Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
1.
J Virol ; 96(22): e0155522, 2022 11 23.
Article in English | MEDLINE | ID: mdl-36317879

ABSTRACT

Porcine epidemic diarrhea virus (PEDV) is a re-emerging enteric coronavirus currently spreading in several nations and inflicting substantial financial damages on the swine industry. The currently available coronavirus vaccines do not provide adequate protection against the newly emerging viral strains. It is essential to study the relationship between host antiviral factors and the virus and to investigate the mechanisms underlying host immune response against PEDV infection. This study shows that heterogeneous nuclear ribonucleoprotein K (hnRNP K), the host protein determined by the transcription factor KLF15, inhibits the replication of PEDV by degrading the nucleocapsid (N) protein of PEDV in accordance with selective autophagy. hnRNP K was found to be capable of recruiting the E3 ubiquitin ligase, MARCH8, aiming to ubiquitinate N protein. Then, it was found that the ubiquitinated N protein could be delivered into autolysosomes for degradation by the cargo receptor NDP52, thereby inhibiting PEDV proliferation. Moreover, based on the enhanced MyD88 expression, we found that hnRNP K activated the interferon 1 (IFN-1) signaling pathway. Overall, the data obtained revealed a new mechanism of hnRNP K-mediated virus restriction wherein hnRNP K suppressed PEDV replication by degradation of viral N protein using the autophagic degradation pathway and by induction of IFN-1 production based on upregulation of MyD88 expression. IMPORTANCE The spread of the highly virulent PEDV in many countries is still leading to several epidemic and endemic outbreaks. To elucidate effective antiviral mechanisms, it is important to study the relationship between host antiviral factors and the virus and to investigate the mechanisms underlying host immune response against PEDV infection. In the work, we detected hnRNP K as a new host restriction factor which can hinder PEDV replication through degrading the nucleocapsid protein based on E3 ubiquitin ligase MARCH8 and the cargo receptor NDP52. In addition, via the upregulation of MyD88 expression, hnRNP K could also activate the interferon (IFN) signaling pathway. This study describes a previously unknown antiviral function of hnRNP K and offers a new vision toward host antiviral factors that regulate innate immune response as well as a protein degradation pathway against PEDV infection.


Subject(s)
Coronavirus Infections , Heterogeneous-Nuclear Ribonucleoprotein K , Interferon Type I , Porcine epidemic diarrhea virus , Virus Replication , Animals , Antiviral Agents , Chlorocebus aethiops , Coronavirus Infections/veterinary , Heterogeneous-Nuclear Ribonucleoprotein K/genetics , Interferons , Myeloid Differentiation Factor 88 , Nucleocapsid Proteins/physiology , Porcine epidemic diarrhea virus/physiology , Swine , Swine Diseases/virology , Ubiquitin-Protein Ligases , Vero Cells , Interferon Type I/immunology
2.
J Virol ; 95(9)2021 04 12.
Article in English | MEDLINE | ID: mdl-33568513

ABSTRACT

Negative-sense RNA viruses (NSVs) rely on prepackaged viral RNA-dependent RNA polymerases (RdRp) to replicate and transcribe their viral genomes. Their replication machinery consists of an RdRp bound to viral RNA which is wound around a nucleoprotein (NP) scaffold, forming a viral ribonucleoprotein complex. NSV NP is known to regulate transcription and replication of genomic RNA; however, its role in maintaining and protecting the viral genetic material is unknown. Here, we exploited host microRNA expression to target NP of influenza A virus and Sendai virus to ascertain how this would impact genomic levels and the host response to infection. We find that in addition to inducing a drastic decrease in genome replication, the antiviral host response in the absence of NP is dramatically enhanced. Additionally, our data show that insufficient levels of NP prevent the replication machinery of these NSVs to process full-length genomes, resulting in aberrant replication products which form pathogen-associated molecular patterns in the process. These dynamics facilitate immune recognition by cellular pattern recognition receptors leading to a strong host antiviral response. Moreover, we observe that the consequences of limiting NP levels are universal among NSVs, including Ebola virus, Lassa virus, and measles virus. Overall, these results provide new insights into viral genome replication of negative-sense RNA viruses and highlight novel avenues for developing effective antiviral strategies, adjuvants, and/or live-attenuated vaccines.IMPORTANCE Negative-sense RNA viruses comprise some of the most important known human pathogens, including influenza A virus, measles virus, and Ebola virus. These viruses possess RNA genomes that are unreadable to the host, as they require specific viral RNA-dependent RNA polymerases in conjunction with other viral proteins, such as nucleoprotein, to be replicated and transcribed. As this process generates a significant amount of pathogen-associated molecular patterns, this phylum of viruses can result in a robust induction of the intrinsic host cellular response. To circumvent these defenses, these viruses form tightly regulated ribonucleoprotein replication complexes in order to protect their genomes from detection and to prevent excessive aberrant replication. Here, we demonstrate the balance that negative-sense RNA viruses must achieve both to replicate efficiently and to avoid induction of the host defenses.


