-related frameshift mutation after amino acid 807 causes the C region to form aberrantly multimeric and aggregated complexes with an unstable secondary structure. This newly revealed domain architecture of DISC1, therefore, provides a powerful framework for understanding the critical role of this protein in a variety of devastating mental illnesses.


Subject(s)
Mutation , Nerve Tissue Proteins/chemistry , Psychotic Disorders/genetics , Schizophrenia/genetics , Frameshift Mutation , Humans , Nerve Tissue Proteins/genetics , Neurons/metabolism , Protein Denaturation , Protein Domains , Protein Folding , Protein Interaction Mapping , Protein Structure, Secondary , Recombinant Proteins/chemistry , Signal Transduction
4.
Biochem Biophys Res Commun ; 497(4): 978-982, 2018 03 18.
Article in English | MEDLINE | ID: mdl-29448102

ABSTRACT

Currently, the most widely used strategies for molecular cloning are sticky-end ligation-based cloning, TA cloning, blunt-end ligation-based cloning and ligase-independent cloning. In this study we have developed a novel mini-vector pANY1 which can simultaneously meet the requirements of all these cloning strategies. In addition, the selection of appropriate restriction digestion sites is difficult in some cases because of the presence of internal sites. In this study, an annealing of PCR products (APP)-based sticky-end cloning strategy was introduced to avoid this issue. Additionally, false positives occur during molecular cloning, which increases the workload of isolating positive clones. The plasmid pANY1 contains a ccdB cassette between multiple cloning sites, which efficiently avoids these false positives. Therefore, this mini-vector should serve as a useful tool with wide applications in biosciences, agriculture, food technologies, etc.


Subject(s)
Cloning, Molecular/methods , Genetic Vectors , Ligation , Methods , Plasmids , Polymerase Chain Reaction
5.
J Struct Biol ; 198(1): 19-27, 2017 04.
Article in English | MEDLINE | ID: mdl-28268178

ABSTRACT

IcsA/VirG is a key virulence factor of the human pathogen Shigella flexneri, acting as both an adhesin and actin-polymerizing factor during infection. We identified a soluble expression construct of the IcsA/VirG α-domain using the ESPRIT library screening system and determined its structure to 1.9Å resolution. In addition to the previously characterized autochaperone domain, our structure reveals a new domain, which shares a common fold with the autochaperone domains of various autotransporters. We further provide insight into the previously structurally uncharacterized ß-helix domain that harbors the polar targeting motif and passenger-associated transport repeat. This structure is the first of any member of the recently identified passenger-associated transport repeat-containing autotransporters. Thus, it provides new insights into the overall architecture of this class of autotransporters, the function of the identified additional autochaperone domain and the structural properties of motifs involved in polar targeting and secretion of the Shigella flexneri virulence factor IcsA/VirG.


Subject(s)
Bacterial Proteins/chemistry , DNA-Binding Proteins/chemistry , Shigella flexneri/pathogenicity , Transcription Factors/chemistry , Type V Secretion Systems/metabolism , Virulence Factors/chemistry , Amino Acid Motifs , Bacterial Proteins/metabolism , DNA-Binding Proteins/metabolism , Humans , Molecular Chaperones/metabolism , Molecular Structure , Protein Domains , Protein Transport , Transcription Factors/metabolism
6.
J Biol Chem ; 289(20): 13903-11, 2014 May 16.
Article in English | MEDLINE | ID: mdl-24634216

ABSTRACT

Mammalian Rif1 is a key regulator of DNA replication timing, double-stranded DNA break repair, and replication fork restart. Dissecting the molecular functions of Rif1 is essential to understand how it regulates such diverse processes. However, Rif1 is a large protein that lacks well defined functional domains and is predicted to be largely intrinsically disordered; these features have hampered recombinant expression of Rif1 and subsequent functional characterization. Here we applied ESPRIT (expression of soluble proteins by random incremental truncation), an in vitro evolution-like approach, to identify high yielding soluble fragments encompassing conserved regions I and II (CRI and CRII) at the C-terminal region of murine Rif1. NMR analysis showed CRI to be intrinsically disordered, whereas CRII is partially folded. CRII binds cruciform DNA with high selectivity and micromolar affinity and thus represents a functional DNA binding domain. Mutational analysis revealed an α-helical region of CRII to be important for cruciform DNA binding and identified critical residues. Thus, we present the first structural study of the mammalian Rif1, identifying a domain that directly links its function to DNA binding. The high specificity of Rif1 for cruciform structures is significant given the role of this key protein in regulating origin firing and DNA repair.


