Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
Add more filters










Publication year range
1.
PLoS Comput Biol ; 16(10): e1008357, 2020 10.
Article in English | MEDLINE | ID: mdl-33079933

ABSTRACT

Icosahedral viruses are under a micrometer in diameter, their infectious genome encapsulated by a shell assembled by a multiscale process, starting from an integer multiple of 60 viral capsid or coat protein (VP) monomers. We predict and validate inter-atomic hotspot interactions between VP monomers that are important for the assembly of 3 types of icosahedral viral capsids: Adeno Associated Virus serotype 2 (AAV2) and Minute Virus of Mice (MVM), both T = 1 single stranded DNA viruses, and Bromo Mosaic Virus (BMV), a T = 3 single stranded RNA virus. Experimental validation is by in-vitro, site-directed mutagenesis data found in literature. We combine ab-initio predictions at two scales: at the interface-scale, we predict the importance (cruciality) of an interaction for successful subassembly across each interface between symmetry-related VP monomers; and at the capsid-scale, we predict the cruciality of an interface for successful capsid assembly. At the interface-scale, we measure cruciality by changes in the capsid free-energy landscape partition function when an interaction is removed. The partition function computation uses atlases of interface subassembly landscapes, rapidly generated by a novel geometric method and curated opensource software EASAL (efficient atlasing and search of assembly landscapes). At the capsid-scale, cruciality of an interface for successful assembly of the capsid is based on combinatorial entropy. Our study goes all the way from resource-light, multiscale computational predictions of crucial hotspot inter-atomic interactions to validation using data on site-directed mutagenesis' effect on capsid assembly. By reliably and rapidly narrowing down target interactions, (no more than 1.5 hours per interface on a laptop with Intel Core i5-2500K @ 3.2 Ghz CPU and 8GB of RAM) our predictions can inform and reduce time-consuming in-vitro and in-vivo experiments, or more computationally intensive in-silico analyses.


Subject(s)
Capsid Proteins , Capsid , Virus Assembly/genetics , Capsid/chemistry , Capsid/metabolism , Capsid Proteins/chemistry , Capsid Proteins/genetics , Capsid Proteins/metabolism , Computer Simulation , Dependovirus/chemistry , Dependovirus/genetics , Dependovirus/metabolism , Minute Virus of Mice/chemistry , Minute Virus of Mice/genetics , Minute Virus of Mice/metabolism , Mutagenesis, Site-Directed
2.
PLoS Pathog ; 16(10): e1009002, 2020 10.
Article in English | MEDLINE | ID: mdl-33064772

ABSTRACT

The autonomous parvovirus Minute Virus of Mice (MVM) localizes to cellular DNA damage sites to establish and sustain viral replication centers, which can be visualized by focal deposition of the essential MVM non-structural phosphoprotein NS1. How such foci are established remains unknown. Here, we show that NS1 localized to cellular sites of DNA damage independently of its ability to covalently bind the 5' end of the viral genome, or its consensus DNA binding sequence. Many of these sites were identical to those occupied by virus during infection. However, localization of the MVM genome to DNA damage sites occurred only when wild-type NS1, but not its DNA-binding mutant was expressed. Additionally, wild-type NS1, but not its DNA binding mutant, could localize a heterologous DNA molecule containing the NS1 binding sequence to DNA damage sites. These findings suggest that NS1 may function as a bridging molecule, helping the MVM genome localize to cellular DNA damage sites to facilitate ongoing virus replication.


Subject(s)
DNA Damage , Minute Virus of Mice/genetics , Minute Virus of Mice/metabolism , Viral Nonstructural Proteins/metabolism , Animals , Cell Line , DNA Replication , DNA, Viral/genetics , DNA-Binding Proteins/genetics , Female , Genome, Viral , Humans , Male , Mice , Parvoviridae Infections/genetics , Parvoviridae Infections/virology , Parvovirus/genetics , Virus Replication
3.
J Chromatogr A ; 1633: 461635, 2020 Dec 06.
Article in English | MEDLINE | ID: mdl-33128974

ABSTRACT

Viral clearance is an important performance metric for the downstream process of monoclonal antibodies (mAbs) due to its impact on patient safety. Anion exchange chromatography (AEX) has been well-accepted in the industry as one of the workhorse techniques for removing viruses, and is considered to be able to achieve high log clearance values under most operating conditions. However, it is not uncommon for viral clearance results on AEX to fall below the desired level despite operating under conditions that should achieve high clearance levels according to conventional wisdom of how this mode of chromatography operates. In this study, a design of experiment (DoE) approach was used to develop a more fundamental understanding of viral clearance during AEX chromatography using Minute Virus of Mice (MVM) on POROS HQ resin. Load pH, conductivity and virus concentration were evaluated as design factors for three mAbs with varying physical and chemical properties. The hydrophobicity and surface charge distributions of the molecules were found to be the most significant factors in influencing viral clearance performance, and the viral clearance trends did not seem to fit with conventional wisdom. To explain this seemingly unconventional behavior, we propose a new mechanism that suggests that interactions between the mAb and the virus have a major contribution on retention of the virus on the resin. This furthered understanding may help improve the predictability, performance and robustness of viral clearance during AEX chromatography.


