Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters










Publication year range
1.
PLoS Pathog ; 17(1): e1009231, 2021 01.
Article in English | MEDLINE | ID: mdl-33471863

ABSTRACT

Liquid-liquid phase separation (LLPS) can drive formation of diverse and essential macromolecular structures, including those specified by viruses. Kaposi's Sarcoma-Associated Herpesvirus (KSHV) genomes associate with the viral encoded Latency-Associated Nuclear Antigen (LANA) to form stable nuclear bodies (NBs) during latent infection. Here, we show that LANA-NB formation and KSHV genome conformation involves LLPS. Using LLPS disrupting solvents, we show that LANA-NBs are partially disrupted, while DAXX and PML foci are highly resistant. LLPS disruption altered the LANA-dependent KSHV chromosome conformation but did not stimulate lytic reactivation. We found that LANA-NBs undergo major morphological transformation during KSHV lytic reactivation to form LANA-associated replication compartments encompassing KSHV DNA. DAXX colocalizes with the LANA-NBs during latency but is evicted from the LANA-associated lytic replication compartments. These findings indicate the LANA-NBs are dynamic super-molecular nuclear structures that partly depend on LLPS and undergo morphological transitions corresponding to the different modes of viral replication.


Subject(s)
Antigens, Viral/chemistry , Co-Repressor Proteins/metabolism , Genome, Viral/genetics , Herpesvirus 8, Human/genetics , Intranuclear Inclusion Bodies/metabolism , Molecular Chaperones/metabolism , Nuclear Proteins/chemistry , Sarcoma, Kaposi/virology , Antigens, Viral/genetics , Cell Line, Tumor , Herpesvirus 8, Human/physiology , Histones/metabolism , Humans , Inclusion Bodies, Viral/chemistry , Inclusion Bodies, Viral/metabolism , Intranuclear Inclusion Bodies/chemistry , Latent Infection , Liquid-Liquid Extraction , Nuclear Proteins/genetics , Plasmids/genetics , Virus Latency , Virus Replication
2.
Adv Exp Med Biol ; 1215: 111-127, 2019.
Article in English | MEDLINE | ID: mdl-31317498

ABSTRACT

Replication and assembly of many viruses occur in viral factories which are specialized intracellular compartments formed during viral infection. For rabies virus, those viral factories are called Negri bodies (NBs). NBs are cytoplasmic inclusion bodies in which viral RNAs (mRNAs as well as genomic and antigenomic RNAs) are synthesized. NBs are spherical, they can fuse together, and can reversibly deform when encountering a physical barrier. All these characteristics are similar to those of eukaryotic membrane-less liquid organelles which contribute to the compartmentalization of the cell interior. Indeed, the liquid nature of NBs has been confirmed by FRAP experiments. The co-expression of rabies virus nucleoprotein N and phosphoprotein P is sufficient to induce the formation of cytoplasmic inclusions recapitulating NBs properties. Remarkably, P and N have features similar to those of cellular proteins involved in liquid organelles formation: N is an RNA-binding protein and P contains intrinsically disordered domains. An overview of the literature indicates that formation of liquid viral factories by phase separation is probably common among Mononegavirales. This allows specific recruitment and concentration of viral proteins. Finally, as virus-associated molecular patterns recognized by cellular sensors of RNA virus replication are probably essentially present in the viral factory, there should be a subtle interplay (which remains to be characterized) between those liquid structures and the cellular proteins which trigger the innate immune response.


Subject(s)
Inclusion Bodies, Viral , Rabies virus , Inclusion Bodies, Viral/chemistry , Inclusion Bodies, Viral/metabolism , RNA, Viral/biosynthesis , Rabies virus/physiology , Viral Proteins/metabolism , Virus Replication
3.
J Biosci ; 44(2)2019 Jun.
Article in English | MEDLINE | ID: mdl-31180044

ABSTRACT

ErbB-3 binding protein 1 (Ebp1) is a host protein which binds ErbB-3 receptor to induce signalling events for cell growth regulation. In addition, Ebp1 also interacts with ribonucleoprotein complexes. In recent times, Ebp1 was found to play an antagonistic role in viral infections caused by Influenza and Rinderpest viruses. In our present work we have tried to understand the role of Ebp1 in Chandipura virus (CHPV) infection. We have observed an induction in Ebp1 expression upon CHPV infection similar to other viruses. However, unlike other viruses an overexpressed Ebp1 only reduces viral protein expression, but does not affect its progeny formation. Additionally, this effect is being carried out in an indirect manner, as there is no interaction between Ebp1 and viral proteins. This is despite Ebp1's presence in viral inclusion bodies.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Host-Pathogen Interactions/genetics , Neurons/metabolism , RNA-Binding Proteins/genetics , Vesiculovirus/genetics , Virus Replication , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Line, Tumor , Chlorocebus aethiops , Gene Expression Regulation , Humans , Inclusion Bodies, Viral/chemistry , Neurons/virology , Plasmids/chemistry , Plasmids/metabolism , RNA-Binding Proteins/metabolism , Signal Transduction , Transfection , Vero Cells , Vesiculovirus/growth & development , Vesiculovirus/metabolism , Viral Plaque Assay
4.
mSphere ; 3(2)2018 04 25.
Article in English | MEDLINE | ID: mdl-29669885