Subject(s)
Influenza A Virus, H1N1 Subtype/physiology , Influenza, Human/virology , Nucleocapsid Proteins/physiology , Respirovirus Infections/virology , Sendai virus/physiology , Virus Replication , A549 Cells , Animals , Chlorocebus aethiops , Dogs , HEK293 Cells , HeLa Cells , Humans , Madin Darby Canine Kidney Cells , Vero Cells , Viral Tropism
3.
Viruses ; 12(7)2020 07 17.
Article in English | MEDLINE | ID: mdl-32708976

ABSTRACT

Arenaviridae is a family of viruses harbouring important emerging pathogens belonging to the Bunyavirales order. Like in other segmented negative strand RNA viruses, the nucleoprotein (NP) is a major actor of the viral life cycle being both (i) the necessary co-factor of the polymerase present in the L protein, and (ii) the last line of defence of the viral genome (vRNA) by physically hiding its presence in the cytoplasm. The NP is also one of the major players interfering with the immune system. Several structural studies of NP have shown that it features two domains: a globular RNA binding domain (NP-core) in its N-terminal and an exonuclease domain (ExoN) in its C-terminal. Further studies have observed that significant conformational changes are necessary for RNA encapsidation. In this review we revisited the most recent structural and functional data available on Arenaviridae NP, compared to other Bunyavirales nucleoproteins and explored the structural and functional implications. We review the variety of structural motif extensions involved in NP-NP binding mode. We also evaluate the major functional implications of NP interactome and the role of ExoN, thus making the NP a target of choice for future vaccine and antiviral therapy.


Subject(s)
Arenaviridae/metabolism , Nucleocapsid Proteins/metabolism , Virus Assembly , Arenaviridae/physiology , Nucleocapsid Proteins/physiology , Protein Structure, Tertiary
4.
Arch Virol ; 165(10): 2279-2289, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32719955

ABSTRACT

In the early stage of virus infection, the pattern recognition receptor (PRR) signaling pathway of the host cell is activated to induce interferon production, activating interferon-stimulated genes (ISGs) that encode antiviral proteins that exert antiviral effects. Viperin is one of the innate antiviral proteins that exert broad-spectrum antiviral effects by various mechanisms. Porcine epidemic diarrhea virus (PEDV) is a coronavirus that causes huge losses to the pig industry. Research on early antiviral responses in the gastrointestinal tract is essential for developing strategies to prevent the spread of PEDV. In this study, we investigated the mechanisms of viperin in PEDV-infected IPEJ-C2 cells. Increased expression of interferon and viperin and decreased replication of PEDV with a clear reduction in the viral load were observed in PEDV-infected IPEC-J2 cells. Amino acids 1-50 of porcine viperin contain an endoplasmic reticulum signal sequence that allows viperin to be anchored to the endoplasmic reticulum and are necessary for its function in inhibiting PEDV proliferation. The interaction of the viperin S-adenosylmethionine domain with the N protein of PEDV was confirmed via confocal laser scanning microscopy and co-immunoprecipitation. This interaction might interfere with viral replication or assembly to reduce virus proliferation. Our results highlight a potential mechanism whereby viperin is able to inhibit PEDV replication and play an antiviral role in innate immunity.


Subject(s)
Antiviral Agents/metabolism , Host Microbial Interactions/physiology , Nucleocapsid Proteins/physiology , Porcine epidemic diarrhea virus/physiology , Animals , Cell Line , Coronavirus Infections/genetics , Coronavirus Infections/immunology , Coronavirus Infections/virology , Host Microbial Interactions/genetics , Host Microbial Interactions/immunology , Immunity, Innate , Interferons/biosynthesis , Mice , Mice, Inbred BALB C , Nucleocapsid Proteins/antagonists & inhibitors , Nucleocapsid Proteins/chemistry , Porcine epidemic diarrhea virus/immunology , Porcine epidemic diarrhea virus/pathogenicity , Protein Interaction Domains and Motifs , Proteins/chemistry , Proteins/genetics , Proteins/physiology , RNA Interference , Swine , Virus Replication
5.
J Virol ; 94(16)2020 07 30.
Article in English | MEDLINE | ID: mdl-32493824