Subject(s)
Biophysical Phenomena , DNA, Cruciform/metabolism , Telomere-Binding Proteins/chemistry , Telomere-Binding Proteins/metabolism , Amino Acid Sequence , Animals , Base Sequence , DNA, Cruciform/genetics , Mice , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Solubility , Substrate Specificity
7.
J Am Chem Soc ; 137(48): 15122-34, 2015 Dec 09.
Article in English | MEDLINE | ID: mdl-26424125

ABSTRACT

Influenza A RNA polymerase complex is formed from three components, PA, PB1, and PB2. PB2 is independently imported into the nucleus prior to polymerase reconstitution. All crystallographic structures of the PB2 C-terminus (residues 536-759) reveal two globular domains, 627 and NLS, that form a tightly packed heterodimer. The molecular basis of the affinity of 627-NLS for importins remained unclear from these structures, apparently requiring large-scale conformational changes prior to importin binding. Using a combination of solution-state NMR, small-angle neutron scattering, small-angle X-ray scattering (SAXS), and Förster resonance energy transfer (FRET), we show that 627-NLS populates a temperature-dependent dynamic equilibrium between closed and open states. The closed state is stabilized by a tripartite salt bridge involving the 627-NLS interface and the linker, that becomes flexible in the open state, with 627 and NLS dislocating into a highly dynamic ensemble. Activation enthalpies and entropies associated with the rupture of this interface were derived from simultaneous analysis of temperature-dependent chemical exchange saturation transfer measurements, revealing a strong temperature dependence of both open-state population and exchange rate. Single-molecule FRET and SAXS demonstrate that only the open-form is capable of binding to importin α and that, upon binding, the 627 domain samples a dynamic conformational equilibrium in the vicinity of the C-terminus of importin α. This intrinsic large-scale conformational flexibility therefore enables 627-NLS to bind importin through conformational selection from a temperature-dependent equilibrium comprising both functional forms of the protein.


Subject(s)
Influenza A Virus, H5N1 Subtype/enzymology , Karyopherins/metabolism , Viral Proteins/metabolism , Crystallography, X-Ray , Fluorescence Resonance Energy Transfer , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Protein Conformation , Solutions , Viral Proteins/chemistry
8.
Nature ; 458(7240): 914-8, 2009 Apr 16.
Article in English | MEDLINE | ID: mdl-19194459

ABSTRACT

The influenza virus polymerase, a heterotrimer composed of three subunits, PA, PB1 and PB2, is responsible for replication and transcription of the eight separate segments of the viral RNA genome in the nuclei of infected cells. The polymerase synthesizes viral messenger RNAs using short capped primers derived from cellular transcripts by a unique 'cap-snatching' mechanism. The PB2 subunit binds the 5' cap of host pre-mRNAs, which are subsequently cleaved after 10-13 nucleotides by the viral endonuclease, hitherto thought to reside in the PB2 (ref. 5) or PB1 (ref. 2) subunits. Here we describe biochemical and structural studies showing that the amino-terminal 209 residues of the PA subunit contain the endonuclease active site. We show that this domain has intrinsic RNA and DNA endonuclease activity that is strongly activated by manganese ions, matching observations reported for the endonuclease activity of the intact trimeric polymerase. Furthermore, this activity is inhibited by 2,4-dioxo-4-phenylbutanoic acid, a known inhibitor of the influenza endonuclease. The crystal structure of the domain reveals a structural core closely resembling resolvases and type II restriction endonucleases. The active site comprises a histidine and a cluster of three acidic residues, conserved in all influenza viruses, which bind two manganese ions in a configuration similar to other two-metal-dependent endonucleases. Two active site residues have previously been shown to specifically eliminate the polymerase endonuclease activity when mutated. These results will facilitate the optimisation of endonuclease inhibitors as potential new anti-influenza drugs.