Subject(s)
Antibodies, Monoclonal/metabolism , Chromatography, Ion Exchange/standards , Minute Virus of Mice/metabolism , Viruses/metabolism , Animals , Anions/chemistry , Antibodies, Monoclonal/chemistry , Mice , Viruses/chemistry
4.
Nanoscale ; 11(19): 9369-9383, 2019 May 16.
Article in English | MEDLINE | ID: mdl-31041970

ABSTRACT

Virus particles and other protein-based supramolecular complexes have a vast nanotechnological potential. However, protein nanostructures are "soft" materials prone to disruption by force. Whereas some non-biological nanoparticles (NPs) may be stronger, for certain applications protein- and virus-based NPs have potential advantages related to their structure, self-assembly, production, engineering, and/or inbuilt functions. Thus, it may be desirable to acquire the knowledge needed to engineer protein-based nanomaterials with a higher strength against mechanical breakage. Here we have used the capsid of the minute virus of mice to experimentally identify individual chemical groups that determine breakage-related properties of a virus particle. Individual amino acid side chains that establish interactions between building blocks in the viral particle were truncated using protein engineering. Indentation experiments using atomic force microscopy were carried out to investigate the role of each targeted side chain in determining capsid strength and brittleness, by comparing the maximum force and deformation each modified capsid withstood before breaking apart. Side chains with major roles in determining capsid strength against breakage included polar groups located in solvent-exposed positions, and did not generally correspond with those previously identified as determinants of mechanical stiffness. In contrast, apolar side chains buried along the intersubunit interfaces that generally determined capsid stiffness had, at most, a minor influence on strength against disruption. Whereas no correlated variations between strength and either stiffness or brittleness were found, brittleness and stiffness were quantitatively correlated. Implications for developing robust protein-based NPs and for acquiring a deeper physics-based perspective of viruses are discussed.


Subject(s)
Amino Acids/chemistry , Capsid Proteins/chemistry , Minute Virus of Mice/metabolism , Nanoparticles/chemistry , Animals , Capsid Proteins/genetics , Capsid Proteins/metabolism , Mechanical Phenomena , Mice , Microscopy, Atomic Force , Mutagenesis, Site-Directed , Nanoparticles/metabolism , Protein Engineering
5.
J Am Chem Soc ; 138(47): 15385-15396, 2016 11 30.
Article in English | MEDLINE | ID: mdl-27933931

ABSTRACT

Understanding the fundamental principles underlying supramolecular self-assembly may facilitate many developments, from novel antivirals to self-organized nanodevices. Icosahedral virus particles constitute paradigms to study self-assembly using a combination of theory and experiment. Unfortunately, assembly pathways of the structurally simplest virus capsids, those more accessible to detailed theoretical studies, have been difficult to study experimentally. We have enabled the in vitro self-assembly under close to physiological conditions of one of the simplest virus particles known, the minute virus of mice (MVM) capsid, and experimentally analyzed its pathways of assembly and disassembly. A combination of electron microscopy and high-resolution atomic force microscopy was used to structurally characterize and quantify a succession of transient assembly and disassembly intermediates. The results provided an experiment-based model for the reversible self-assembly pathway of a most simple (T = 1) icosahedral protein shell. During assembly, trimeric capsid building blocks are sequentially added to the growing capsid, with pentamers of building blocks and incomplete capsids missing one building block as conspicuous intermediates. This study provided experimental verification of many features of self-assembly of a simple T = 1 capsid predicted by molecular dynamics simulations. It also demonstrated atomic force microscopy imaging and automated analysis, in combination with electron microscopy, as a powerful single-particle approach to characterize at high resolution and quantify transient intermediates during supramolecular self-assembly/disassembly reactions. Finally, the efficient in vitro self-assembly achieved for the oncotropic, cell nucleus-targeted MVM capsid may facilitate its development as a drug-encapsidating nanoparticle for anticancer targeted drug delivery.


