Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 131
Filter
1.
mBio ; 12(6): e0224121, 2021 12 21.
Article in English | MEDLINE | ID: mdl-34809451

ABSTRACT

Current seasonal influenza virus vaccines do not induce robust immune responses to neuraminidase. Several factors, including immunodominance of hemagglutinin over neuraminidase, instability of neuraminidase in vaccine formulations, and variable, nonstandardized amounts of neuraminidase in the vaccines, may contribute to this effect. However, vaccines that induce strong antineuraminidase immune responses would be beneficial, as they are highly protective. Furthermore, antigenic drift is slower for neuraminidase than for hemagglutinin, potentially providing broader coverage. Here, we designed stabilized recombinant versions of neuraminidase by replacing the N-terminal cytoplasmic domain, transmembrane, and extracellular stalk with tetramerization domains from the measles or Sendai virus phosphoprotein or from an Arabidopsis thaliana transcription factor. The measles virus tetramerization domain-based construct, termed N1-MPP, was chosen for further evaluation, as it retained antigenicity, neuraminidase activity, and structural integrity and provided robust protection in vivo against lethal virus challenge in the mouse model. We tested N1-MPP as a standalone vaccine, admixed with seasonal influenza virus vaccines, or given with seasonal influenza virus vaccines but in the other leg of the mouse. Admixture with different formulations of seasonal vaccines led to a weak neuraminidase response, suggesting a dominant effect of hemagglutinin over neuraminidase when administered in the same formulation. However, administration of neuraminidase alone or with seasonal vaccine administered in the alternate leg of the mouse induced robust antibody responses. Thus, this recombinant neuraminidase construct is a promising vaccine antigen that may enhance and broaden protection against seasonal influenza viruses. IMPORTANCE Influenza virus infections remain a high risk to human health, causing up to 650,000 deaths worldwide every year, with an enormous burden on the health care system. Since currently available seasonal vaccines are only partially effective and often mismatched to the circulating strains, a broader protective influenza virus vaccine is needed. Here, we generated a recombinant influenza virus vaccine candidate based on the more conserved neuraminidase surface glycoprotein in order to induce a robust and broader protective immune response against a variety of circulating influenza virus strains.


Subject(s)
Influenza Vaccines/immunology , Influenza, Human/prevention & control , Measles virus/immunology , Neuraminidase/immunology , Phosphoproteins/immunology , Viral Proteins/immunology , Amino Acid Sequence , Animals , Antibodies, Viral/immunology , Antigenic Drift and Shift , Cross Reactions , Humans , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H1N1 Subtype/physiology , Influenza Vaccines/administration & dosage , Influenza Vaccines/chemistry , Influenza Vaccines/genetics , Influenza, Human/immunology , Influenza, Human/virology , Measles virus/chemistry , Measles virus/genetics , Mice , Mice, Inbred BALB C , Neuraminidase/administration & dosage , Neuraminidase/chemistry , Neuraminidase/genetics , Phosphoproteins/chemistry , Phosphoproteins/genetics , Protein Domains , Sequence Alignment , Vaccination , Viral Proteins/administration & dosage , Viral Proteins/chemistry , Viral Proteins/genetics
2.
Curr Opin Virol ; 41: 59-67, 2020 04.
Article in English | MEDLINE | ID: mdl-32570195

ABSTRACT

The measles virus replication complex represents a potentially important, but as yet relatively unexplored target for viral inhibition. Little is known about the molecular mechanisms that underpin replication and transcription in paramyxoviruses. In recent years it has become clear that conformational dynamics play an important role in paramyxoviral replication, and that a complete understanding of the viral cycle requires a description of the structural plasticity of the different components. Here, we review recent progress in this direction, covering the dynamics of the nucleocapsid assembly process, high resolution structure and dynamics of protein:RNA interactions, and the investigation of the role of intrinsic conformational disorder in pre-assembly nucleoprotein/phosphoprotein complexes. Finally, we discuss the role of viral factories in the form of phase-separated membraneless organelles formed by measles virus phospho and nucleoproteins that promote the assembly of nucleocapsid structures.


Subject(s)
Measles virus/physiology , Measles/virology , Nucleocapsid/chemistry , RNA, Viral/genetics , Virus Replication , Animals , Humans , Measles virus/chemistry , Measles virus/genetics , Nucleocapsid/genetics , Nucleocapsid/metabolism , Nucleoproteins/chemistry , Nucleoproteins/genetics , Nucleoproteins/metabolism , RNA, Viral/chemistry , RNA, Viral/metabolism
3.
Curr Opin Virol ; 41: 52-58, 2020 04.
Article in English | MEDLINE | ID: mdl-32413678