ABSTRACT

Heteroaryldihydropyrimidines (HAPs) are compounds that inhibit hepatitis B virus (HBV) replication by modulating viral capsid assembly. While their biophysical effects on capsid assembly in vitro have been previously studied, the effect of HAP treatment on capsid protein (Cp) in individual HBV-infected cells remains unknown. We report here that the HAP Bay 38-7690 promotes aggregation of recombinant Cp in vitro and causes a time- and dose-dependent decrease of Cp in infected cells, consistent with previously studied HAPs. Interestingly, immunofluorescence analysis showed Cp aggregating in nuclear foci of Bay 38-7690-treated infected cells in a time- and dose-dependent manner. We found these foci to be associated with promyelocytic leukemia (PML) nuclear bodies (NBs), which are structures that affect many cellular functions, including DNA damage response, transcription, apoptosis, and antiviral responses. Cp aggregation is not an artifact of the cell system used, as it is observed in HBV-expressing HepAD38 cells, in HepG2 cells transfected with an HBV-expressing plasmid, and in HepG2-NTCP cells infected with HBV. Use of a Cp overexpression vector without HBV sequences shows that aggregation is independent of viral replication, and use of an HBV-expressing plasmid harboring a HAP resistance mutation in Cp abrogated the aggregation, demonstrating that the effect is due to direct compound-Cp interactions. These studies provide novel insight into the effects of HAP-based treatment at a single-cell level.IMPORTANCE Despite the availability of effective vaccines and treatments, HBV remains a significant global health concern, with more than 240 million individuals chronically infected. Current treatments are highly effective at controlling viral replication and disease progression but rarely cure infections. Therefore, much emphasis is being placed on finding therapeutics with new drug targets, such as viral gene expression, covalently closed circular DNA formation and stability, capsid formation, and host immune modulators, with the ultimate goal of an HBV cure. Understanding the mechanisms by which novel antiviral agents act will be imperative for the development of curative HBV therapies.


Subject(s)
Antiviral Agents/pharmacology , Capsid Proteins/chemistry , Hepatitis B virus/drug effects , Inclusion Bodies, Viral/chemistry , Protein Aggregates/drug effects , Pyridines/pharmacology , Pyrimidines/pharmacology , Capsid/chemistry , Capsid/drug effects , Capsid Proteins/genetics , Fluorescent Antibody Technique , Hep G2 Cells , Hepatitis B/drug therapy , Hepatitis B virus/physiology , Humans , Recombinant Proteins/chemistry , Virus Assembly/drug effects , Virus Replication/drug effects
5.
J Virol ; 90(16): 7268-7284, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27252530

ABSTRACT

UNLABELLED: A hallmark of Ebola virus (EBOV) infection is the formation of viral inclusions in the cytoplasm of infected cells. These viral inclusions contain the EBOV nucleocapsids and are sites of viral replication and nucleocapsid maturation. Although there is growing evidence that viral inclusions create a protected environment that fosters EBOV replication, little is known about their role in the host response to infection. The cellular stress response is an effective antiviral strategy that leads to stress granule (SG) formation and translational arrest mediated by the phosphorylation of a translation initiation factor, the α subunit of eukaryotic initiation factor 2 (eIF2α). Here, we show that selected SG proteins are sequestered within EBOV inclusions, where they form distinct granules that colocalize with viral RNA. These inclusion-bound (IB) granules are functionally and structurally different from canonical SGs. Formation of IB granules does not indicate translational arrest in the infected cells. We further show that EBOV does not induce formation of canonical SGs or eIF2α phosphorylation at any time postinfection but is unable to fully inhibit SG formation induced by different exogenous stressors, including sodium arsenite, heat, and hippuristanol. Despite the sequestration of SG marker proteins into IB granules, canonical SGs are unable to form within inclusions, which we propose might be mediated by a novel function of VP35, which disrupts SG formation. This function is independent of VP35's RNA binding activity. Further studies aim to reveal the mechanism for SG protein sequestration and precise function within inclusions. IMPORTANCE: Although progress has been made developing antiviral therapeutics and vaccines against the highly pathogenic Ebola virus (EBOV), the cellular mechanisms involved in EBOV infection are still largely unknown. To better understand these intracellular events, we investigated the cellular stress response, an antiviral pathway manipulated by many viruses. We show that EBOV does not induce formation of stress granules (SGs) in infected cells and is therefore unrestricted by their concomitant translational arrest. We identified SG proteins sequestered within viral inclusions, which did not impair protein translation. We further show that EBOV is unable to block SG formation triggered by exogenous stress early in infection. These findings provide insight into potential targets of therapeutic intervention. Additionally, we identified a novel function of the interferon antagonist VP35, which is able to disrupt SG formation.