Subject(s)
Endonucleases/metabolism , Influenza A Virus, H3N2 Subtype/enzymology , Protein Subunits/chemistry , Protein Subunits/metabolism , RNA Caps/metabolism , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/metabolism , Viral Proteins/chemistry , Viral Proteins/metabolism , Amino Acid Sequence , Animals , Catalytic Domain , Endonucleases/chemistry , Enzyme Stability , Histidine/metabolism , Humans , Influenza A Virus, H5N1 Subtype/enzymology , Gammainfluenzavirus/enzymology , Manganese/metabolism , Manganese/pharmacology , Models, Molecular , Molecular Sequence Data
9.
Nature ; 461(7264): 664-8, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19794495

ABSTRACT

A key step in many chromatin-related processes is the recognition of histone post-translational modifications by effector modules such as bromodomains and chromo-like domains of the Royal family. Whereas effector-mediated recognition of single post-translational modifications is well characterized, how the cell achieves combinatorial readout of histones bearing multiple modifications is poorly understood. One mechanism involves multivalent binding by linked effector modules. For example, the tandem bromodomains of human TATA-binding protein-associated factor-1 (TAF1) bind better to a diacetylated histone H4 tail than to monoacetylated tails, a cooperative effect attributed to each bromodomain engaging one acetyl-lysine mark. Here we report a distinct mechanism of combinatorial readout for the mouse TAF1 homologue Brdt, a testis-specific member of the BET protein family. Brdt associates with hyperacetylated histone H4 (ref. 7) and is implicated in the marked chromatin remodelling that follows histone hyperacetylation during spermiogenesis, the stage of spermatogenesis in which post-meiotic germ cells mature into fully differentiated sperm. Notably, we find that a single bromodomain (BD1) of Brdt is responsible for selectively recognizing histone H4 tails bearing two or more acetylation marks. The crystal structure of BD1 bound to a diacetylated H4 tail shows how two acetyl-lysine residues cooperate to interact with one binding pocket. Structure-based mutagenesis that reduces the selectivity of BD1 towards diacetylated tails destabilizes the association of Brdt with acetylated chromatin in vivo. Structural analysis suggests that other chromatin-associated proteins may be capable of a similar mode of ligand recognition, including yeast Bdf1, human TAF1 and human CBP/p300 (also known as CREBBP and EP300, respectively). Our findings describe a new mechanism for the combinatorial readout of histone modifications in which a single effector module engages two marks on a histone tail as a composite binding epitope.


Subject(s)
Histones/chemistry , Histones/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Acetylation , Allosteric Regulation , Animals , Binding Sites , COS Cells , Chlorocebus aethiops , Chromatin/chemistry , Chromatin/metabolism , Crystallography, X-Ray , Lysine/metabolism , Mice , Models, Molecular , Nuclear Proteins/genetics , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Substrate Specificity
10.
J Biol Chem ; 287(3): 2022-31, 2012 Jan 13.
Article in English | MEDLINE | ID: mdl-22130666

ABSTRACT

The human genome encodes six isoforms of importin α that show greater than 60% sequence similarity and remarkable substrate specificity. The isoform importin α5 can bind phosphorylated cargos such as STAT1 and Epstein-Barr Virus Nuclear Antigen 1, as well as the influenza virus polymerase subunit PB2. In this work, we have studied the interaction of the nucleoporin Nup50 with importin α5. We show that the first 47 residues of Nup50 bind to the C terminus of importin α5 like a "clip," stabilizing the closed conformation of ARM 10. In vitro, Nup50 binds with high affinity either to empty importin α5 or to a preassembled complex of importin α5 bound to the C-terminal domain of the import cargo PB2, resulting in a trimeric complex. By contrast, PB2 can only bind with high affinity to importin α5 in the absence of Nup50. This suggests that Nup50 primary function may not be to actively displace the import cargo from importin α5 but rather to prevent cargo rebinding in preparation for recycling. This is the first evidence for a nucleoporin modulating the import reaction by directly altering the three-dimensional structure of an import adaptor.