ABSTRACT

Measles virus, a member of the genus Morbillivirus, is highly contagious and still shows considerable mortality with over 100000 deaths annually, although efficient attenuated vaccines exist. Recent studies of measles virus haemagglutinin (MeV-H) and its receptor, including crystallographic and electron microscopic structural analyses combined with functional assays, have revealed how the MeV-H protein recognizes its cognate receptors, SLAM and Nectin-4, and how the glycan shield ensures effective vaccination. In addition, the crystal structure of the MeV-F protein indicated its similarity to those of other paramyxoviruses. Taking into account these data, several models of viral entry/membrane fusion of measles viruses and related paramyxoviruses have been proposed. Furthermore, anti-MeV-F inhibitors targeted to specific regions to inhibit MeV-F protein activation were reported, with potency for preventing MeV infection. The inhibitors targeted for entry events may potentially be applied to treatment of MeV-derived diseases, although escape mutations and drug profiles should be considered.


Subject(s)
Hemagglutinins, Viral/chemistry , Measles virus/physiology , Measles/virology , Virus Internalization , Animals , Hemagglutinins, Viral/genetics , Hemagglutinins, Viral/metabolism , Humans , Measles/genetics , Measles/metabolism , Measles virus/chemistry , Measles virus/genetics , Protein Binding , Receptors, Virus/chemistry , Receptors, Virus/genetics , Receptors, Virus/metabolism
4.
J Am Chem Soc ; 142(25): 11092-11101, 2020 06 24.
Article in English | MEDLINE | ID: mdl-32323533

ABSTRACT

Intrinsically disordered proteins (IDPs), which in isolation do not adopt a well-defined tertiary structure but instead populate a structurally heterogeneous ensemble of interconverting states, play important roles in many biological pathways. IDPs often fold into ordered states upon binding to their physiological interaction partners (a so-called "folding-upon-binding" process), but it has proven difficult to obtain an atomic-level description of the structural mechanisms by which they do so. Here, we describe in atomic detail the folding-upon-binding mechanism of an IDP segment to its binding partner, as observed in unbiased molecular dynamics simulations. In our simulations, we observed over 70 binding and unbinding events between the α-helical molecular recognition element (α-MoRE) of the intrinsically disordered C-terminal domain of the measles virus nucleoprotein (NTAIL) and the X domain (XD) of the measles virus phosphoprotein complex. We found that folding-upon-binding primarily occurred through induced-folding pathways (in which intermolecular contacts form before or concurrently with the secondary structure of the disordered protein)-an observation supported by previous experiments-and that the transition state ensemble was characterized by formation of just a few key intermolecular contacts and was otherwise highly structurally heterogeneous. We found that when a large amount of helical content was present early in a transition path, NTAIL typically unfolded and then refolded after additional intermolecular contacts formed. We also found that, among conformations with similar numbers of intermolecular contacts, those with less helical content had a higher probability of ultimately forming the native complex than conformations with more helical content, which were more likely to unbind. These observations suggest that even after intermolecular contacts have formed, disordered regions can have a kinetic advantage over folded regions in the folding-upon-binding process.


Subject(s)
Intrinsically Disordered Proteins/metabolism , Nucleocapsid Proteins/metabolism , Peptide Fragments/metabolism , Phosphoproteins/metabolism , Protein Folding , Intrinsically Disordered Proteins/chemistry , Measles virus/chemistry , Molecular Dynamics Simulation , Nucleocapsid Proteins/chemistry , Peptide Fragments/chemistry , Phosphoproteins/chemistry , Protein Binding , Protein Conformation, alpha-Helical , Protein Domains
5.
Arch Biochem Biophys ; 671: 255-261, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31326517

ABSTRACT

Intrinsically Disordered Proteins (IDPs) are a class of protein that exert their function despite lacking a well-defined three-dimensional structure, which is sometimes achieved only upon binding to their natural ligands. This feature implies the folding of IDPs to be generally coupled with a binding event, representing an interesting challenge for kinetic studies. In this review, we recapitulate some of the most important findings of IDPs binding-induced folding mechanisms obtained by analyzing their binding kinetics. Furthermore, by focusing on the interaction between the Measles virus NTAIL protein, a prototypical IDP, and its physiological partner, the X domain, we recapitulate the major theoretical and experimental approaches that were used to describe binding induced folding.