ABSTRACT

Ebola virus (EBOV) inclusion bodies (IBs) are cytoplasmic sites of nucleocapsid formation and RNA replication, housing key steps in the virus life cycle that warrant further investigation. During infection, IBs display dynamic properties regarding their size and location. The contents of IBs also must transition prior to further viral maturation, assembly, and release, implying additional steps in IB function. Interestingly, the expression of the viral nucleoprotein (NP) alone is sufficient for the generation of IBs, indicating that it plays an important role in IB formation during infection. In addition to NP, other components of the nucleocapsid localize to IBs, including VP35, VP24, VP30, and the RNA polymerase L. We previously defined and solved the crystal structure of the C-terminal domain of NP (NP-Ct), but its role in virus replication remained unclear. Here, we show that NP-Ct is necessary for IB formation when NP is expressed alone. Interestingly, we find that NP-Ct is also required for the production of infectious virus-like particles (VLPs), and that defective VLPs with NP-Ct deletions are significantly reduced in viral RNA content. Furthermore, coexpression of the nucleocapsid component VP35 overcomes deletion of NP-Ct in triggering IB formation, demonstrating a functional interaction between the two proteins. Of all the EBOV proteins, only VP35 is able to overcome the defect in IB formation caused by the deletion of NP-Ct. This effect is mediated by a novel protein-protein interaction between VP35 and NP that controls both regulation of IB formation and RNA replication itself and that is mediated by a newly identified functional domain of NP, the central domain.IMPORTANCE Inclusion bodies (IBs) are cytoplasmic sites of RNA synthesis for a variety of negative-sense RNA viruses, including Ebola virus. In addition to housing important steps in the viral life cycle, IBs protect new viral RNA from innate immune attack and contain specific host proteins whose function is under study. A key viral factor in Ebola virus IB formation is the nucleoprotein, NP, which also is important in RNA encapsidation and synthesis. In this study, we have identified two domains of NP that control inclusion body formation. One of these, the central domain (CD), interacts with viral protein VP35 to control both inclusion body formation and RNA synthesis. The other is the NP C-terminal domain (NP-Ct), whose function has not previously been reported. These findings contribute to a model in which NP and its interactions with VP35 link the establishment of IBs to the synthesis of viral RNA.


Subject(s)
Ebolavirus/metabolism , Inclusion Bodies, Viral/metabolism , Nucleoproteins/physiology , Cell Line , Ebolavirus/pathogenicity , Genome, Viral/genetics , HEK293 Cells , Hemorrhagic Fever, Ebola/virology , Humans , Inclusion Bodies/metabolism , Nucleocapsid/metabolism , Nucleocapsid/physiology , Nucleocapsid Proteins/metabolism , Nucleocapsid Proteins/physiology , Nucleoproteins/metabolism , RNA/biosynthesis , RNA, Viral/genetics , Transcription Factors/metabolism , Viral Proteins/metabolism , Viral Regulatory and Accessory Proteins/metabolism , Viral Regulatory and Accessory Proteins/physiology , Virion/metabolism , Virus Replication/physiology
7.
J Virol ; 93(10)2019 05 15.
Article in English | MEDLINE | ID: mdl-30867297

ABSTRACT

Andes virus (ANDV) causes hantavirus pulmonary syndrome (HPS) and is the only hantavirus shown to spread person to person and cause a highly lethal HPS-like disease in Syrian hamsters. The unique ability of ANDV N protein to inhibit beta interferon (IFNß) induction may contribute to its virulence and spread. Here we analyzed IFNß regulation by ANDV N protein substituted with divergent residues from the nearly identical Maporal virus (MAPV) N protein. We found that MAPV N fails to inhibit IFNß signaling and that replacing ANDV residues 252 to 296 with a hypervariable domain (HVD) from MAPV N prevents IFNß regulation. In addition, changing ANDV residue S386 to the histidine present in MAPV N or the alanine present in other hantaviruses prevented ANDV N from regulating IFNß induction. In contrast, replacing serine with phosphoserine-mimetic aspartic acid (S386D) in ANDV N robustly inhibited interferon regulatory factor 3 (IRF3) phosphorylation and IFNß induction. Additionally, the MAPV N protein gained the ability to inhibit IRF3 phosphorylation and IFNß induction when ANDV HVD and H386D replaced MAPV residues. Mass spectroscopy analysis of N protein from ANDV-infected cells revealed that S386 is phosphorylated, newly classifying ANDV N as a phosphoprotein and phosphorylated S386 as a unique determinant of IFN regulation. In this context, the finding that the ANDV HVD is required for IFN regulation by S386 but dispensable for IFN regulation by D386 suggests a role for HVD in kinase recruitment and S386 phosphorylation. These findings delineate elements within the ANDV N protein that can be targeted to attenuate ANDV and suggest targeting cellular kinases as potential ANDV therapeutics.IMPORTANCE ANDV contains virulence determinants that uniquely permit it to spread person to person and cause highly lethal HPS in immunocompetent hamsters. We discovered that ANDV S386 and an ANDV-specific hypervariable domain permit ANDV N to inhibit IFN induction and that IFN regulation is directed by phosphomimetic S386D substitutions in ANDV N. In addition, MAPV N proteins containing D386 and ANDV HVD gained the ability to inhibit IFN induction. Validating these findings, mass spectroscopy analysis revealed that S386 of ANDV N protein is uniquely phosphorylated during ANDV infection. Collectively, these findings reveal new paradigms for ANDV N protein as a phosphoprotein and IFN pathway regulator and suggest new mechanisms for hantavirus regulation of cellular kinases and signaling pathways. Our findings define novel IFN-regulating virulence determinants of ANDV, identify residues that can be modified to attenuate ANDV for vaccine development, and suggest the potential for kinase inhibitors to therapeutically restrict ANDV replication.