Subject(s)
Capsid/metabolism , Capsid/ultrastructure , Microscopy, Atomic Force , Minute Virus of Mice/metabolism , Minute Virus of Mice/ultrastructure , Molecular Dynamics Simulation , Virus Assembly , Capsid/chemistry , Microscopy, Electron , Minute Virus of Mice/chemistry , Particle Size , Surface Properties
6.
PLoS One ; 9(1): e86909, 2014.
Article in English | MEDLINE | ID: mdl-24475195

ABSTRACT

The recognition of sialic acids by two strains of minute virus of mice (MVM), MVMp (prototype) and MVMi (immunosuppressive), is an essential requirement for successful infection. To understand the potential for recognition of different modifications of sialic acid by MVM, three types of capsids, virus-like particles, wild type empty (no DNA) capsids, and DNA packaged virions, were screened on a sialylated glycan microarray (SGM). Both viruses demonstrated a preference for binding to 9-O-methylated sialic acid derivatives, while MVMp showed additional binding to 9-O-acetylated and 9-O-lactoylated sialic acid derivatives, indicating recognition differences. The glycans recognized contained a type-2 Galß1-4GlcNAc motif (Neu5Acα2-3Galß1-4GlcNAc or 3'SIA-LN) and were biantennary complex-type N-glycans with the exception of one. To correlate the recognition of the 3'SIA-LN glycan motif as well as the biantennary structures to their natural expression in cell lines permissive for MVMp, MVMi, or both strains, the N- and O-glycans, and polar glycolipids present in three cell lines used for in vitro studies, A9 fibroblasts, EL4 T lymphocytes, and the SV40 transformed NB324K cells, were analyzed by MALDI-TOF/TOF mass spectrometry. The cells showed an abundance of the sialylated glycan motifs recognized by the viruses in the SGM and previous glycan microarrays supporting their role in cellular recognition by MVM. Significantly, the NB324K showed fucosylation at the non-reducing end of their biantennary glycans, suggesting that recognition of these cells is possibly mediated by the Lewis X motif as in 3'SIA-Le(X) identified in a previous glycan microarray screen.


Subject(s)
Mice/virology , Minute Virus of Mice/metabolism , N-Acetylneuraminic Acid/metabolism , Animals , Capsid/metabolism , Cell Line , Fibroblasts , Humans , Lymphocytes , Microarray Analysis , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Virion/metabolism
7.
PLoS Pathog ; 10(1): e1003891, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24415942

ABSTRACT

Parvoviruses halt cell cycle progression following initiation of their replication during S-phase and continue to replicate their genomes for extended periods of time in arrested cells. The parvovirus minute virus of mice (MVM) induces a DNA damage response that is required for viral replication and induction of the S/G2 cell cycle block. However, p21 and Chk1, major effectors typically associated with S-phase and G2-phase cell cycle arrest in response to diverse DNA damage stimuli, are either down-regulated, or inactivated, respectively, during MVM infection. This suggested that parvoviruses can modulate cell cycle progression by another mechanism. In this work we show that the MVM-induced, p21- and Chk1-independent, cell cycle block proceeds via a two-step process unlike that seen in response to other DNA-damaging agents or virus infections. MVM infection induced Chk2 activation early in infection which led to a transient S-phase block associated with proteasome-mediated CDC25A degradation. This step was necessary for efficient viral replication; however, Chk2 activation and CDC25A loss were not sufficient to keep infected cells in the sustained G2-arrested state which characterizes this infection. Rather, although the phosphorylation of CDK1 that normally inhibits entry into mitosis was lost, the MVM induced DDR resulted first in a targeted mis-localization and then significant depletion of cyclin B1, thus directly inhibiting cyclin B1-CDK1 complex function and preventing mitotic entry. MVM infection thus uses a novel strategy to ensure a pseudo S-phase, pre-mitotic, nuclear environment for sustained viral replication.


Subject(s)
Cyclin B1/metabolism , Minute Virus of Mice/metabolism , Mitosis , Parvoviridae Infections/metabolism , Animals , CDC2 Protein Kinase , Cell Line , Checkpoint Kinase 2/genetics , Checkpoint Kinase 2/metabolism , Cyclin B1/genetics , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , Enzyme Activation/genetics , G2 Phase Cell Cycle Checkpoints/genetics , Humans , Mice , Minute Virus of Mice/genetics , Parvoviridae Infections/genetics , Parvoviridae Infections/pathology , S Phase Cell Cycle Checkpoints/genetics , cdc25 Phosphatases/genetics , cdc25 Phosphatases/metabolism
8.
Biochem J ; 455(1): 87-94, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23875612

ABSTRACT

Viruses constitute paradigms to study conformational dynamics in biomacromolecular assemblies. Infection by the parvovirus MVM (minute virus of mice) requires a conformational rearrangement that involves the intracellular externalization through capsid channels of the 2Nt (N-terminal region of VP2). We have investigated the role in this process of conserved glycine residues in an extended glycine-rich tract located immediately after 2Nt. Based on the virus structure, residues with hydrophobic side chains of increasing volume were substituted for glycine residues 31 or 33. Mutations had no effect on capsid assembly or stability, but inhibited virus infectivity. All mutations, except those to alanine residues which had minor effects, impaired 2Nt externalization in nuclear maturing virions and in purified virions, to an extent that correlated with the side chain size. Different biochemical and biophysical analyses were consistent with this result. Importantly, all of the tested glycine residue replacements impaired the capacity of the virion to initiate infection, at ratios correlating with their restrictive effects on 2Nt externalization. Thus small residues within the evolutionarily conserved glycine-rich tract facilitate 2Nt externalization through the capsid channel, as required by this virus to initiate cell entry. The results demonstrate the exquisite dependence on geometric constraints of a biologically relevant translocation event in a biomolecular complex.