Subject(s)
Intrinsically Disordered Proteins/metabolism , Measles virus/chemistry , Protein Folding , Viral Proteins/metabolism , Intrinsically Disordered Proteins/chemistry , Kinetics , Protein Binding , Protein Domains , Viral Proteins/chemistry
6.
Comput Biol Chem ; 80: 384-389, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31112822

ABSTRACT

The hemagglutinin (H) protein of measles viruses (MeV) mediates binding to the cellular receptors, CD46,human signaling lymphocyte activation molecule and nectin-4. Vaccine strains primarily contain H-proteins possessing MeV-H: Y481 and can utilize CD46. Reports suggest that a single amino acid change in MeV-H at position 481 in wild type strains renders them inefficient in utilizing CD46. The in-depth molecular mechanism by which substitutions at 481 and another reported critical residue position 546 affects CD46 binding affinity however remains elusive. We used molecular docking studies of CD46 with MeV-H possessing Y481 N/D to understand the in-depth molecular mechanism involved. It was found that loss in either of the hydrogen bond (H-bond) contacts (MeV-H:481-CD46:65, MeV-H:546-CD46:63) in the central contact region prevented efficient CD46 binding. Y481 N could form the specific H-bond, while G546S H-bond could be formed only in conjunction with Y481, revealing the significance of these residues in determining CD46 receptor binding potential. Elucidating the underlying molecular mechanism of receptor usage by the MeV has implications to understanding cellular tropism, viral pathogenesis and therapy.


Subject(s)
Hemagglutinins, Viral/metabolism , Measles virus/chemistry , Membrane Cofactor Protein/metabolism , Receptors, Virus/metabolism , Amino Acid Sequence , Binding Sites , Hemagglutinins, Viral/chemistry , Humans , Hydrogen Bonding , Membrane Cofactor Protein/chemistry , Molecular Docking Simulation , Protein Binding , Receptors, Virus/chemistry
7.
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
8.
ACS Chem Biol ; 14(3): 337-341, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30715849

ABSTRACT

The interaction between NTAIL and XD from the measles virus represents a paradigmatic example of molecular recognition between an intrinsically disordered protein and a folded partner. By binding to XD, a small portion of NTAIL (classically denoted as MoRE) undergoes a disorder-to-order transition, populating an α-helical structure, while the reminder of the protein remains disordered. Here, we demonstrate an unexpected crosstalk between such a disordered region and the adjacent molecular recognition element (MoRE). This result was obtained by producing a series of truncation and site-directed variants of NTAIL while measuring the effects on the kinetics of folding and binding. We show that the disordered region of NTAIL exerts its inhibitory role by slowing the folding step of the MoRE, thereby tuning the affinity of the interaction.


Subject(s)
Intrinsically Disordered Proteins/chemistry , Measles virus/chemistry , Phosphoproteins/chemistry , Viral Proteins/chemistry , Binding Sites , Kinetics , Measles virus/metabolism , Models, Molecular , Protein Binding , Protein Conformation, alpha-Helical , Protein Folding
9.
Structure ; 27(4): 660-668.e4, 2019 04 02.
Article in English | MEDLINE | ID: mdl-30799076

ABSTRACT

Nipah virus is a highly lethal zoonotic pathogen found in Southeast Asia that has caused human encephalitis outbreaks with 40%-70% mortality. NiV encodes its own RNA-dependent RNA polymerase within the large protein, L. Efficient polymerase activity requires the phosphoprotein, P, which tethers L to its template, the viral nucleocapsid. P is a multifunctional protein with modular domains. The central P multimerization domain is composed of a long, tetrameric coiled coil. We investigated the importance of structural features found in this domain for polymerase function using a newly constructed NiV bicistronic minigenome assay. We identified a conserved basic patch and central kink in the coiled coil that are important for polymerase function, with R555 being absolutely essential. This basic patch and central kink are conserved in the related human pathogens measles and mumps viruses, suggesting that this mechanism may be conserved.


Subject(s)
DNA-Directed RNA Polymerases/chemistry , Genome, Viral , Nipah Virus/chemistry , Phosphoproteins/chemistry , RNA-Dependent RNA Polymerase/chemistry , Viral Proteins/chemistry , Amino Acid Sequence , Binding Sites , Cloning, Molecular , Conserved Sequence , Crystallography, X-Ray , DNA-Directed RNA Polymerases/genetics , DNA-Directed RNA Polymerases/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Humans , Measles virus/chemistry , Measles virus/enzymology , Measles virus/genetics , Models, Molecular , Mumps virus/chemistry , Mumps virus/enzymology , Mumps virus/genetics , Nipah Virus/enzymology , Nipah Virus/genetics , Phosphoproteins/genetics , Phosphoproteins/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Interaction Domains and Motifs , Protein Multimerization , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Viral Proteins/genetics , Viral Proteins/metabolism
10.
Virol J ; 15(1): 160, 2018 10 16.
Article in English | MEDLINE | ID: mdl-30326905