Subject(s)
Cell Nucleus/chemistry , Nuclear Pore Complex Proteins/chemistry , Nuclear Proteins/chemistry , alpha Karyopherins/chemistry , Active Transport, Cell Nucleus/physiology , Cell Nucleus/genetics , Cell Nucleus/metabolism , Humans , Nuclear Pore Complex Proteins/genetics , Nuclear Pore Complex Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Binding , Protein Structure, Quaternary , Protein Structure, Tertiary , alpha Karyopherins/genetics , alpha Karyopherins/metabolism
11.
Proc Natl Acad Sci U S A ; 107(37): 16078-83, 2010 Sep 14.
Article in English | MEDLINE | ID: mdl-20805464

ABSTRACT

During viral replication, herpesviruses package their DNA into the procapsid by means of the terminase protein complex. In human cytomegalovirus (herpesvirus 5), the terminase is composed of subunits UL89 and UL56. UL89 cleaves the long DNA concatemers into unit-length genomes of appropriate length for encapsidation. We used ESPRIT, a high-throughput screening method, to identify a soluble purifiable fragment of UL89 from a library of 18,432 randomly truncated ul89 DNA constructs. The purified protein was crystallized and its three-dimensional structure was solved. This protein corresponds to the key nuclease domain of the terminase and shows an RNase H/integrase-like fold. We demonstrate that UL89-C has the capacity to process the DNA and that this function is dependent on Mn(2+) ions, two of which are located at the active site pocket. We also show that the nuclease function can be inactivated by raltegravir, a recently approved anti-AIDS drug that targets the HIV integrase.


Subject(s)
Cytomegalovirus/enzymology , DNA Packaging , DNA, Viral/chemistry , Viral Proteins/chemistry , Catalytic Domain , Crystallography, X-Ray , Cytomegalovirus/genetics , DNA, Viral/genetics , Models, Molecular , Mutation , Protein Structure, Tertiary , Viral Proteins/genetics
12.
J Biol Chem ; 286(12): 10439-48, 2011 Mar 25.
Article in English | MEDLINE | ID: mdl-21216958

ABSTRACT

In the adaptation of avian viruses to mammalian hosts, mutations in the viral polymerase, notably in the PB2 subunit, play an important role. A PB2 C-terminal domain rich in putative host adaptation residues has been shown to bind importin α nuclear import receptors. Adaptation has been proposed to involve binding of PB2 to importins of the new host. To date PB2-importin complexes have been characterized semiquantitatively with no precise measurement of binding parameters. To investigate the effects of adaptive mutations on importin interaction and selectivity, surface plasmon resonance was used to compare the binding rate constants and affinities of avian H5N1 and human H3N2 PB2 C-terminal variants with importin isoforms human α 1, 3, 5 and 7, and avian α 1. Using purified proteins eliminates host environment effects and permits measurement of intrinsic affinities and rates of complex formation and dissociation. Two effects were observed: first, adaptive mutations D701N, R702K, and S714R in the nuclear localization signal domain increased 2-4-fold the association rates with avian and human importins; second, measurement of different structural forms of the PB2 C terminus demonstrated that the upstream 627 domain reduced binding affinity, consistent with a steric clash predicted from crystal structures. From these kinetic data, structural analyses, and the data of others, a model is proposed in which an increase in charged surface residues during host adaptation increases the association rate of PB2 to cytoplasmic importins and where the C-terminal 627-nuclear localization signal domain may reorganize upon importin binding, consistent with a role in active polymerase assembly.


Subject(s)
Adaptation, Physiological/physiology , Host-Pathogen Interactions/physiology , Influenza A Virus, H3N2 Subtype/enzymology , Influenza A Virus, H5N1 Subtype/enzymology , RNA-Dependent RNA Polymerase/chemistry , Viral Proteins/chemistry , alpha Karyopherins/chemistry , Amino Acid Substitution , Cell Count , Humans , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/immunology , Models, Molecular , Mutation, Missense , Protein Binding/physiology , Protein Structure, Tertiary , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism , alpha Karyopherins/genetics , alpha Karyopherins/metabolism
13.
Nat Struct Mol Biol ; 14(3): 229-33, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17310249

ABSTRACT

The trimeric influenza virus polymerase, comprising subunits PA, PB1 and PB2, is responsible for transcription and replication of the segmented viral RNA genome. Using a novel library-based screening technique called expression of soluble proteins by random incremental truncation (ESPRIT), we identified an independently folded C-terminal domain from PB2 and determined its solution structure by NMR. Using green fluorescent protein fusions, we show that both the domain and the full-length PB2 subunit are efficiently imported into the nucleus dependent on a previously overlooked bipartite nuclear localization sequence (NLS). The crystal structure of the domain complexed with human importin alpha5 shows how the last 20 residues unfold to permit binding to the import factor. The domain contains three surface residues implicated in adaptation from avian to mammalian hosts. One of these tethers the NLS-containing peptide to the core of the domain in the unbound state.