Subject(s)
Cytoplasm/virology , Ebolavirus/growth & development , Host-Pathogen Interactions , Immunologic Factors/analysis , Inclusion Bodies, Viral/virology , Stress, Physiological , Viral Regulatory and Accessory Proteins/metabolism , Animals , Cell Line , Cytoplasmic Granules/metabolism , Ebolavirus/immunology , Heat-Shock Proteins/analysis , Humans , Inclusion Bodies, Viral/chemistry
6.
Clin Chim Acta ; 448: 206-10, 2015 Aug 25.
Article in English | MEDLINE | ID: mdl-26164385

ABSTRACT

BACKGROUND: Development of indirect enzyme-linked immunosorbent assays (ELISAs) often utilizes synthetic peptides or recombinant proteins from Escherichia coli as immobilized antigens. Because inclusion bodies (IBs) formed during recombinant protein expression in E. coli are commonly thought as misfolded aggregates, only refolded proteins from IBs are used to develop new or in-house diagnostic assays. However, the promising utilities of IBs as nanomaterials and immobilized enzymes as shown in recent studies have led us to explore the potential use of IBs of recombinant Epstein-Barr virus viral capsid antigen p18 (VCA p18) as immobilized antigens in ELISAs for serologic detection of nasopharyngeal carcinoma (NPC). METHODS: Thioredoxin fusion VCA p18 (VCA-Trx) and IBs of VCA p18 without fusion tags (VCA-IBs) were purified from E. coli. The diagnostic performances of IgG/VCA-IBs, IgG/VCA-Denat-IBs (using VCA-IBs coated in 8mol/l urea), IgG/VCA-Trx, and IgG/VCA-Peptide assays were compared by screening 100 NPC case-control pairs. RESULTS: The IgG/VCA-Denat-IBs assay showed the best area under the receiver operating characteristic curve (AUC: 0.802; p<0.05), while the AUCs for the IgG/VCA-IBs, IgG/VCA-Trx, and IgG/VCA-Peptide assays were comparable (AUC: 0.740, 0.727, and 0.741, respectively). CONCLUSION: We improved the diagnostic performance of the ELISA significantly using IBs of recombinant VCA p18.


Subject(s)
Antigens, Viral/immunology , Capsid Proteins/immunology , Enzyme-Linked Immunosorbent Assay , Immobilized Proteins/immunology , Inclusion Bodies, Viral/immunology , Nasopharyngeal Neoplasms/diagnosis , Nasopharyngeal Neoplasms/virology , Antigens, Viral/chemistry , Capsid Proteins/chemistry , Humans , Immobilized Proteins/chemistry , Inclusion Bodies, Viral/chemistry , Nasopharyngeal Neoplasms/immunology , Recombinant Proteins/chemistry , Recombinant Proteins/immunology
7.
Protein Expr Purif ; 113: 35-43, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25959459

ABSTRACT

Japanese Encephalitis (JE) is a mosquito borne arboviral infection caused by Japanese Encephalitis Virus (JEV). It is a major cause of viral encephalitis in Asian countries including India. In the present study, we have used a Tymovirus [i.e. Physalis Mottle Virus (PhMV) coat protein (CP)], which forms virus like particles (VLPs) as a template to display immunodominant epitopes of JEV envelope (E) protein. The immunodominant epitopes of JEV were inserted at the N-terminus of the wild type PhMV CP, and these constructs were cloned and expressed in Escherichia coli. The chimeric proteins were purified from the inclusion bodies and evaluated for VLP formation. The purified protein was identified by Western blotting and VLP formation was studied and confirmed by transmission electron microscopy and dynamic light scattering. Finally, the immunogenicity was studied in mice. Our results indicate that the chimeric protein with JEV epitopes assembled efficiently to form VLPs generating neutralizing antibodies. Hence, we report the purified chimeric VLP would be a potent vaccine candidate, which needs to be evaluated in a mouse challenge model.