ABSTRACT

BACKGROUND: Measles (MEV) and mumps virus (MUV) are enveloped, non-segmented, negative single stranded RNA viruses of the family Paramyxoviridae, and are the cause of measles and mumps, respectively, both preventable by vaccination. Aside from proteins coded by the viral genome, viruses are considered to contain host cell proteins (HCPs). The presence of extracellular vesicles (ECVs), which are often co-purified with viruses due to their similarity in size, density and composition, also contributes to HCPs detected in virus preparations, and this has often been neglected. The aim was to identify which virus-coded proteins are present in MEV and MUV virions, and to try to detect which HCPs, if any, are incorporated inside the virions or adsorbed on their outer surface, and which are more likely to be a contamination from co-purified ECVs. METHODS: MUV, MEV and ECVs were purified by ultracentrifugation, hydrophobic interaction chromatography and immunoaffinity chromatography, proteins in the samples were resolved by SDS-PAGE and subjected to identification by MALDI-TOF/TOF-MS. A comparative analysis of HCPs present in all samples was carried out. RESULTS: By proteomics approach, it was verified that almost all virus-coded proteins are present in MEV and MUV particles. Protein C in MEV which was until now considered to be non-structural viral protein, was found to be present inside the MeV virions. Results on the presence of HCPs in differently purified virus preparations imply that actin, annexins, cyclophilin A, moesin and integrin ß1 are part of the virions. CONCLUSIONS: All HCPs detected in the viruses are present in ECVs as well, indicating their possible function in vesicle formation, or that most of them are only present in ECVs. Only five HCPs were constantly present in purified virus preparations, regardless of the purification method used, implying they are likely the integral part of the virions. The approach described here is helpful for further investigation of HCPs in other virus preparations.


Subject(s)
Measles virus/chemistry , Measles/virology , Mumps virus/chemistry , Mumps/virology , Proteome/analysis , Viral Proteins/analysis , Virion/chemistry , Animals , Chlorocebus aethiops , Hydrophobic and Hydrophilic Interactions , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Vero Cells
11.
J Pharm Biomed Anal ; 161: 73-82, 2018 Nov 30.
Article in English | MEDLINE | ID: mdl-30145452

ABSTRACT

Protein thermal shift assay (TSA) has been extensively used in investigation of protein stabilization (for protein biopharmaceutics stabilization, protein crystallization studies or screening of recombinant proteins) and drug discovery (screening of ligands or inhibitors). This work aimed to analyze thermal shift assay results in comparison to protein polymerization (multimerization and aggregation) propensity and test the most stabilizing formulations for their stabilization effect on enveloped viruses. Influence of protein concentration, buffer pH and molarity was tested on three proteins (immunoglobulin G, ovalbumin, and albumin) and results showed that each of these factors has an impact on determined shift in protein melting point Tm, and the impact was similar for all three proteins. In case of ovalbumin, molecular dynamics simulations were performed with the goal to understanding molecular basis of protein's thermal stability dependence on pH. Effect of three denaturing agents in a wide concentration range on Tm showed nicely that chemical denaturation occurs only at the highest concentrations. Results showed similar effect on Tm for most formulations on different proteins. Most successful formulations were tested for enveloped virus stabilizing potential using cell culture infectivity assay (CCID50) and results showed lack of correlation with TSA results. Only weak correlation of Tm shift and protein polymerization measured by SEC-HPLC was obtained, meaning that polymerization cannot be predicted from Tm shifts.


Subject(s)
Measles virus/chemistry , Mumps virus/chemistry , Protein Stability , Viral Envelope Proteins/chemistry , Albumins/chemistry , Cells, Cultured , Drug Compounding , Drug Stability , Guanidine/chemistry , Hydrogen-Ion Concentration , Immunoglobulin G/chemistry , Measles virus/pathogenicity , Molecular Dynamics Simulation , Mumps virus/pathogenicity , Ovalbumin/chemistry , Polymerization , Potassium Cyanide/chemistry , Protein Denaturation/drug effects , Transition Temperature , Urea/chemistry
12.
Biochim Biophys Acta Gen Subj ; 1861(11 Pt A): 2543-2550, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28890401

ABSTRACT

Intrinsically disordered proteins (IDPs) possess a peculiar amino acid composition that makes them very soluble. Nevertheless, they can encounter aggregation in physiological and pathological contexts. In this work, we addressed the issue of how electrostatic charges can influence aggregation propensity by using the N-terminus moiety of the measles virus phosphoprotein, PNT, as a model IDP. Taking advantage of the high sequence designability of IDPs, we have produced an array of PNT variants sharing the same hydrophobicity, but differing in net charges per residue and isoelectric points (pI). The solubility and conformational properties of these proteins were analysed through biochemical and biophysical techniques in a wide range of pH values and compared with those of the green fluorescence protein (GFP), a globular protein with lower net charge per residue, but similar hydrophobicity. Tested proteins showed a solubility minimum close to their pI, as expected, but the pH-dependent decrease of solubility was not uniform and driven by the net charge per residue of each variant. A parallel behaviour was observed also in fusion proteins between PNT variants and GFP, which minimally contributes to the solubility of chimeras. Our data suggest that the overall solubility of a protein can be dictated by protein regions endowed with higher net charge per residue and, hence, prompter to respond to pH changes. This finding could be exploited for biotechnical purposes, such as the design of solubility/aggregation tags, and in studies aimed to clarify the pathological and physiological behaviour of IDPs.