Subject(s)
Capsid Proteins/chemistry , Minute Virus of Mice/genetics , Peptides/chemistry , Virion/genetics , Virus Release/physiology , Amino Acid Substitution , Animals , Capsid/chemistry , Capsid/metabolism , Capsid Proteins/genetics , Capsid Proteins/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Nucleus/virology , Humans , Kinetics , Mice , Minute Virus of Mice/chemistry , Minute Virus of Mice/metabolism , Models, Molecular , Mutation , Peptides/genetics , Peptides/metabolism , Protein Structure, Tertiary , Protein Transport , Thermodynamics , Virion/chemistry , Virus Assembly
9.
Lab Anim ; 47(2): 116-21, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23492514

ABSTRACT

Mouse parvoviruses are among the most prevalent infectious pathogens in contemporary mouse colonies. To improve the efficiency of routine screening for mouse parvovirus infections, a multiplex polymerase chain reaction (PCR) assay targeting the VP gene was developed. The assay detected minute virus of mice (MVM), mouse parvovirus (MPV) and a mouse housekeeping gene (α-actin) and was able to specifically detect MVM and MPV at levels as low as 50 copies. Co-infection with the two viruses with up to 200-fold differences in viral concentrations can easily be detected. The multiplex PCR assay developed here could be a useful tool for monitoring mouse health and the viral contamination of biological materials.


Subject(s)
Mice , Multiplex Polymerase Chain Reaction/methods , Parvoviridae Infections/diagnosis , Parvovirus/isolation & purification , Rodent Diseases/diagnosis , Actins/genetics , Actins/metabolism , Animals , Minute Virus of Mice/genetics , Minute Virus of Mice/isolation & purification , Minute Virus of Mice/metabolism , Parvoviridae Infections/virology , Parvovirus/genetics , Parvovirus/metabolism , Rodent Diseases/virology , Sensitivity and Specificity , Sequence Analysis, DNA , Viral Proteins/genetics , Viral Proteins/metabolism
10.
Cancer Gene Ther ; 19(7): 468-75, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22539062

ABSTRACT

Rodent autonomous parvoviruses (PVs) are endowed with oncotropic properties and represent virotherapeutics with inherent oncolytic features. This work aimed to evaluate the capacity of Minute Virus of Mice (MVMp) to act as an adjuvant stimulating a mouse glioblastoma-specific immune response. MVMp was shown to induce cell death through apoptosis in glioma GL261 cells. Antigen-presenting cells (APCs) provide the initial cue for innate and adaptive immune responses, and thus MVMp-infected GL261 cells were tested for their ability to activate dendritic cells (DCs) and microglia (MG), two distinct cell types that are able to act as APCs. MG and discrete DC subsets were activated after co-culture with MVMp-infected glioma GL261 cells, as evidenced by upregulation of specific activation markers (CD80, CD86) and release of proinflammatory cytokines (tumor necrosis factor-α and interleukin-6). The in vivo analysis of immunodeficient and immunocompetent mice revealed a clear difference in their susceptibility to MVMp-mediated tumor suppression. Immunocompetent mice were fully protected from tumor outgrowth of GL261 cells infected ex vivo with MVMp. In contrast, immunodeficient animals were less competent for MVMp-dependent tumor inhibition, with only 20% of the recipients being protected, arguing for an additional immune component to allow full tumor suppression. In keeping with this conclusion, immunocompetent mice engrafted with MVMp-infected glioma cells developed a level of anti-tumor immunity with isolated splenocytes producing elevated levels of interferon-γ. In rechallenge experiments using uninfected GL261 cells, we could show complete protection against the tumor, arguing for the induction of a T-cell-mediated, tumor-specific, long-term memory response. These findings indicate that the anticancer effect of PVs can be traced back not only for their direct oncolytic effect, but also to their ability to break tumor tolerance.