Subject(s)
Interferon-beta/metabolism , Nucleocapsid Proteins/metabolism , Orthohantavirus/metabolism , Animals , Chlorocebus aethiops , Endothelial Cells/virology , HEK293 Cells , Orthohantavirus/pathogenicity , Hantavirus Infections/virology , Host-Pathogen Interactions , Humans , Interferon-beta/physiology , Interferons/metabolism , Interferons/physiology , Nucleocapsid Proteins/physiology , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Serine/metabolism , Signal Transduction , Vero Cells , Virulence , Virulence Factors/metabolism , Virus Replication
8.
J Zhejiang Univ Sci B ; 19(7): 570-580, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29971995

ABSTRACT

Porcine epidemic diarrhea virus (PEDV) is a highly infectious pathogen that can cause severe diseases in pigs and result in enormous economic losses in the worldwide swine industry. Previous studies revealed that PEDV exhibits an obvious capacity for modulating interferon (IFN) signaling or expression. The newly discovered type III IFN, which plays a crucial role in antiviral immunity, has strong antiviral activity against PEDV proliferation in IPEC-J2 cells. In this study, we aimed to investigate the effect of PEDV nucleocapsid (N) protein on type III IFN-λ. We found that the N proteins of ten PEDV strains isolated between 2013 and 2017 from different local farms shared high nucleotide identities, while the N protein of the CV777 vaccine strain formed a monophyletic branch in the phylogenetic tree. The N protein of the epidemic strain could antagonize type III IFN, but not type I or type II IFN expression induced by polyinosinic-polycytidylic acid (poly(I:C)) in IPEC-J2 cells. Subsequently, we demonstrated that the inhibition of poly(I:C)-induced IFN-λ3 production by PEDV N protein was dependent on the blocking of nuclear factor-κB (NF-κB) nuclear translocation. These findings might help increase understanding of the pathogenesis of PEDV and its mechanisms for evading the host immune response.


Subject(s)
Interferons/antagonists & inhibitors , Interleukins/antagonists & inhibitors , NF-kappa B/metabolism , Nucleocapsid Proteins/physiology , Porcine epidemic diarrhea virus/physiology , Porcine epidemic diarrhea virus/pathogenicity , Active Transport, Cell Nucleus , Animals , Coronavirus Infections/immunology , Coronavirus Infections/veterinary , Coronavirus Infections/virology , Coronavirus Nucleocapsid Proteins , Genes, Viral , Host-Pathogen Interactions/immunology , Interferons/biosynthesis , Interferons/genetics , Interleukins/biosynthesis , Interleukins/genetics , Nucleocapsid Proteins/genetics , Nucleocapsid Proteins/immunology , Porcine epidemic diarrhea virus/genetics , Promoter Regions, Genetic , Swine , Swine Diseases/immunology , Swine Diseases/virology
9.
J Virol ; 92(4)2018 02 15.
Article in English | MEDLINE | ID: mdl-29212928

ABSTRACT

Autographa californica multiple nucleopolyhedrovirus (AcMNPV) orf75 (ac75) is a highly conserved gene of unknown function. In this study, we constructed an ac75 knockout AcMNPV bacmid and investigated the role of ac75 in the baculovirus life cycle. The expression and distribution of the Ac75 protein were characterized, and its interaction with another viral protein was analyzed to further understand its function. Our data indicated that ac75 was required for the nuclear egress of nucleocapsids, intranuclear microvesicle formation, and subsequent budded virion (BV) formation, as well as occlusion-derived virion (ODV) envelopment and embedding of ODVs into polyhedra. Western blot analyses showed that two forms, of 18 and 15 kDa, of FLAG-tagged Ac75 protein were detected. Ac75 was associated with both nucleocapsid and envelope fractions of BVs but with only the nucleocapsid fraction of ODVs; the 18-kDa form was associated with only BVs, whereas the 15-kDa form was associated with both types of virion. Ac75 was localized predominantly in the intranuclear ring zone during infection and exhibited a nuclear rim distribution during the early phase of infection. A phase separation assay suggested that Ac75 was not an integral membrane protein. A coimmunoprecipitation assay revealed an interaction between Ac75 and the integral membrane protein Ac76, and bimolecular fluorescence complementation assays identified the sites of the interaction within the cytoplasm and at the nuclear membrane and ring zone in AcMNPV-infected cells. Our results have identified ac75 as a second gene that is required for both the nuclear egress of nucleocapsids and the formation of intranuclear microvesicles.IMPORTANCE During the baculovirus life cycle, the morphogenesis of both budded virions (BVs) and occlusion-derived virions (ODVs) is proposed to involve a budding process at the nuclear membrane, which occurs while nucleocapsids egress from the nucleus or when intranuclear microvesicles are produced. However, the exact mechanism of virion morphogenesis remains unknown. In this study, we identified ac75 as a second gene, in addition to ac93, that is essential for the nuclear egress of nucleocapsids, intranuclear microvesicle formation, and subsequent BV formation, as well as ODV envelopment and embedding of ODVs into polyhedra. Ac75 is not an integral membrane protein. However, it interacts with an integral membrane protein (Ac76) and is associated with the nuclear membrane. These data enhance our understanding of the commonalities between nuclear egress of nucleocapsids and intranuclear microvesicle formation and may help to reveal insights into the mechanism of baculovirus virion morphogenesis.