Subject(s)
Amino Acid Sequence/genetics , Intrinsically Disordered Proteins/chemistry , Protein Aggregates/genetics , Viral Proteins/chemistry , Hydrogen-Ion Concentration , Hydrophobic and Hydrophilic Interactions , Intrinsically Disordered Proteins/genetics , Kinetics , Measles virus/chemistry , Measles virus/genetics , Protein Conformation , Protein Folding , Solubility , Static Electricity , Viral Proteins/genetics
13.
Curr Opin Virol ; 24: 105-114, 2017 06.
Article in English | MEDLINE | ID: mdl-28601688

ABSTRACT

The paramyxovirus family comprises major human and animal pathogens such as measles virus (MeV), mumps virus (MuV), the parainfluenzaviruses, Newcastle disease virus (NDV), and the highly pathogenic zoonotic hendra (HeV) and nipah (NiV) viruses. Paramyxovirus particles are pleomorphic, with a lipid envelope, nonsegmented RNA genomes of negative polarity, and densely packed glycoproteins on the virion surface. A number of crystal structures of different paramyxovirus proteins and protein fragments were solved, but the available information concerning overall virion organization remains limited. However, recent studies have reported cryo-electron tomography-based reconstructions of Sendai virus (SeV), MeV, NDV, and human parainfluenza virus type 3 (HPIV3) particles and a surface assessment of NiV-derived virus-like particles (VLPs), which have yielded innovative hypotheses concerning paramyxovirus particle assembly, budding, and organization. Following a summary of the current insight into paramyxovirus virion morphology, this review will focus on discussing the implications of these particle reconstructions on the present models of paramyxovirus assembly and infection.


Subject(s)
Paramyxoviridae/chemistry , Viral Matrix Proteins/chemistry , Viral Matrix Proteins/physiology , Virion/chemistry , Cryoelectron Microscopy , Electron Microscope Tomography , Genome, Viral , Humans , Measles virus/chemistry , Newcastle disease virus/chemistry , Nipah Virus/chemistry , Paramyxoviridae/physiology , Paramyxoviridae/ultrastructure , Viral Fusion Proteins/chemistry , Virion/metabolism , Virus Assembly , Virus Release
14.
Biochemistry ; 56(29): 3780-3786, 2017 07 25.
Article in English | MEDLINE | ID: mdl-28661120

ABSTRACT

Intrinsically disordered proteins (IDPs) are functionally active despite lacking a well-defined three-dimensional structure. Such proteins often undergo a disorder-to-order transition, or induced folding, when binding to their specific physiological partner. Because of cooperativity, the folding and binding steps typically appear as a single event, and therefore, induced folding is extremely difficult to characterize experimentally. In this perspective, the interaction between the disordered C-terminal domain of the measles virus nucleoprotein NTAIL and the folded X domain of the viral phosphoprotein (XD) is particularly interesting because the inherent complexity of the observed kinetics allows characterization of the binding and folding steps individually. Here we present a detailed structural description of the folding and binding events occurring in the recognition between NTAIL and XD. This result was achieved by measuring the effect of single-amino acid substitutions in NTAIL on the reaction mechanism. Analysis of the experimental data allowed us (i) to identify the key residues involved in the initial recognition between the two molecules and (ii) to depict the general features of the folding pathway of NTAIL. Furthermore, an analysis of the changes in stability obtained for the whole set of variants highlights how the sequence of this IDP has not been selected during evolution to fold efficiently. This feature might be a consequence of the weakly funneled nature of the energy landscape of IDPs in their unbound state and represents a plausible explanation of their highly dynamic nature even in the bound state, typically defined as "fuzziness".


Subject(s)
Intrinsically Disordered Proteins/chemistry , Measles virus/chemistry , Nucleoproteins/chemistry , Protein Engineering , Protein Folding , Viral Proteins/chemistry , Intrinsically Disordered Proteins/genetics , Measles virus/genetics , Nucleocapsid Proteins , Nucleoproteins/genetics , Protein Domains , Viral Proteins/genetics
15.
Biomedica ; 37(0): 121-132, 2017 Apr 01.
Article in Spanish | MEDLINE | ID: mdl-28527274