Subject(s)
Capsid Proteins/metabolism , Immunodominant Epitopes/metabolism , Inclusion Bodies, Viral/metabolism , Membrane Glycoproteins/metabolism , Recombinant Fusion Proteins/metabolism , Tymovirus/genetics , Viral Envelope Proteins/metabolism , Animals , Antibodies, Viral/blood , Capsid Proteins/chemistry , Capsid Proteins/genetics , Capsid Proteins/immunology , Immunodominant Epitopes/chemistry , Immunodominant Epitopes/genetics , Immunodominant Epitopes/immunology , Inclusion Bodies, Viral/chemistry , Inclusion Bodies, Viral/immunology , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , Protein Refolding , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology
8.
J Virol ; 89(3): 1640-51, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25410852

ABSTRACT

UNLABELLED: The rabies virus (RABV) phosphoprotein P is a multifunctional protein: it plays an essential role in viral transcription and replication, and in addition, RABV P has been identified as an interferon antagonist. Here, a yeast two-hybrid screen revealed that RABV P interacts with the focal adhesion kinase (FAK). The binding involved the 106-to-131 domain, corresponding to the dimerization domain of P and the C-terminal domain of FAK containing the proline-rich domains PRR2 and PRR3. The P-FAK interaction was confirmed in infected cells by coimmunoprecipitation and colocalization of FAK with P in Negri bodies. By alanine scanning, we identified a single mutation in the P protein that abolishes this interaction. The mutant virus containing a substitution of Ala for Arg in position 109 in P (P.R109A), which did not interact with FAK, is affected at a posttranscriptional step involving protein synthesis and viral RNA replication. Furthermore, FAK depletion inhibited viral protein expression in infected cells. This provides the first evidence of an interaction of RABV with FAK that positively regulates infection. IMPORTANCE: Rabies virus exhibits a small genome that encodes a limited number of viral proteins. To maintain efficient virus replication, some of them are multifunctional, such as the phosphoprotein P. We and others have shown that P establishes complex networks of interactions with host cell components. These interactions have revealed much about the role of P and about host-pathogen interactions in infected cells. Here, we identified another cellular partner of P, the focal adhesion kinase (FAK). Our data shed light on the implication of FAK in RABV infection and provide evidence that P-FAK interaction has a proviral function.


Subject(s)
Focal Adhesion Protein-Tyrosine Kinases/metabolism , Host-Pathogen Interactions , Phosphoproteins/metabolism , Protein Interaction Mapping , Rabies virus/physiology , Viral Structural Proteins/metabolism , Virus Replication , Animals , Cell Line , DNA Mutational Analysis , Humans , Immunoprecipitation , Inclusion Bodies, Viral/chemistry , Inclusion Bodies, Viral/virology , Microscopy, Confocal , Molecular Chaperones , Mutagenesis, Site-Directed , Protein Binding , Two-Hybrid System Techniques
9.
Mol Plant Microbe Interact ; 27(3): 215-26, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24405034

ABSTRACT

A unique feature shared by all plant viruses of the Potyviridae family is the induction of characteristic pinwheel-shaped inclusion bodies in the cytoplasm of infected cells. These cylindrical inclusions are composed of the viral-encoded cylindrical inclusion helicase (CI protein). Its helicase activity was characterized and its involvement in replication demonstrated through different reverse genetics approaches. In addition to replication, the CI protein is also involved in cell-to-cell and long-distance movements, possibly through interactions with the recently discovered viral P3N-PIPO protein. Studies over the past two decades demonstrate that the CI protein is present in several cellular compartments interacting with viral and plant protein partners likely involved in its various roles in different steps of viral infection. Furthermore, the CI protein acts as an avirulence factor in gene-for-gene interactions with dominant-resistance host genes and as a recessive-resistance overcoming factor. Although a significant amount of data concerning the potential functions and subcellular localization of this protein has been published, no synthetic review is available on this important multifunctional protein. In this review, we compile and integrate all information relevant to the current understanding of this viral protein structure and function and present a mode of action for CI, combining replication and movement.


Subject(s)
Genome, Viral/physiology , Inclusion Bodies, Viral/metabolism , Plant Diseases/virology , Plants/virology , Potyviridae/enzymology , RNA Helicases/metabolism , Amino Acid Sequence , Host-Pathogen Interactions , Inclusion Bodies, Viral/chemistry , Inclusion Bodies, Viral/ultrastructure , Models, Biological , Molecular Sequence Data , Plant Viruses/enzymology , Plant Viruses/physiology , Plant Viruses/ultrastructure , Plants/ultrastructure , Plasmodesmata/ultrastructure , Plasmodesmata/virology , Potyviridae/physiology , Potyviridae/ultrastructure , RNA Helicases/chemistry , RNA Helicases/ultrastructure , Sequence Alignment , Viral Proteins/chemistry , Viral Proteins/metabolism , Viral Proteins/ultrastructure
10.
Virol J ; 10: 133, 2013 Apr 27.
Article in English | MEDLINE | ID: mdl-23622425