Subject(s)
Active Transport, Cell Nucleus , Cell Nucleus/metabolism , Orthomyxoviridae/enzymology , Protein Subunits/chemistry , Protein Subunits/metabolism , Viral Proteins/chemistry , Viral Proteins/metabolism , Amino Acid Sequence , Cell Survival , Crystallography, X-Ray , Humans , Magnetic Resonance Spectroscopy , Molecular Sequence Data , Nuclear Localization Signals , Protein Structure, Secondary , Protein Structure, Tertiary , Solubility , Solutions , alpha Karyopherins/chemistry
14.
Methods ; 55(1): 38-43, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21723393

ABSTRACT

When expression or crystallisation of a protein target in its wild-type full-length form proves problematic, a common strategy is to divide it into subconstructs comprising one or more domains. Rational construct design is not always successful, especially with targets for which there are few similar sequences to generate multiple sequence alignments. Even when this is possible, expression constructs may still fail to yield soluble protein, commonly expressing insolubly or at unusable yields. To address this, several new methods have been described that borrow concepts from the field of directed evolution whereby a random library is generated encompassing construct diversity; this is then screened to identify soluble constructs empirically. Here, we review progress in this area.


Subject(s)
Automation, Laboratory/methods , Gene Library , Genetic Vectors/chemistry , High-Throughput Screening Assays , Proteins/chemistry , Cloning, Molecular , DNA Fragmentation , Deoxyribonucleases , Escherichia coli , Gene Expression , Genes, Reporter , Genetic Vectors/genetics , Green Fluorescent Proteins/analysis , Humans , Mutagenesis , Proteins/genetics , Proteins/metabolism , Solubility
15.
J Struct Biol ; 175(2): 189-97, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21515383

ABSTRACT

Here we present ORF-selector ESPRIT, a 9-fold enhanced version of our technology for screening incremental truncation libraries to identify soluble high yielding constructs of challenging proteins. Gene fragments are truncated at both termini to access internal domains and the resulting reading frame problem is addressed by an unbiased, intein-based open reading frame selection yielding only in-frame DNA inserts. This enriched library is then subcloned into a standard high-level expression plasmid where tens of thousands of constructs can be assayed in a two-step process using colony- and liquid-handling robots to isolate rare highly expressing clones useful for production of multi milligram quantities of purifiable proteins. The p85α protein was used to benchmark the system resulting in isolation of all known domains, either alone or in tandem. The human kinase IKK1 was then screened resulting in purification of a predicted internal domain. This strategy provides an integrated, facile route to produce soluble proteins from challenging and poorly understood target genes at quantities compatible with structural biology, screening applications and immunisation studies. The high genetic diversity that can be sampled opens the way to study more diverse systems including multisubunit complexes.


Subject(s)
Cloning, Molecular/methods , Gene Library , Open Reading Frames , Peptide Fragments/genetics , Recombinant Proteins/genetics , Automation, Laboratory , Class Ia Phosphatidylinositol 3-Kinase/biosynthesis , Class Ia Phosphatidylinositol 3-Kinase/chemistry , Class Ia Phosphatidylinositol 3-Kinase/genetics , Escherichia coli/genetics , Genetic Vectors , Humans , I-kappa B Kinase/biosynthesis , I-kappa B Kinase/chemistry , I-kappa B Kinase/genetics , Peptide Fragments/biosynthesis , Peptide Fragments/chemistry , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Reference Values , Solubility
16.
J Biol Chem ; 285(37): 28411-7, 2010 Sep 10.
Article in English | MEDLINE | ID: mdl-20538599