Subject(s)
Glioma/immunology , Minute Virus of Mice/immunology , Oncolytic Viruses/immunology , Adaptive Immunity , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Cell Line, Tumor , Coculture Techniques , Dendritic Cells/cytology , Dendritic Cells/immunology , Glioma/pathology , Glioma/prevention & control , Mice , Mice, Inbred C57BL , Microglia/cytology , Microglia/immunology , Minute Virus of Mice/genetics , Minute Virus of Mice/metabolism , Neoplasm Transplantation , Oncolytic Virotherapy , Oncolytic Viruses/genetics , Oncolytic Viruses/metabolism , Tumor Necrosis Factor-alpha/metabolism
11.
Transgenic Res ; 18(1): 45-57, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18800235

ABSTRACT

The aim of this study was to determine the susceptibility of murine embryonic stem (mESCs) to mouse hepatitis virus (MHV-A59) and mouse minute virus (MMVp) and the effect of these viruses on germline transmission (GLT) and the serological status of recipients and pups. When recipients received 10 blastocysts, each injected with 10(0) TCID(50) MHV-A59, three out of five recipients and four out of 14 pups from three litters became seropositive. When blastocysts were injected with 10(-5) TCID(50) MMVp, all four recipients and 14 pups from four litters remained seronegative. The mESCs replicated MHV-A59 but not MMVp, MHV-A59 being cytolytic for mESCs. Exposure of mESCs to the viruses over four to five passages but not for 6 h affected GLT. Recipients were seropositive for MHV-A59 but not for MMVp when mESCs were cultured with the virus over four or five passages. The data show that GLT is affected by virus-contaminated mESCs.


Subject(s)
Blastocyst/virology , Coronavirus Infections/virology , Embryonic Stem Cells/virology , Germ Cells/physiology , Hepatitis, Viral, Animal/transmission , Minute Virus of Mice/pathogenicity , Murine hepatitis virus/pathogenicity , Parvoviridae Infections/virology , Animals , Embryo Transfer , Female , Fertilization in Vitro , Flow Cytometry , Hepatitis, Viral, Animal/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Minute Virus of Mice/isolation & purification , Minute Virus of Mice/metabolism , Murine hepatitis virus/isolation & purification , Murine hepatitis virus/metabolism , Receptors, Virus/metabolism , Seroepidemiologic Studies , Virus Replication
12.
Mol Biol Cell ; 19(6): 2544-52, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18385513

ABSTRACT

CRM1 exports proteins that carry a short leucine-rich peptide signal, the nuclear export signal (NES), from the nucleus. Regular NESs must have low affinity for CRM1 to function optimally. We previously generated artificial NESs with higher affinities for CRM1, termed supraphysiological NESs. Here we identify a supraphysiological NES in an endogenous protein, the NS2 protein of parvovirus Minute Virus of Mice (MVM). NS2 interacts with CRM1 without the requirement of RanGTP, whereas addition of RanGTP renders the complex highly stable. Mutation of a single hydrophobic residue that inactivates regular NESs lowers the affinity of the NS2 NES for CRM1 from supraphysiological to regular. Mutant MVM harboring this regular NES is compromised in viral nuclear export and productivity. In virus-infected mouse fibroblasts we observe colocalization of NS2, CRM1 and mature virions, which is dependent on the supraphysiological NS2 NES. We conclude that supraphysiological NESs exist in nature and that the supraphysiological NS2 NES has a critical role in active nuclear export of mature MVM particles before cell lysis.


Subject(s)
Cell Nucleus/metabolism , Cell Nucleus/virology , Minute Virus of Mice/metabolism , Nuclear Export Signals , Viral Nonstructural Proteins/metabolism , Active Transport, Cell Nucleus , Amino Acid Sequence , Animals , Cell Line , Humans , Karyopherins/metabolism , Mice , Minute Virus of Mice/pathogenicity , Molecular Chaperones/chemistry , Molecular Chaperones/metabolism , Molecular Sequence Data , Nuclear Envelope/metabolism , Nuclear Pore Complex Proteins/chemistry , Nuclear Pore Complex Proteins/metabolism , Protein Binding , Protein Transport , Receptors, Cytoplasmic and Nuclear/metabolism , Viral Nonstructural Proteins/chemistry , Virion/metabolism , ran GTP-Binding Protein/metabolism , Exportin 1 Protein
13.
Proc Natl Acad Sci U S A ; 105(11): 4150-5, 2008 Mar 18.
Article in English | MEDLINE | ID: mdl-18334651

ABSTRACT

In a previous study, we showed that the DNA molecule within a spherical virus (the minute virus of mice) plays an architectural role by anisotropically increasing the mechanical stiffness of the virus. A finite element model predicted that this mechanical reinforcement is a consequence of the interaction between crystallographically visible, short DNA patches and the inner capsid wall. We have now tested this model by using protein engineering. Selected amino acid side chains have been truncated to specifically remove major interactions between the capsid and the visible DNA patches, and the effect of the mutations on the stiffness of virus particles has been measured using atomic force microscopy. The mutations do not affect the stiffness of the empty capsid; however, they significantly reduce the difference in stiffness between the DNA-filled virion and the empty capsid. The results (i) reveal that intermolecular interactions between individual chemical groups contribute to the mechanical properties of a supramolecular assembly and (ii) identify specific protein-DNA interactions as the origin of the anisotropic increase in the rigidity of a virus. This study also demonstrates that it is possible to control the mechanical properties of a protein nanoparticle by the rational application of protein engineering based on a mechanical model.