ABSTRACT

BACKGROUND: Aquareovirus particle is comprised of central core and outer capsid, which is built by seven structural proteins (VP1-VP7). The protein VP6 has been identified to be a clamp protein of stabilizing inner core frame VP3, and bridging outer shell protein VP5. However, the biological properties of VP6 in viral life cycle remain unknown. RESULTS: The recombinant VP6 (rVP6) of aquareovirus was expressed in E. coli, and the polyclonal antibody against VP6 was generated by using purified rVP6 in this study. Following the preparation of VP6 antibody, the VP6 component in aquareovirus infected cells and purified viral particles was detected by Immunoblotting (IB) assay. Furthermore, using Immunofluorescence (IF) microscopy, singly transfected VP6 protein was observed to exhibit a diffuse distribution mainly in the cytoplasm, while it appeared inclusion phenotype in infected cells. Meanwhile, inclusion structures were also identified when VP6 was coexpressed with nonstructural protein NS80 in cotransfected cells. CONCLUSIONS: VP6 can be recruited by NS80 to its inclusions in both infected and transfected cells. The colocalization of VP6 and NS80 is corresponding to their homologous proteins σ2 and µNS in MRV. Our results suggest that VP6 may play a significant role in viral replication and particle assembly.


Subject(s)
Inclusion Bodies, Viral/chemistry , Reoviridae/physiology , Viral Proteins/analysis , Virus Assembly , Virus Replication , Animals , Antibodies, Viral , Cell Line , Immunoblotting , Microscopy, Fluorescence , Protein Binding , Protein Interaction Mapping
11.
J Biotechnol ; 155(3): 284-6, 2011 Sep 20.
Article in English | MEDLINE | ID: mdl-21802457

ABSTRACT

We have recently developed a versatile tagging system (IC-tagging) that causes relocation of the tagged proteins to ARV muNS-derived intracellular globular inclusions. In the present study we demonstrate (i) that the IC-tag can be successfully fused either to the amino or carboxyl terminus of the protein to be tagged and (ii) that IC-tagged proteins are able to interact between them and perform complex reactions that require such interactions while integrated into muNS inclusions, increasing the versatility of the IC-tagging system. Also, our studies with the DsRed protein add some light on the structure/function relationship of the evolution of DsRed chromophore.


Subject(s)
Inclusion Bodies, Viral/metabolism , Molecular Probe Techniques , Viral Proteins/metabolism , Animals , Chlorocebus aethiops , Immunohistochemistry , Inclusion Bodies, Viral/chemistry , Luminescent Proteins/chemistry , Luminescent Proteins/metabolism , Microscopy, Fluorescence , Models, Molecular , Orthoreovirus, Avian/chemistry , Protein Binding , Protein Structure, Tertiary , Vero Cells , Viral Proteins/chemistry , Red Fluorescent Protein
12.
Traffic ; 12(9): 1179-95, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21736684

ABSTRACT

The viral factories of mammalian reovirus (MRV) are cytoplasmic structures that serve as sites of viral genome replication and particle assembly. A 721-aa MRV non-structural protein, µNS, forms the factory matrix and recruits other viral proteins to these structures. In this report, we show that µNS contains a conserved C-proximal sequence (711-LIDFS-715) that is similar to known clathrin-box motifs and is required for recruitment of clathrin to viral factories. Clathrin recruitment by µNS occurs independently of infecting MRV particles or other MRV proteins. Ala substitution for a single Leu residue (mutation L711A) within the putative clathrin-binding motif of µNS inhibits clathrin recruitment, but does not prevent formation or expansion of viral factories. Notably, clathrin-dependent cellular functions, including both endocytosis and secretion, are disrupted in cells infected with MRV expressing wild-type, but not L711A, µNS. These results identify µNS as a novel adaptor-like protein that recruits cellular clathrin to viral factories, disrupting normal functions of clathrin in cellular membrane trafficking. To our knowledge, this is the only viral or bacterial protein yet shown to interfere with clathrin functions in this manner. The results additionally establish a new approach for studies of clathrin functions, based on µNS-mediated sequestration.