ABSTRACT

Introducción. El virus del Zika (ZIKV) es un flavivirus con envoltura, transmitido a los seres humanos principalmente por el vector Aedes aegypti. La infección por ZIKV se ha asociado con un gran neurotropismo y con efectos neuropáticos, como el síndrome de Guillain-Barré en el adulto y la microcefalia fetal y posnatal, así como con un síndrome de infección congénita similar al producido por el virus de la rubéola (RV).Objetivo. Comparar las estructuras moleculares de la proteína de envoltura E del virus del Zika (E-ZIKV) y de la E1 del virus de la rubéola (E1-RV), y plantear posibles implicaciones en el neurotropismo y en las alteraciones del sistema nervioso asociadas con el ZIKV.Materiales y métodos. La secuencia de aminoácidos de la proteína E-ZIKV (PDB: 5iZ7) se alineó con la de la glucopreteína E1 del virus de la rubéola (PDB: 4ADG). Los elementos de la estructura secundaria se determinaron usando los programas Vector NTI Advance®, DSSP y POSA, así como herramientas de gestión de datos (AlignX®). Uno de los criterios principales de comparación y alineación fue la asignación de residuos estructuralmente equivalentes, con más de 70 % de identidad.Resultados. La organización estructural de la proteína E-ZIKV (PDB: 5iZ7) fue similar a la de E1-RV (PDB: 4ADG) (70 a 80 % de identidad), y se observó una correspondencia con la estructura definida para las glucoproteínas de fusión de membrana de clase II de los virus con envoltura. E-ZIKV y E1-RV exhibieron elementos estructurales de fusión muy conservados en la región distal del dominio II, asociados con la unión a los receptores celulares de entrada del virus de la rubéola (glucoproteína de mielina del oligodendrocito, Myelin Oligodendrocyte Glycoprotein, MOG), y con los receptores celulares Axl del ZIKV y de otros flavivirus.Conclusión. La comparación de las proteínas E-ZIKV y E1-RV es un paso necesario hacia la definición de otros factores moleculares determinantes del neurotropismo y la patogenia del ZIKV, el cual puede contribuir a generar estrategias de diagnóstico, prevención y tratamiento de las complicaciones neurológicas inducidas por el ZIKV.


Subject(s)
Measles virus/chemistry , Serine Endopeptidases/chemistry , Serine Endopeptidases/metabolism , Viral Envelope Proteins/metabolism , Viral Proteins/chemistry , Zika Virus/chemistry , Humans , Measles virus/pathogenicity , Measles virus/physiology , Molecular Biology , Viral Proteins/genetics , Viral Proteins/physiology , Zika Virus/pathogenicity , Zika Virus/physiology
16.
Biomédica (Bogotá) ; 37(supl.1): 121-132, abr. 2017. graf
Article in Spanish | LILACS | ID: biblio-888518

ABSTRACT

Resumen Introducción. El virus del Zika (ZIKV) es un flavivirus con envoltura, transmitido a los seres humanos principalmente por el vector Aedes aegypti. La infección por ZIKV se ha asociado con un gran neurotropismo y con efectos neuropáticos, como el síndrome de Guillain-Barré en el adulto y la microcefalia fetal y posnatal, así como con un síndrome de infección congénita similar al producido por el virus de la rubéola (RV). Objetivo. Comparar las estructuras moleculares de la proteína de envoltura E del virus del Zika (E-ZIKV) y de la E1 del virus de la rubéola (E1-RV), y plantear posibles implicaciones en el neurotropismo y en las alteraciones del sistema nervioso asociadas con el ZIKV. Materiales y métodos. La secuencia de aminoácidos de la proteína E-ZIKV (PDB: 5iZ7) se alineó con la de la glucopreteína E1 del virus de la rubéola (PDB: 4ADG). Los elementos de la estructura secundaria se determinaron usando los programas Vector NTI Advance®, DSSP y POSA, así como herramientas de gestión de datos (AlignX®). Uno de los criterios principales de comparación y alineación fue la asignación de residuos estructuralmente equivalentes, con más de 70 % de identidad. Resultados. La organización estructural de la proteína E-ZIKV (PDB: 5iZ7) fue similar a la de E1-RV (PDB: 4ADG) (70 a 80 % de identidad), y se observó una correspondencia con la estructura definida para las glucoproteínas de fusión de membrana de clase II de los virus con envoltura. E-ZIKV y E1-RV exhibieron elementos estructurales de fusión muy conservados en la región distal del dominio II, asociados con la unión a los receptores celulares de entrada del virus de la rubéola (glucoproteína de mielina del oligodendrocito, Myelin Oligodendrocyte Glycoprotein, MOG), y con los receptores celulares Axl del ZIKV y de otros flavivirus. Conclusión. La comparación de las proteínas E-ZIKV y E1-RV es un paso necesario hacia la definición de otros factores moleculares determinantes del neurotropismo y la patogenia del ZIKV, el cual puede contribuir a generar estrategias de diagnóstico, prevención y tratamiento de las complicaciones neurológicas inducidas por el ZIKV.