ABSTRACT

The heterotrimeric RNA-dependent RNA polymerase of influenza viruses catalyzes RNA replication and transcription activities in infected cell nuclei. The nucleotide polymerization activity is common to both replication and transcription processes, with an additional cap-snatching function being employed during transcription to steal short 5'-capped RNA primers from host mRNAs. Cap-binding, endonuclease, and polymerase activities have long been studied biochemically, but structural studies on the polymerase and its subunits have been hindered by difficulties in producing sufficient quantities of material. Recently, because of heightened effort and advances in expression and crystallization technologies, a series of high resolution structures of individual domains have been determined. These shed light on intrinsic activities of the polymerase, including cap snatching, subunit association, and nucleocytoplasmic transport, and open up the possibility of structure-guided development of new polymerase inhibitors. Furthermore, the activity of influenza polymerase is highly host- and cell type-specific, being dependent on the identity of a few key amino acid positions in the different subunits, especially in the C-terminal region of PB2. New structures demonstrate the surface exposure of these residues, consistent with ideas that they might modulate interactions with host-specific factors that enhance or restrict activity. Recent proteomic and genome-wide interactome and RNA interference screens have suggested the identities of some of these potential regulators of polymerase function.


Subject(s)
Influenza A virus/enzymology , RNA-Directed DNA Polymerase/chemistry , Active Transport, Cell Nucleus , Animals , Catalytic Domain , Cell Nucleus/enzymology , Cell Nucleus/virology , Crystallography, X-Ray , Humans , Protein Structure, Quaternary , RNA Caps/biosynthesis , RNA, Viral/biosynthesis , RNA-Directed DNA Polymerase/metabolism , Structure-Activity Relationship , Transcription, Genetic , Virus Replication/physiology
17.
J Rheumatol ; 48(6): 867-876, 2021 06.
Article in English | MEDLINE | ID: mdl-33132218

ABSTRACT

OBJECTIVE: The modified Rodnan skin score (mRSS) remains the preferred method for skin assessment in systemic sclerosis (SSc). There are concerns regarding high interobserver variability of mRSS and negative clinical trials utilizing mRSS as the primary endpoint. High-frequency ultrasound (HFUS) allows objective assessment of cutaneous fibrosis in SSc. We investigated the relationship between HFUS with both mRSS and dermal collagen. METHODS: Skin thickness (ST), echogenicity, and novel shear wave elastography (SWE) were assessed in 53 patients with SSc and 15 healthy controls (HCs) at the finger, hand, forearm, and abdomen. The relationship between HFUS parameters with mRSS (n = 53) and dermal collagen (10 patients with SSc and 10 HCs) was investigated. Intraobserver repeatability of HFUS was calculated using intraclass correlation coefficients (ICCs). RESULTS: HFUS assessment of ST (hand/forearm) and SWE (finger/hand) correlated with local mRSS at some sites. Subclinical abnormalities in ST, echogenicity, and SWE were present in clinically uninvolved SSc skin. Additionally, changes in echogenicity and SWE were sometimes apparent despite objectively normal ST on HFUS. ST, SWE, and local mRSS correlated strongly with collagen quantification (r = 0.697, 0.709, 0.649, respectively). Intraobserver repeatability was high for all HFUS parameters (ICCs for ST = 0.946-0.978; echogenicity = 0.648-0.865; and SWE = 0.953-0.973). CONCLUSION: Our data demonstrate excellent reproducibility and reassuring convergent validity with dermal collagen content. Detection of subclinical abnormalities is an additional benefit of HFUS. The observed correlations with collagen quantification support further investigation of HFUS as an alternative to mRSS in clinical trial settings.


Subject(s)
Scleroderma, Systemic , Collagen , Hand/diagnostic imaging , Humans , Reproducibility of Results , Scleroderma, Systemic/diagnostic imaging , Skin/diagnostic imaging , Ultrasonography
18.
J Struct Biol ; 172(1): 66-74, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20206698