Subject(s)
Clathrin/metabolism , Inclusion Bodies, Viral/metabolism , Orthoreovirus, Mammalian/physiology , Protein Transport/physiology , Reoviridae Infections/metabolism , Viral Nonstructural Proteins/metabolism , Adaptor Protein Complex 1/genetics , Adaptor Protein Complex 1/metabolism , Adaptor Protein Complex 2/genetics , Adaptor Protein Complex 2/metabolism , Animals , Cell Line , Clathrin/chemistry , Clathrin/genetics , Coated Pits, Cell-Membrane/metabolism , Inclusion Bodies, Viral/chemistry , Mice , Orthoreovirus, Mammalian/pathogenicity , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Viral Nonstructural Proteins/genetics , Virus Replication
13.
Virol J ; 7: 350, 2010 Nov 29.
Article in English | MEDLINE | ID: mdl-21114853

ABSTRACT

BACKGROUND: During rotavirus replication cycle, electron-dense cytoplasmic inclusions named viroplasms are formed, and two non-structural proteins, NSP2 and NSP5, have been shown to localize in these membrane-free structures. In these inclusions, replication of dsRNA and packaging of pre-virion particles occur. Despite the importance of viroplasms in the replication cycle of rotavirus, the information regarding their formation, and the possible sites of their nucleation during the early stages of infection is scarce. Here, we analyzed the formation of viroplasms after infection of MA104 cells with the rotavirus strain RRV, using different multiplicities of infection (MOI), and different times post-infection. The possibility that viroplasms formation is nucleated by the entering viral particles was investigated using fluorescently labeled purified rotavirus particles. RESULTS: The immunofluorescent detection of viroplasms, using antibodies specific to NSP2 showed that both the number and size of viroplasms increased during infection, and depend on the MOI used. Small-size viroplasms predominated independently of the MOI or time post-infection, although at MOI's of 2.5 and 10 the proportion of larger viroplasms increased. Purified RRV particles were successfully labeled with the Cy5 mono reactive dye, without decrease in virus infectivity, and the labeled viruses were clearly observed by confocal microscope. PAGE gel analysis showed that most viral proteins were labeled; including the intermediate capsid protein VP6. Only 2 out of 117 Cy5-labeled virus particles colocalized with newly formed viroplasms at 4 hours post-infection. CONCLUSIONS: The results presented in this work suggest that during rotavirus infection the number and size of viroplasm increases in an MOI-dependent manner. The Cy5 in vitro labeled virus particles were not found to colocalize with newly formed viroplasms, suggesting that they are not involved in viroplasm nucleation.


Subject(s)
Inclusion Bodies, Viral/virology , Rotavirus/growth & development , Rotavirus/pathogenicity , Virus Assembly , Virus Replication , Animals , Capsid Proteins/analysis , Cell Line , Inclusion Bodies, Viral/chemistry , Macaca mulatta , Microscopy, Confocal , Viral Nonstructural Proteins/analysis
14.
Virus Res ; 152(1-2): 96-103, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20600394

ABSTRACT

Mal de Río Cuarto virus (MRCV) is a plant virus of the genus Fijivirus within the family Reoviridae that infects several monocotyledonous species and is transmitted by planthoppers in a persistent and propagative manner. Other members of the family replicate in viral inclusion bodies (VIBs) termed viroplasms that are formed in the cytoplasm of infected plant and insect cells. In this study, the protein coded by the first ORF of MRCV segment S9 (P9-1) was shown to establish cytoplasmic inclusion bodies resembling viroplasms after transfection of Spodoptera frugiperda insect cells. In accordance, MRCV P9-1 self-associates giving rise to high molecular weight complexes when expressed in bacteria. Strong self-interaction was also evidenced by yeast two-hybrid assays. Furthermore, biochemical characterization showed that MRCV P9-1 bound single stranded RNA and had ATPase activity. Finally, the MRCV P9-1 region required for the formation of VIB-like structures was mapped to the protein carboxy-terminal half. This extensive functional and biochemical characterization of MRCV P9-1 revealed further similarities between plant and animal reovirus viroplasm proteins.


Subject(s)
Inclusion Bodies, Viral/metabolism , Reoviridae/metabolism , Spodoptera/virology , Viral Proteins/metabolism , Amino Acid Motifs , Animals , Inclusion Bodies, Viral/chemistry , Inclusion Bodies, Viral/genetics , Open Reading Frames , Reoviridae/chemistry , Reoviridae/genetics , Viral Proteins/chemistry , Viral Proteins/genetics
15.
J Virol ; 83(7): 2892-906, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19176625