Subject(s)
Protein Engineering , Viral Proteins/chemistry , Viral Proteins/metabolism , Microscopy, Atomic Force , Minute Virus of Mice/chemistry , Minute Virus of Mice/genetics , Minute Virus of Mice/metabolism , Models, Molecular , Mutation/genetics , Viral Proteins/genetics , Viral Proteins/ultrastructure , Virion/chemistry , Virion/metabolism , Virion/ultrastructure
14.
J Biol Chem ; 281(35): 25670-7, 2006 Sep 01.
Article in English | MEDLINE | ID: mdl-16822863

ABSTRACT

Sialic acid binding is required for infectious cell surface receptor recognition by parvovirus minute virus of mice (MVM). We have utilized a glycan array consisting of approximately 180 different carbohydrate structures to identify the specific sialosides recognized by the prototype (MVMp) and immunosuppressive (MVMi) strains of MVM plus three virulent mutants of MVMp, MVMp-I362S, MVMp-K368R, and MVMp-I362S/K368R. All of the MVM capsids specifically bound to three structures with a terminal sialic acid-linked alpha2-3 to a common Galbeta1-4GlcNAc motif: Neu5Acalpha2-3Galbeta1-4GlcNAcbeta1-4Galbeta1-4GlcNAc (3'SiaLN-LN), Neu5Acalpha2-3Galbeta1-4GlcNAcbeta1-3Galbeta1-4GlcNAcbeta1-3Galbeta1-4GlcNAc (3'SiaLN-LN-LN), and Neu5Acalpha2-3Galbeta1-4(Fucalpha1-3)-GlcNAcbeta1-3Galbeta1-4(Fucalpha1-3)GlcNAcbeta1-3Galbeta1-4(Fucalpha1-3)GlcNAc (sLe(x)-Le(x)-Le(x)). In addition, MVMi also recognized four multisialylated glycans with terminal alpha2-8 linkages: Neu5Acalpha2-8Neu5Acalpha2-8Neu5Acalpha ((Sia)(3)), Neu5Acalpha2-8Neu5Acalpha2-3Galbeta1-4Glc (GD3), Neu5Acalpha2-8Neu5Acalpha2-8Neu5Acalpha2-3Galbeta1-4Glc (GT3), and Neu5Acalpha2-8Neu5Acalpha2-3(GalNAcbeta1-4)Galbeta1-4Glc (GD2). Interestingly, the virulent MVMp-K368R mutant also recognized GT3. Analysis of the relative binding affinities using a surface plasmon resonance biospecific interaction (BIAcore) assay showed the wild-type MVMp and MVMi capsids binding with higher affinity to selected glycans compared with the virulent MVMp mutants. The reduced affinity of the virulent MVMp mutants are consistent with previous in vitro cell binding assays that had shown weaker binding to permissive cells compared with wild-type MVMp. This study identifies the sialic acid structures recognized by MVM. It also provides rationale for the tropism of MVM for malignant transformed cells that contain sLe(x) motifs and the neurotropism of MVMi, which is likely mediated via interactions with multisialylated glycans known to be tumor cell markers. Finally, the observations further implicate a decreased binding affinity for sialic acid in the in vivo adaptation of MVMp to a virulent phenotype.


Subject(s)
Minute Virus of Mice/metabolism , Minute Virus of Mice/pathogenicity , N-Acetylneuraminic Acid/chemistry , Binding Sites , Capsid/chemistry , Models, Molecular , Mutation , Phenotype , Polysaccharides/chemistry , Protein Binding , Virulence
15.
J Mol Biol ; 360(5): 1081-93, 2006 Jul 28.
Article in English | MEDLINE | ID: mdl-16814321

ABSTRACT

The structural and functional relevance of amino acid residues surrounding cavities within the hydrophobic core of the protein subunits that form the capsid of parvoviruses has been investigated. Several of the evolutionarily conserved, hydrophobic residues that delimit these cavities in the capsid of the minute virus of mice were replaced by other hydrophobic residues that would affect the size and/or shape of the cavity. When four or more methylene-sized groups were introduced, or six or more groups removed, capsid assembly was drastically impaired. In contrast, the introduction or removal of up to three groups had no significant effect on capsid assembly or thermostability. However, many of these mutations affected a capsid conformational transition needed for viral infectivity. Replacement of some polar residues around the largest cavity showed that capsid assembly requires a carboxylate buried within this cavity, but both aspartate and glutamate are structurally accepted. Again, only the aspartate allowed the production of infectious viruses, because of a specific role in encapsidation of the viral genome. These observations provide evidence of a remarkable structural tolerance to mutation of the hydrophobic core of the protein subunits in a viral capsid, and of an involvement of core residues and internal cavities in capsid functions needed for infectivity.