Subject(s)
Cell Nucleus/virology , Nucleocapsid Proteins/physiology , Nucleopolyhedroviruses/physiology , Virus Replication , Active Transport, Cell Nucleus , Animals , Cell Nucleus/ultrastructure , Gene Knockout Techniques , Microscopy, Electron , Nuclear Envelope/metabolism , Nucleocapsid Proteins/genetics , Sf9 Cells , Spodoptera
10.
Microb Pathog ; 107: 81-87, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28330747

ABSTRACT

Peste des petits ruminants virus (PPRV), belonging to paramyxoviruses, has six structure proteins (such as matrix protein (M), nucleocapsid proteins (N), fusion protein (F) and hemagglutinin protein (H)) and could cause high morbidity and mortality in sheep and goats. Although a vaccine strain of PPRV has been rescued and co-expression of M and N could yield PPRV-like particles, the roles of structure proteins in virion assembly and release have not been investigated in detail. In this study, plasmids carrying PPRV cDNA sequences encoding the N, M, H, and F proteins were expressed in Vero cells. The co-expression of all four proteins resulted in the release of virus-like particles (VLPs) with similar release efficiency to that of authentic virions. Moreover, the co-expression of M together with F also resulted in efficient VLPs release. In the absence of M protein, the expression of no combination of the other proteins resulted in particle release. In summary, a VLPs production system for PPRV has been established and M protein is necessary for promoting the assembly and release of VLPs, of which the predominant protein is M protein. Further study will be focused on the immunogenicity of the VLPs.


Subject(s)
Peste-des-petits-ruminants virus/metabolism , Peste-des-petits-ruminants virus/physiology , Vero Cells/metabolism , Viral Matrix Proteins/metabolism , Animals , Antibodies, Viral , Chlorocebus aethiops/metabolism , Chlorocebus aethiops/physiology , DNA, Complementary , DNA, Viral , Hemagglutinins, Viral/metabolism , Hemagglutinins, Viral/physiology , Mice , Nucleocapsid Proteins/metabolism , Nucleocapsid Proteins/physiology , Peste-des-petits-ruminants virus/genetics , Peste-des-petits-ruminants virus/immunology , Viral Fusion Proteins/metabolism , Viral Fusion Proteins/physiology
11.
J Clin Invest ; 126(3): 1012-22, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26878170

ABSTRACT

Paget's disease (PD) is characterized by focal and dramatic bone resorption and formation. Treatments that target osteoclasts (OCLs) block both pagetic bone resorption and formation; therefore, PD offers key insights into mechanisms that couple bone resorption and formation. Here, we evaluated OCLs from 3 patients with PD and determined that measles virus nucleocapsid protein (MVNP) was expressed in 70% of these OCLs. Moreover, transgenic mice with OCL-specific expression of MVNP (MVNP mice) developed PD-like bone lesions that required MVNP-dependent induction of high IL-6 expression levels in OCLs. In contrast, mice harboring a knockin of p62P394L (p62-KI mice), which is the most frequent PD-associated mutation, exhibited increased bone resorption, but not formation. Evaluation of OCLs from MVNP, p62-KI, and WT mice revealed increased IGF1 expression in MVNP-expressing OCLs that resulted from the high IL-6 expression levels in these cells. IL-6, in turn, increased the expression of coupling factors, specifically ephrinB2 on OCLs and EphB4 on osteoblasts (OBs). IGF1 enhanced ephrinB2 expression on OCLs and OB differentiation. Importantly, ephrinB2 and IGF1 levels were increased in MVNP-expressing OCLs from patients with PD and MVNP-transduced human OCLs compared with levels detected in controls. Further, anti-IGF1 or anti-IGF1R blocked Runx2 and osteocalcin upregulation in OBs cocultured with MVNP-expressing OCLs. These results suggest that in PD, MVNP upregulates IL-6 and IGF1 in OCLs to increase ephrinB2-EphB4 coupling and bone formation.


Subject(s)
Measles virus/physiology , Nucleocapsid Proteins/physiology , Osteitis Deformans/pathology , Osteoblasts/physiology , Animals , Case-Control Studies , Cell Differentiation , Cells, Cultured , Coculture Techniques , Ephrin-B2/metabolism , Humans , Insulin-Like Growth Factor I/metabolism , Interleukin-6/physiology , Mice, Knockout , Osteitis Deformans/virology , Osteoclasts/physiology , Receptor, EphB4/metabolism
12.
Biochem J ; 464(1): 109-21, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25062117

ABSTRACT

The hantaviral zoonotic diseases pose a significant threat to human health due to the lack of potential antiviral therapeutics or a vaccine against hantaviruses. N (Sin Nombre hantavirus nucleocapsid protein) augments mRNA translation. N binds to both the mRNA 5' cap and 40S ribosomal subunit via RPS19 (ribosomal protein S19). N with the assistance of the viral mRNA 5'-UTR preferentially favours the translation of a downstream ORF. We identified and characterized the RPS19-binding domain at the N-terminus of N. Its deletion did not influence the secondary structure, but affected the conformation of trimeric N molecules. The N variant lacking the RPS19-binding region was able to bind both the mRNA 5' cap and panhandle-like structure, formed by the termini of viral genomic RNA. In addition, the N variant formed stable trimers similar to wild-type N. Use of this variant in multiple experiments provided insights into the mechanism of ribosome loading during N-mediated translation strategy. The present study suggests that N molecules individually associated with the mRNA 5' cap and RPS19 of the 40S ribosomal subunit undergo N-N interaction to facilitate the engagement of N-associated ribosomes at the mRNA 5' cap. This has revealed new targets for therapeutic intervention of hantavirus infection.