ABSTRACT

Mammalian reoviruses are nonenveloped particles containing a genome of 10 double-stranded RNA (dsRNA) gene segments. Reovirus replication occurs within viral inclusions, which are specialized nonmembranous cytoplasmic organelles formed by viral nonstructural and structural proteins. Although these structures serve as sites for several major events in the reovirus life cycle, including dsRNA synthesis, gene segment assortment, and genome encapsidation, biochemical mechanisms of virion morphogenesis within inclusions have not been elucidated because much remains unknown about inclusion anatomy and functional organization. To better understand how inclusions support viral replication, we have used RNA interference (RNAi) and reverse genetics to define functional domains in two inclusion-associated proteins, muNS and mu2, which are interacting partners essential for inclusion development and viral replication. Removal of muNS N-terminal sequences required for association with mu2 or another muNS-binding protein, sigmaNS, prevented the capacity of muNS to support viral replication without affecting inclusion formation, indicating that muNS-mu2 and muNS-sigmaNS interactions are necessary for inclusion function but not establishment. In contrast, introduction of changes into the muNS C-terminal region, including sequences that form a putative oligomerization domain, precluded inclusion formation as well as viral replication. Mutational analysis of mu2 revealed a critical dependence of viral replication on an intact nucleotide/RNA triphosphatase domain and an N-terminal cluster of basic amino acid residues conforming to a nuclear localization motif. Another domain in mu2 governs the capacity of viral inclusions to affiliate with microtubules and thereby modulates inclusion morphology, either globular or filamentous. However, viral variants altered in inclusion morphology displayed equivalent replication efficiency. These studies reveal a modular functional organization of inclusion proteins muNS and mu2, define the importance of specific amino acid sequences and motifs in these proteins for viral replication, and demonstrate the utility of complementary RNAi-based and reverse genetic approaches for studies of reovirus replication proteins.


Subject(s)
Reoviridae/physiology , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Replication , Animals , Cell Line , DNA Mutational Analysis , Gene Silencing , Humans , Inclusion Bodies, Viral/chemistry , Inclusion Bodies, Viral/virology , Mice , RNA Interference , Sequence Deletion
16.
Virology ; 386(1): 168-82, 2009 Mar 30.
Article in English | MEDLINE | ID: mdl-19178924

ABSTRACT

We examined the structure of lipid-raft membranes in respiratory syncytial virus infected cells. Cholesterol depletion studies using methyl-beta-cyclodextrin suggested that membrane cholesterol was required for virus filament formation, but not inclusion bodies. In addition, virus filament formation coincided with elevated 3-hydroxy-3-methylglutaryl-coenzyme A reductase expression, suggesting an increase in requirement for endogenous cholesterol synthesis during virus assembly. Lipid raft membranes were examined by mass spectrometry, which suggested that virus infection induced subtle changes in the lipid composition of these membrane structures. This analysis revealed increased levels of raft-associated phosphatidylinositol (PI) and phosphorylated PI during RSV infection, which correlated with the appearance of phosphatidylinositol 4,5-bisphosphate and phosphatidylinositol 3,4,5-triphosphate (PIP(3)) within virus inclusion bodies, and inhibiting the synthesis of PIP(3) impaired the formation of progeny virus. Collectively, our analysis suggests that RSV infection induces specific changes in the composition of raft-associated lipids, and that these changes play an important role in virus maturation.


Subject(s)
Lipid Metabolism , Lipids/analysis , Membrane Microdomains/chemistry , Respiratory Syncytial Viruses/physiology , Virus Assembly , Cell Line , Humans , Hydroxymethylglutaryl-CoA-Reductases, NADP-dependent/metabolism , Inclusion Bodies, Viral/chemistry , Mass Spectrometry
17.
J Electron Microsc (Tokyo) ; 56(3): 77-81, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17967810

ABSTRACT

When rice dwarf virus (RDV), a member of the Reoviridae family, infects leafhopper cells, formation of protruding tubules composed of nonstructural viral protein Pns 10 can be observed. We examined the three-dimensional (3D) structure of these tubules containing RDV particles using electron tomography. The thin section of RDV-infected leafhopper vector cells in monolayers was subjected to double-tilt tomography. The tomographic 3D map provides a more reliable estimation of the real dimensions of the structure compared with the 2D image of the thin section. Docking of particle models made from atomic coordinates of RDV into the tomogram revealed that the inner diameter of the tubule was close to the outer diameter of the RDV particle. Fourier-transform of the reconstituted tubule image from the purified Pns 10 protein in vitro revealed a helical structure of the tubule.


Subject(s)
Reoviridae/ultrastructure , Animals , Hemiptera/virology , Imaging, Three-Dimensional , Inclusion Bodies, Viral/chemistry , Inclusion Bodies, Viral/ultrastructure , Insect Vectors , Microscopy, Electron, Transmission , Models, Molecular , Oryza/virology , Plant Diseases/virology , Reoviridae/chemistry , Reoviridae/pathogenicity , Tomography , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/ultrastructure
18.
Virol J ; 4: 76, 2007 Jul 24.
Article in English | MEDLINE | ID: mdl-17650322