Abstract Introduction: Zika virus (ZIKV) is an enveloped flavivirus transmitted to humans mainly by Aedes aegypti. ZIKV infection has been associated with high neurotropism and neuropathic effects such as the Guillain-Barré syndrome in adults, and fetal and postnatal microcephaly and the congenital Zika virus syndrome similar to that produced by rubella virus (VR). Objective: To compare Zika virus membrane protein E (E-ZIKV) and rubella virus membrane protein E1 (E1-RV), and to propose possible implications for neurotropism and nervous system disorders associated with ZIKV infections. Materials and methods: The amino acid sequence of E-ZIKV protein (PDB: 5iZ7) was aligned to that of rubella virus glycoprotein E1 (PDB: 4ADG). The secondary structure elements were determined using the programs Vector NTI Advance®, DSSP, and POSA, and integrated data management tools (AlignX®). One of the main comparison and alignment criteria was the allocation of structurally equivalent residues with more than 70% identity. Results: E-ZIKV structural organization (PDB: 5iZ7) was similar to that of E1-RV (PDB: 4ADG) (70%-80% identity), and it was consistent with relevant structural features of viral membrane class II fusion glycoproteins. E-ZIKV and E1-RV exhibited highly conserved fusion structural elements at the distal region of domain II, which has been associated with the RV myelin oligodendrocyte glycoprotein and Axl cell receptors in ZIKV and other flaviviruses. Conclusion: The comparison of E-ZIKV and E1-RV proteins constitutes an essential step towards the definition of ZIKV neurotropism and pathogenesis molecular determinants, and for the adoption of diagnosis, prevention and treatment strategies against neurological complications induced by ZIKV infection.


Subject(s)
Humans , Viral Proteins/chemistry , Serine Endopeptidases/metabolism , Serine Endopeptidases/chemistry , Viral Envelope Proteins/metabolism , Zika Virus/chemistry , Measles virus/chemistry , Viral Proteins/physiology , Viral Proteins/genetics , Zika Virus/physiology , Zika Virus/pathogenicity , Measles virus/physiology , Measles virus/pathogenicity , Molecular Biology
17.
J Virol Methods ; 245: 53-60, 2017 07.
Article in English | MEDLINE | ID: mdl-28315717

ABSTRACT

In this study we evaluated the long-term stability of a microarray-based serological screening platform, containing antigens of influenza A, measles and Streptococcus pneumoniae, as part of a preparedness research program aiming to develop assays for syndromic disease detection. Spotted microarray slides were kept at four different storage regimes with varying temperature and humidity conditions. We showed that under the standard storage condition in a temperature-controlled (21°C) and desiccated environment (0% relative humidity), microarray slides remained stable for at least 22 months without loss of antigen quality, whereas the other three conditions (37°C, desiccated; Room temperature, non-desiccated; Frozen, desiccated) produced acceptable results for some antigens (influenza A, S.pneumoniae), but not for others (measles). We conclude that these arrays for multiplex antibody testing can be prepared and stored for prolonged periods of time, which aids laboratory-preparedness and facilitates sero-epidemiological studies.


Subject(s)
Antigens, Bacterial/chemistry , Antigens, Viral/chemistry , Drug Storage , Protein Array Analysis , Specimen Handling/methods , Desiccation , Drug Stability , Humans , Immunologic Tests/instrumentation , Influenza A virus/chemistry , Influenza, Human/diagnosis , Measles/diagnosis , Measles virus/chemistry , Pneumococcal Infections/diagnosis , Seroepidemiologic Studies , Specimen Handling/instrumentation , Streptococcus pneumoniae/chemistry , Temperature , Time Factors
18.
J Virol ; 91(1)2017 Jan 01.
Article in English | MEDLINE | ID: mdl-27733647

ABSTRACT

Measles virus (MV) infection is undergoing resurgence and remains one of the leading causes of death among young children worldwide despite the availability of an effective measles vaccine. MV infects its target cells by coordinated action of the MV hemagglutinin (H) and fusion (F) envelope glycoproteins; upon receptor engagement by H, the prefusion F undergoes a structural transition, extending and inserting into the target cell membrane and then refolding into a postfusion structure that fuses the viral and cell membranes. By interfering with this structural transition of F, peptides derived from the heptad repeat (HR) regions of F can inhibit MV infection at the entry stage. In previous work, we have generated potent MV fusion inhibitors by dimerizing the F-derived peptides and conjugating them to cholesterol. We have shown that prophylactic intranasal administration of our lead fusion inhibitor efficiently protects from MV infection in vivo We show here that peptides tagged with lipophilic moieties self-assemble into nanoparticles until they reach the target cells, where they are integrated into cell membranes. The self-assembly feature enhances biodistribution and the half-life of the peptides, while integration into the target cell membrane increases fusion inhibitor potency. These factors together modulate in vivo efficacy. The results suggest a new framework for developing effective fusion inhibitory peptides. IMPORTANCE: Measles virus (MV) infection causes an acute illness that may be associated with infection of the central nervous system (CNS) and severe neurological disease. No specific treatment is available. We have shown that fusion-inhibitory peptides delivered intranasally provide effective prophylaxis against MV infection. We show here that specific biophysical properties regulate the in vivo efficacy of MV F-derived peptides.