Subject(s)
Nucleocapsid Proteins/physiology , Orthohantavirus/physiology , Peptide Chain Initiation, Translational/physiology , Ribosomal Proteins/metabolism , Amino Acid Sequence , Binding Sites/physiology , HeLa Cells , Humans , Molecular Sequence Data , Ribosomal Proteins/genetics
13.
Biochemistry ; 52(46): 8226-36, 2013 Nov 19.
Article in English | MEDLINE | ID: mdl-24144434

ABSTRACT

The human immunodeficiency virus type-1 (HIV-1) nucleocapsid (NC) protein is a chaperone that facilitates nucleic acid conformational changes to produce the most thermodynamically stable arrangement. The critical role of NC in many steps of the viral life cycle makes it an attractive therapeutic target. The chaperone activity of NC depends on its nucleic acid aggregating ability, duplex destabilizing activity, and rapid on-off binding kinetics. During the minus-strand transfer step of reverse transcription, NC chaperones the annealing of highly structured transactivation response region (TAR) RNA to the complementary TAR DNA. In this work, the role of different functional domains of NC in facilitating 59-nucleotide TAR RNA-DNA annealing was probed by using chemically synthesized peptides derived from full-length (55 amino acids) HIV-1 NC: NC(1-14), NC(15-35), NC(1-28), NC(1-35), NC(29-55), NC(36-55), and NC(11-55). Most of these peptides displayed significantly reduced annealing kinetics, even when present at concentrations much higher than that of wild-type (WT) NC. In addition, these truncated NC constructs generally bind more weakly to single-stranded DNA and are less effective nucleic acid aggregating agents than full-length NC, consistent with the loss of both electrostatic and hydrophobic contacts. However, NC(1-35) displayed annealing kinetics, nucleic acid binding, and aggregation activity that were very similar to those of WT NC. Thus, we conclude that the N-terminal zinc finger, flanked by the N-terminus and linker domains, represents the minimal sequence that is necessary and sufficient for chaperone function in vitro. In addition, covalent continuity of the 35 N-terminal amino acids of NC is critical for full activity. Thus, although the hydrophobic pocket formed by residues proximal to the C-terminal zinc finger has been a major focus of recent anti-NC therapeutic strategies, NC(1-35) represents an alternative target for therapeutics aimed at disrupting NC's chaperone function.


Subject(s)
HIV-1/genetics , Molecular Chaperones/metabolism , Nucleocapsid Proteins/physiology , Zinc Fingers/physiology , DNA, Viral/chemistry , DNA, Viral/metabolism , HIV Long Terminal Repeat/physiology , Molecular Chaperones/chemistry , Nucleocapsid Proteins/chemistry , Protein Structure, Tertiary , RNA, Viral/chemistry , RNA, Viral/metabolism , Zinc Fingers/genetics
14.
J Virol ; 87(17): 9486-500, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23637410

ABSTRACT

The coronavirus nucleocapsid (N) protein plays a multifunctional role in the virus life cycle, from regulation of replication and transcription and genome packaging to modulation of host cell processes. These functions are likely to be facilitated by interactions with host cell proteins. The potential interactome of the infectious bronchitis virus (IBV) N protein was mapped using stable isotope labeling with amino acids in cell culture (SILAC) coupled to a green fluorescent protein-nanotrap pulldown methodology and liquid chromatography-tandem mass spectrometry. The addition of the SILAC label allowed discrimination of proteins that were likely to specifically bind to the N protein over background binding. Overall, 142 cellular proteins were selected as potentially binding to the N protein, many as part of larger possible complexes. These included ribosomal proteins, nucleolar proteins, translation initiation factors, helicases, and hnRNPs. The association of selected cellular proteins with IBV N protein was confirmed by immunoblotting, cosedimentation, and confocal microscopy. Further, the localization of selected proteins in IBV-infected cells as well as their activity during virus infection was assessed by small interfering RNA-mediated depletion, demonstrating the functional importance of cellular proteins in the biology of IBV. This interactome not only confirms previous observations made with other coronavirus and IBV N proteins with both overexpressed proteins and infectious virus but also provides novel data that can be exploited to understand the interaction between the virus and the host cell.