Subject(s)
Capsid Proteins/chemistry , Minute Virus of Mice/metabolism , Amino Acid Substitution , Animals , Capsid/metabolism , Capsid/ultrastructure , Capsid Proteins/genetics , Capsid Proteins/metabolism , Cells, Cultured , DNA, Viral/chemistry , Hydrophobic and Hydrophilic Interactions , Mice , Microscopy, Electron, Transmission , Minute Virus of Mice/genetics , Minute Virus of Mice/pathogenicity , Models, Molecular , Mutagenesis, Site-Directed , Mutation
16.
J Gen Virol ; 87(Pt 5): 1197-1201, 2006 May.
Article in English | MEDLINE | ID: mdl-16603521

ABSTRACT

Vectors derived from the autonomous parvovirus Minute virus of mice, MVM(p), are promising tools for the gene therapy of cancer. The validation of their in vivo anti-tumour effect is, however, hampered by the difficulty to produce high-titre stocks. In an attempt to increase vector titres, host cells were subjected to low oxygen tension (hypoxia). It has been shown that a number of viruses are produced at higher titres under these conditions. This is the case, among others, for another member of the family Parvoviridae, the erythrovirus B19 virus. Hypoxia stabilizes a hypoxia-inducible transcription factor (HIF-1alpha) that interacts with a 'hypoxia-responsive element' (HRE), the consensus sequence of which ((A)/(G)CGTG) is present in the B19 and MVM promoters. Whilst the native P4 promoter was induced weakly in hypoxia, vector production was reduced dramatically, and adding HRE elements to the P4 promoter of the vector did not alleviate this reduction. Hypoxia has many effects on cell metabolism. Therefore, even if the P4 promoter is activated, the cellular factors that are required for the completion of the parvoviral life cycle may not be expressed.


Subject(s)
Cell Hypoxia , Genetic Vectors/biosynthesis , Minute Virus of Mice/metabolism , Animals , Cell Line , Cobalt/pharmacology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Minute Virus of Mice/genetics , Promoter Regions, Genetic
17.
J Virol ; 80(2): 1015-24, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16379002

ABSTRACT

Minute virus of mice (MVM) enters the host cell via receptor-mediated endocytosis. Although endosomal processing is required, its role remains uncertain. In particular, the effect of low endosomal pH on capsid configuration and nuclear delivery of the viral genome is unclear. We have followed the progression and structural transitions of DNA full-virus capsids (FC) and empty capsids (EC) containing the VP1 and VP2 structural proteins and of VP2-only virus-like particles (VLP) during the endosomal trafficking. Three capsid rearrangements were detected in FC: externalization of the VP1 N-terminal sequence (N-VP1), cleavage of the exposed VP2 N-terminal sequence (N-VP2), and uncoating of the full-length genome. All three capsid modifications occurred simultaneously, starting as early as 30 min after internalization, and all of them were blocked by raising the endosomal pH. In particles lacking viral single-stranded DNA (EC and VLP), the N-VP2 was not exposed and thus it was not cleaved. However, the EC did externalize N-VP1 with kinetics similar to those of FC. The bulk of all the incoming particles (FC, EC, and VLP) accumulated in lysosomes without signs of lysosomal membrane destabilization. Inside lysosomes, capsid degradation was not detected, although the uncoated DNA of FC was slowly degraded. Interestingly, at any time postinfection, the amount of structural proteins of the incoming virions accumulating in the nuclear fraction was negligible. These results indicate that during the early endosomal trafficking, the MVM particles are structurally modified by low-pH-dependent mechanisms. Regardless of the structural transitions and protein composition, the majority of the entering viral particles and genomes end in lysosomes, limiting the efficiency of MVM nuclear translocation.


Subject(s)
Capsid Proteins/metabolism , Endosomes/metabolism , Minute Virus of Mice/physiology , Parvoviridae Infections/virology , Viral Structural Proteins/metabolism , Animals , Cell Line , DNA, Viral , Hydrogen-Ion Concentration , Mice , Minute Virus of Mice/metabolism , Virus Replication
18.
J Virol ; 78(19): 10685-94, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15367635

ABSTRACT

It is uncertain whether nonenveloped karyophilic virus particles may actively traffic from the nucleus outward. The unordered amino-terminal domain of the VP2 major structural protein (2Nt) of the icosahedral parvovirus minute virus of mice (MVM) is internal in empty capsids, but it is exposed outside of the shell through the fivefold axis of symmetry in virions with an encapsidated single-stranded DNA genome, as well as in empty capsids subjected to a heat-induced structural transition. In productive infections of transformed and normal fibroblasts, mature MVM virions were found to efficiently exit from the nucleus prior to cell lysis, in contrast to the extended nuclear accumulation of empty capsids. Newly formed mutant viruses lacking the three phosphorylated serine residues of 2Nt were hampered in their exit from the human transformed NB324K nucleus, in correspondence with the capacity of 2Nt to drive microinjected phosphorylated heated capsids out of the nucleus. However, in normal mouse A9 fibroblasts, in which the MVM capsid was phosphorylated at similar sites but with a much lower rate, the nuclear exit of virions and microinjected capsids harboring exposed 2Nt required the infection process and was highly sensitive to inhibition of the exportin CRM1 in the absence of a demonstrable interaction. Thus, the MVM virion exits the nucleus by accessing nonconventional export pathways relying on cell physiology that can be intensified by infection but in which the exposure of 2Nt remains essential for transport. The flexible 2Nt nuclear transport signal may illustrate a common structural solution used by nonenveloped spherical viruses to propagate in undamaged host tissues.