Subject(s)
Hemagglutinins, Viral/immunology , Measles Vaccine/administration & dosage , Measles virus/drug effects , Measles/prevention & control , Nanoparticles/administration & dosage , Peptides/immunology , Viral Fusion Proteins/immunology , Administration, Intranasal , Amino Acid Sequence , Animals , Brain/drug effects , Brain/immunology , Cholesterol/chemistry , Female , Half-Life , Hemagglutinins, Viral/chemistry , Humans , Lung/drug effects , Lung/immunology , Male , Measles/immunology , Measles/mortality , Measles/virology , Measles Vaccine/chemical synthesis , Measles virus/chemistry , Measles virus/immunology , Nanoparticles/chemistry , Peptides/chemical synthesis , Sigmodontinae , Survival Analysis , Viral Fusion Proteins/chemistry , Virus Internalization/drug effects
19.
J Gen Virol ; 98(2): 143-154, 2017 02.
Article in English | MEDLINE | ID: mdl-27911256

ABSTRACT

The fusion (F) protein of measles virus performs refolding from the thermodynamically metastable prefusion form to the highly stable postfusion form via an activated unstable intermediate stage, to induce membrane fusion. Some amino acids involved in the fusion regulation cluster in the heptad repeat B (HR-B) domain of the stalk region, among which substitution of residue 465 by various amino acids revealed that fusion activity correlates well with its side chain length from the Cα (P<0.01) and van der Waals volume (P<0.001), except for Phe, Tyr, Trp, Pro and His carrying ring structures. Directed towards the head region, longer side chains of the non-ring-type 465 residues penetrate more deeply into the head region and may disturb the hydrophobic interaction between the stalk and head regions and cause destabilization of the molecule by lowering the energy barrier for refolding, which conferred the F protein enhanced fusion activity. Contrarily, the side chain of ring-type 465 residues turned away from the head region, resulting in not only no contact with the head region but also extensive coverage of the HR-B surface, which may prevent the dissociation of the HR-B bundle for initiation of membrane fusion and suppress fusion activity. Located in the HR-B domain just at the junction between the head and stalk regions, amino acid 465 is endowed with a possible ability to either destabilize or stabilize the F protein depending on its molecular volume and the direction of the side chain, regulating fusion activity of measles virus F protein.


Subject(s)
Measles virus/chemistry , Measles/virology , Membrane Fusion , Viral Fusion Proteins/chemistry , Amino Acid Sequence , Amino Acid Substitution , Animals , Chlorocebus aethiops , Humans , Measles virus/ultrastructure , Protein Domains , Protein Folding , Protein Stability , Thermodynamics , Vero Cells
20.
PLoS Pathog ; 12(12): e1006058, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27936158

ABSTRACT

Measles virus (MeV) and all Paramyxoviridae members rely on a complex polymerase machinery to ensure viral transcription and replication. Their polymerase associates the phosphoprotein (P) and the L protein that is endowed with all necessary enzymatic activities. To be processive, the polymerase uses as template a nucleocapsid made of genomic RNA entirely wrapped into a continuous oligomer of the nucleoprotein (N). The polymerase enters the nucleocapsid at the 3'end of the genome where are located the promoters for transcription and replication. Transcription of the six genes occurs sequentially. This implies ending and re-initiating mRNA synthesis at each intergenic region (IGR). We explored here to which extent the binding of the X domain of P (XD) to the C-terminal region of the N protein (NTAIL) is involved in maintaining the P/L complex anchored to the nucleocapsid template during the sequential transcription. Amino acid substitutions introduced in the XD-binding site on NTAIL resulted in a wide range of binding affinities as determined by combining protein complementation assays in E. coli and human cells and isothermal titration calorimetry. Molecular dynamics simulations revealed that XD binding to NTAIL involves a complex network of hydrogen bonds, the disruption of which by two individual amino acid substitutions markedly reduced the binding affinity. Using a newly designed, highly sensitive dual-luciferase reporter minigenome assay, the efficiency of re-initiation through the five measles virus IGRs was found to correlate with NTAIL/XD KD. Correlatively, P transcript accumulation rate and F/N transcript ratios from recombinant viruses expressing N variants were also found to correlate with the NTAIL to XD binding strength. Altogether, our data support a key role for XD binding to NTAIL in maintaining proper anchor of the P/L complex thereby ensuring transcription re-initiation at each intergenic region.


Subject(s)
Measles/virology , Nucleoproteins/metabolism , Viral Proteins/metabolism , Virus Replication/physiology , Calorimetry , Circular Dichroism , DNA, Intergenic , Humans , Mass Spectrometry , Measles/metabolism , Measles virus/chemistry , Measles virus/metabolism , Models, Molecular , Nucleocapsid Proteins , Nucleoproteins/chemistry , Protein Binding , Transcription, Genetic , Viral Proteins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...