Subject(s)
Host-Pathogen Interactions/physiology , Infectious bronchitis virus/physiology , Nucleocapsid Proteins/physiology , Animals , Chlorocebus aethiops , Coronavirus Nucleocapsid Proteins , Gene Knockdown Techniques , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Host-Pathogen Interactions/genetics , Humans , Infectious bronchitis virus/genetics , Nucleocapsid Proteins/genetics , Protein Binding , Proteome/genetics , Proteome/metabolism , RNA, Small Interfering/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Ribosomes/metabolism , Vero Cells
15.
Proc Natl Acad Sci U S A ; 110(10): E861-8, 2013 Mar 05.
Article in English | MEDLINE | ID: mdl-23404707

ABSTRACT

We present an unconventional approach to antiviral drug discovery, which is used to identify potent small molecules against rabies virus. First, we conceptualized viral capsid assembly as occurring via a host-catalyzed biochemical pathway, in contrast to the classical view of capsid formation by self-assembly. This suggested opportunities for antiviral intervention by targeting previously unappreciated catalytic host proteins, which were pursued. Second, we hypothesized these host proteins to be components of heterogeneous, labile, and dynamic multi-subunit assembly machines, not easily isolated by specific target protein-focused methods. This suggested the need to identify active compounds before knowing the precise protein target. A cell-free translation-based small molecule screen was established to recreate the hypothesized interactions involving newly synthesized capsid proteins as host assembly machine substrates. Hits from the screen were validated by efficacy against infectious rabies virus in mammalian cell culture. Used as affinity ligands, advanced analogs were shown to bind a set of proteins that effectively reconstituted drug sensitivity in the cell-free screen and included a small but discrete subfraction of cellular ATP-binding cassette family E1 (ABCE1), a host protein previously found essential for HIV capsid formation. Taken together, these studies advance an alternate view of capsid formation (as a host-catalyzed biochemical pathway), a different paradigm for drug discovery (whole pathway screening without knowledge of the target), and suggest the existence of labile assembly machines that can be rendered accessible as next-generation drug targets by the means described.


Subject(s)
Antiviral Agents/pharmacology , Host-Pathogen Interactions/drug effects , Rabies virus/drug effects , Rabies virus/physiology , Viral Proteins/physiology , Amino Acid Sequence , Animals , Cell-Free System , Chlorocebus aethiops , Drug Discovery , Host-Pathogen Interactions/physiology , Humans , Microbial Sensitivity Tests , Molecular Sequence Data , Nucleocapsid Proteins/chemistry , Nucleocapsid Proteins/genetics , Nucleocapsid Proteins/physiology , Protein Interaction Domains and Motifs , Rabies virus/genetics , Vero Cells , Viral Proteins/chemistry , Viral Proteins/genetics , Virus Assembly/drug effects
16.
Bone ; 53(1): 269-76, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23262029

ABSTRACT

Measles virus plays an important role as an environmental factor in the pathogenesis of Paget's disease (PD). Previous studies have shown that IL-6 is increased in the bone marrow of Paget's patients and that measles virus nucleocapsid protein (MVNP) induces IL-6 secretion by pagetic osteoclasts. Further, IL-6 plays a critical role in the development of pagetic osteoclasts and bone lesions induced by PD, but the mechanisms regulating IL-6 production by MVNP remain unclear. Our current studies revealed that MVNP expression in osteoclast precursors down-regulated Sirt1 mRNA and protein, a negative regulator of NF-κB activity, which is a key factor for IL-6 expression. MVNP expression in NIH3T3 cells also elevated Il-6 transcription and impaired the expression of Sirt1 mRNA both under basal conditions and upon activation of the Sirt1 upstream regulator FoxO3 by LY294002 (a PI3K/AKT inhibitor). Luciferase activity assays showed that constitutively active FoxO3 abolished the repressive effect of MVNP on reporters driven by either FoxO3 response elements or the Sirt1 promoter. Further, protein stability assays revealed that FoxO3 was degraded more rapidly in MVNP-expressing cells than in control cells following the addition of cycloheximide. Similarly, co-transfection of MVNP and FoxO3 into HEK293 cells demonstrated that MVNP decreased the protein levels of over-expressed FoxO3 in a dose-dependent manner. Treatment with the proteasome inhibitor, MG132, blocked the MVNP-triggered decrease of FoxO3, and the treatment with the serine/threonine phosphatase inhibitor, calyculin A, revealed that MVNP increased phosphorylation of FoxO3. Further, over-expression of Sirt1 or treatment with the Sirt1 activator resveratrol blocked the increase in Il-6 transcription by MVNP. Finally, resveratrol reduced the numbers of TRAP positive multi-nuclear cells in bone marrow cultures from TRAP-MVNP transgenic mice to wild type levels. These results indicate that MVNP decreases FoxO3/Sirt1 signaling to enhance the levels of IL-6, which in part mediate MVNP's contribution to the development of Paget's disease.