ABSTRACT

BACKGROUND: Profilins are critical to cytoskeletal dynamics in eukaryotes; however, little is known about their viral counterparts. In this study, a poxviral profilin homolog, ectromelia virus strain Moscow gene 141 (ECTV-PH), was investigated by a variety of experimental and bioinformatics techniques to characterize its interactions with cellular and viral proteins. RESULTS: Profilin-like proteins are encoded by all orthopoxviruses sequenced to date, and share over 90% amino acid (aa) identity. Sequence comparisons show highest similarity to mammalian type 1 profilins; however, a conserved 3 aa deletion in mammalian type 3 and poxviral profilins suggests that these homologs may be more closely related. Structural analysis shows that ECTV-PH can be successfully modelled onto both the profilin 1 crystal structure and profilin 3 homology model, though few of the surface residues thought to be required for binding actin, poly(L-proline), and PIP2 are conserved. Immunoprecipitation and mass spectrometry identified two proteins that interact with ECTV-PH within infected cells: alpha-tropomyosin, a 38 kDa cellular actin-binding protein, and the 84 kDa product of vaccinia virus strain Western Reserve (VACV-WR) 148, which is the truncated VACV counterpart of the orthopoxvirus A-type inclusion (ATI) protein. Western and far-western blots demonstrated that the interaction with alpha-tropomyosin is direct, and immunofluorescence experiments suggest that ECTV-PH and alpha-tropomyosin may colocalize to structures that resemble actin tails and cellular protrusions. Sequence comparisons of the poxviral ATI proteins show that although full-length orthologs are only present in cowpox and ectromelia viruses, an ~ 700 aa truncated ATI protein is conserved in over 90% of sequenced orthopoxviruses. Immunofluorescence studies indicate that ECTV-PH localizes to cytoplasmic inclusion bodies formed by both truncated and full-length versions of the viral ATI protein. Furthermore, colocalization of ECTV-PH and truncated ATI protein to protrusions from the cell surface was observed. CONCLUSION: These results suggest a role for ECTV-PH in intracellular transport of viral proteins or intercellular spread of the virus. Broader implications include better understanding of the virus-host relationship and mechanisms by which cells organize and control the actin cytoskeleton.


Subject(s)
Ectromelia virus/physiology , Profilins/metabolism , Tropomyosin/metabolism , Viral Proteins/metabolism , Actins/chemistry , Animals , Blotting, Western , Cell Line , Chlorocebus aethiops , Cytoplasm/chemistry , Ectromelia virus/genetics , Immunoprecipitation , Inclusion Bodies, Viral/chemistry , Microscopy, Confocal , Microscopy, Fluorescence , Models, Molecular , Phylogeny , Profilins/genetics , Protein Binding , Protein Structure, Tertiary , Sequence Homology, Amino Acid
19.
BMC Microbiol ; 7: 38, 2007 May 15.
Article in English | MEDLINE | ID: mdl-17504539

ABSTRACT

BACKGROUND: Although more than 100 Chlamydia pneumoniae hypothetical proteins have been predicted to be inclusion membrane proteins, only a few have been experimentally demonstrated to be in the inclusion membrane. Using antibodies raised with fusion proteins, we characterized four such hypothetical proteins encoded by two gene clusters (Cpn0146-147 and Cpn0284-285) in the C. pneumoniae genome. RESULTS: Cpn0146 and 0147 were detected in the inclusion membrane while Cpn0284 and 0285 inside inclusion and mainly associated with reticulate bodies although all four proteins contain an N-terminal bi-lobed hydrophobic region, a signature motif assigned to inclusion membrane proteins. These four hypothetical proteins were only detected in cells infected with C. pneumoniae but not other chlamydial species, with Cpn0147 at 6 hours and Cpn0146, 0284 & 0285 at 24 hours after infection. Cpn0146 & 147 but not Cpn0284 and 285 co-localized with a host cell endoplasmic reticulum marker, a property known to be possessed by some chlamydial inclusion membrane proteins, when expressed in the host cell cytosol via transgenes. However, the endoplasmic reticulum localization of the C. pneumoniae inclusion membrane proteins did not result in inhibition of the subsequent C. pneumoniae infection. CONCLUSION: The hypothetical proteins Cpn0146 & 0147 were localized in the C. pneumoniae inclusion membrane while Cpn0284 & 0285 within the inclusion although all four were predicted to be Inc proteins, suggesting the need to experimentally characterize the predicted Inc proteins.


Subject(s)
Bacterial Proteins/analysis , Chlamydophila pneumoniae/chemistry , Inclusion Bodies, Viral/chemistry , Membrane Proteins/analysis , Antibodies, Bacterial/immunology , Endoplasmic Reticulum/chemistry , Fluorescent Antibody Technique, Indirect , HeLa Cells , Humans , Microscopy, Confocal , Microscopy, Fluorescence
SELECTION OF CITATIONS
SEARCH DETAIL
...