ABSTRACT

Expression of sufficient quantities of soluble protein for structural biology and other applications is often a very difficult task, especially when multimilligram quantities are required. In order to improve yield, solubility or crystallisability of a protein, it is common to subclone shorter genetic constructs corresponding to single- or multi-domain fragments. However, it is not always clear where domain boundaries are located, especially when working on novel targets with little or no sequence similarity to other proteins. Several methods have been described employing aspects of directed evolution to the recombinant expression of challenging proteins. These combine the construction of a random library of genetic constructs of a target with a screening or selection process to identify solubly expressing protein fragments. Here we review several datasets from the ESPRIT (Expression of Soluble Proteins by Random Incremental Truncation) technology to provide a view on its capabilities. Firstly, we demonstrate how it functions using the well-characterised NF-kappaB p50 transcription factor as a model system. Secondly, application of ESPRIT to the challenging PB2 subunit of influenza polymerase has led to several novel atomic resolution structures; here we present an overview of the screening phase of that project. Thirdly, analysis of the human kinase TBK1 is presented to show how the ESPRIT technology rapidly addresses the compatibility of challenging targets with the Escherichia coli expression system.


Subject(s)
Biotechnology/methods , Gene Library , Proteins/genetics , Humans , NF-kappa B p50 Subunit/chemistry , NF-kappa B p50 Subunit/genetics , NF-kappa B p50 Subunit/metabolism , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , Proteins/chemistry , Proteins/metabolism , Reproducibility of Results , Solubility , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/metabolism
19.
PLoS Pathog ; 4(8): e1000136, 2008 Aug 29.
Article in English | MEDLINE | ID: mdl-18769709

ABSTRACT

Understanding how avian influenza viruses adapt to human hosts is critical for the monitoring and prevention of future pandemics. Host specificity is determined by multiple sites in different viral proteins, and mutation of only a limited number of these sites can lead to inter-species transmission. Several of these sites have been identified in the viral polymerase, the best characterised being position 627 in the PB2 subunit. Efficient viral replication at the relatively low temperature of the human respiratory tract requires lysine 627 rather than the glutamic acid variant found systematically in avian viruses. However, the molecular mechanism by which any of these host specific sites determine host range are unknown, although adaptation to host factors is frequently evoked. We used ESPRIT, a library screening method, to identify a new PB2 domain that contains a high density of putative host specific sites, including residue 627. The X-ray structure of this domain (denoted the 627-domain) exhibits a novel fold with the side-chain of Lys627 solvent exposed. The structure of the K627E mutated domain shows no structural differences but the charge reversal disrupts a striking basic patch on the domain surface. Five other recently proposed host determining sites of PB2 are also located on the 627-domain surface. The structure of the complete C-terminal region of PB2 comprising the 627-domain and the previously identified NLS-domain, which binds the host nuclear import factor importin alpha, was also determined. The two domains are found to pack together with a largely hydrophilic interface. These data enable a three-dimensional mapping of approximately half of PB2 sites implicated in cross-species transfer onto a single structural unit. Their surface location is consistent with roles in interactions with other viral proteins or host factors. The identification and structural characterization of these well-defined PB2 domains will help design experiments to elucidate the effects of mutations on polymerase-host factor interactions.


Subject(s)
Adaptation, Physiological , Amino Acid Substitution , Influenza A Virus, H3N2 Subtype/enzymology , Mutation, Missense , RNA-Dependent RNA Polymerase/chemistry , Viral Proteins/chemistry , Crystallography, X-Ray , Host-Pathogen Interactions , Humans , Influenza A Virus, H3N2 Subtype/genetics , Influenza, Human/enzymology , Influenza, Human/genetics , Influenza, Human/metabolism , Protein Structure, Tertiary/genetics , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism , alpha Karyopherins/chemistry , alpha Karyopherins/metabolism
20.
Virologie (Montrouge) ; 14(6): 435-445, 2010 Dec 01.
Article in French | MEDLINE | ID: mdl-36151627

ABSTRACT

Influenza virus polymerase transcribes and replicates the viral RNA genome within the context of a ribonucleoprotein complex that has been hitherto remarkably intractable to structural analysis. In the last three years, crystal structures of independent domains covering roughly half of the heterotrimeric polymerase have been determined. These include the cap-binding and endonuclease domains, critical for the unique cap-snatching mechanism of mRNA transcription, and the major inter-subunit interfaces. In addition a cryoelectron microscopy structure of the entire ribonucleoprotein complex has been determined opening the way to the construction of a quasi-atomic model of the influenza replication machinery. These results provide the first detailed structure-function insights into polymerase assembly, transcription and host adaptation and will have an impact on anti-influenza drug design.

SELECTION OF CITATIONS
SEARCH DETAIL