Subject(s)
Capsid Proteins/metabolism , Cell Nucleus/virology , Minute Virus of Mice/metabolism , Receptors, Cytoplasmic and Nuclear , Active Transport, Cell Nucleus , Animals , Capsid Proteins/chemistry , Cell Line , Cell Nucleus/metabolism , Cell Transformation, Viral , Humans , Karyopherins/metabolism , Mice , Minute Virus of Mice/growth & development , Phosphorylation , Virion/metabolism , Exportin 1 Protein
19.
Virology ; 324(2): 350-60, 2004 Jul 01.
Article in English | MEDLINE | ID: mdl-15207621

ABSTRACT

Minute virus of mice (MVM) infection is disrupted by proteasome inhibitors. Here, we show that inhibition of the ubiquitin-proteasome pathway did not affect viral entry and had influence neither on the natural proteolytic cleavage of VP2 to VP3 nor on the externalization of the N terminal of VP1. In both MG132-treated and untreated cells, MVM particles accumulated progressively in the perinuclear region. However, in MG132-treated cells, MVM was not able to penetrate into the nuclei, remaining blocked in the perinuclear region without capsid disassembly. MVM was similarly sensitive to MG132 in the two cell lines tested, A9 and NB324K. After releasing from the reversible MG132 block, MVM recovered the ability to translocate to the nuclei and replicate. Analysis of viral capsid proteins during internalization showed no evidence of capsid ubiquitination or degradation. We examined the effect of MG132 on two other parvoviruses, canine (CPV) and bovine parvovirus (BPV). Similarly to MVM, CPV infection was sensitive to MG132; however, BPV infection, as previously shown for adeno-associated viruses (AAVs), was not disturbed. These findings suggest that parvoviruses follow divergent strategies for nuclear transport, some of them requiring active proteasomes.


Subject(s)
Adenosine Triphosphatases/metabolism , Cysteine Endopeptidases/metabolism , Minute Virus of Mice/metabolism , Multienzyme Complexes/metabolism , Ubiquitin/metabolism , Active Transport, Cell Nucleus/drug effects , Animals , Capsid/metabolism , Cell Line , Cysteine Proteinase Inhibitors/pharmacology , Intracellular Space/metabolism , Leupeptins/pharmacology , Mice , Minute Virus of Mice/drug effects , Proteasome Endopeptidase Complex , Time Factors , Virus Replication
20.
J Virol ; 76(20): 10307-19, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12239307

ABSTRACT

The small nonstructural NS2 proteins of parvovirus minute virus of mice (MVMp) were previously shown to interact with the nuclear export receptor Crm1. We report here the analysis of two MVM mutant genomic clones generating NS2 proteins that are unable to interact with Crm1 as a result of amino acid substitutions within their nuclear export signal (NES) sequences. Upon transfection of human and mouse cells, the MVM-NES21 and MVM-NES22 mutant genomic clones were proficient in synthesis of the four virus-encoded proteins. While the MVM-NES22 clone was further able to produce infectious mutant virions, no virus could be recovered from cells transfected with the MVM-NES21 clone. Whereas the defect of MVM-NES21 appeared to be complex, the phenotype of MVM-NES22 could be traced back to a novel distinct NS2 function. Infection of mouse cells with the MVM-NES22 mutant led to stronger nuclear retention not only of the NS2 proteins but also of infectious progeny MVM particles. This nuclear sequestration correlated with a severe delay in the release of mutant virions in the medium and with prolonged survival of the infected cell populations compared with wild-type virus-treated cultures. This defect could explain, at least in part, the small size of the plaques generated by the MVM-NES22 mutant when assayed on mouse indicator cells. Altogether, our data indicate that the interaction of MVMp NS2 proteins with the nuclear export receptor Crm1 plays a critical role at a late stage of the parvovirus life cycle involved in release of progeny viruses.


Subject(s)
Karyopherins/metabolism , Minute Virus of Mice/metabolism , Receptors, Cytoplasmic and Nuclear , Viral Nonstructural Proteins/metabolism , Active Transport, Cell Nucleus , Animals , Cell Line , Cell Nucleus/metabolism , Mice , Minute Virus of Mice/genetics , Mutagenesis , Transfection , Viral Nonstructural Proteins/genetics , Virion , Exportin 1 Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...