Subject(s)
Down-Regulation , Forkhead Transcription Factors/metabolism , Interleukin-6/metabolism , Measles virus/physiology , Nucleocapsid Proteins/physiology , Osteitis Deformans/metabolism , Signal Transduction , Sirtuin 1/metabolism , Animals , Base Sequence , Blotting, Western , DNA Primers , Forkhead Box Protein O3 , Forkhead Transcription Factors/physiology , Mice , NIH 3T3 Cells , Reverse Transcriptase Polymerase Chain Reaction , Sirtuin 1/genetics , Transcription, Genetic/physiology
17.
PLoS One ; 7(12): e51534, 2012.
Article in English | MEDLINE | ID: mdl-23236513

ABSTRACT

Retroviral nucleocapsid proteins harbor nucleic acid chaperoning activities that mostly rely on the N-terminal basic residues and the CCHC zinc finger motif. Such chaperoning is essential for virus replication, notably for genomic RNA selection and packaging in virions, and for reverse transcription of genomic RNA into DNA. Recent data revealed that HIV-1 nucleocapsid restricts reverse transcription during virus assembly--a process called late reverse transcription--suggesting a regulation between RNA packaging and late reverse transcription. Indeed, mutating the HIV-1 nucleocapsid basic residues or the two zinc fingers caused a reduction in RNA incorporated and an increase in newly made viral DNA in the mutant virions. MoMuLV nucleocapsid has an N-terminal basic region similar to HIV-1 nucleocapsid but a unique zinc finger. This prompted us to investigate whether the N-terminal basic residues and the zinc finger of MoMuLV and HIV-1 nucleocapsids play a similar role in genomic RNA packaging and late reverse transcription. To this end, we analyzed the genomic RNA and viral DNA contents of virions produced by cells transfected with MoMuLV molecular clones where the zinc finger was mutated or completely deleted or with a deletion of the N-terminal basic residues of nucleocapsid. All mutant virions showed a strong defect in genomic RNA content indicating that the basic residues and zinc finger are important for genomic RNA packaging. In contrast to HIV-1 nucleocapsid-mutants, the level of viral DNA in mutant MoMuLV virions was only slightly increased. These results confirm that the N-terminal basic residues and zinc finger of MoMuLV nucleocapsid are critical for genomic RNA packaging but, in contrast to HIV-1 nucleocapsid, they most probably do not play a role in the control of late reverse transcription. In addition, these results suggest that virus formation and late reverse transcription proceed according to distinct mechanisms for MuLV and HIV-1.


Subject(s)
HIV-1/physiology , Leukemia Virus, Murine/physiology , Nucleocapsid Proteins/physiology , RNA, Viral/physiology , Reverse Transcription/physiology , Virus Assembly/physiology , Amino Acid Sequence , HEK293 Cells , HIV-1/genetics , Humans , Leukemia Virus, Murine/genetics , Molecular Sequence Data , Mutagenesis , Nucleocapsid Proteins/genetics , Oligonucleotides/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Reverse Transcription/genetics , Zinc Fingers/genetics , Zinc Fingers/physiology
18.
Virus Res ; 155(1): 168-74, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20875468

ABSTRACT

We previously reported that nucleoprotein (N) is related to the different pathogenicities of the virulent rabies virus strain Nishigahara (Ni) and avirulent strain Ni-CE and also that Ni N, but not Ni-CE N, functions to evade retinoic acid-inducible gene I (RIG-I)-mediated innate immunity. There are three amino acid differences between Ni and Ni-CE N (at positions 273, 394 and 395), indicating that one of these mutations or a combination of mutations is important for the pathogenicity and evasion of innate immunity. We generated Ni-CE mutants in which the amino acids in Ni-CE N were replaced with those of Ni in all combinations. Among the mutants, CE(NiN273/394) with mutations at positions 273 and 394 evaded activation of RIG-I-mediated signaling most efficiently and also showed the highest pathogenicity. This correlation reinforces the relation between evasion of host RIG-I-mediated innate immunity and pathogenicity of rabies virus.


Subject(s)
DEAD-box RNA Helicases/immunology , Immune Evasion , Nucleocapsid Proteins/physiology , Rabies virus/pathogenicity , Virulence Factors/physiology , Amino Acid Substitution/genetics , Amino Acids/genetics , Animals , Cell Line , Cricetinae , DEAD Box Protein 58 , Disease Models, Animal , Female , Humans , Lethal Dose 50 , Mice , Mutagenesis, Site-Directed , Nucleocapsid Proteins/genetics , Rabies/pathology , Rabies/virology , Rabies virus/genetics , Survival Analysis , Virulence , Virulence Factors/genetics
19.
J Virol ; 84(19): 9793-9, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20668089

ABSTRACT

The interaction of Nipah virus (NiV) nucleocapsid (N) protein with phosphoprotein (P) during nucleocapsid assembly is the essential process in the viral life cycle, since only the encapsidated RNA genome can be used for replication. To identify the region responsible for N-P interaction, we utilized fluorescent protein tags to visualize NiV N and P proteins in live cells and analyzed their cellular localization. N protein fused to monomeric enhanced cyan fluorescence protein (N-ECFP) exhibited a dotted pattern in transfected cells, while P protein fused to monomeric red fluorescent protein (P-mRFP) showed diffuse distribution. When the two proteins were coexpressed, P-mRFP colocalized with N-ECFP dots. N-ECFP mutants with serial amino acid deletions were generated to search for the region(s) responsible for this N-P colocalization. We found that, in addition to the 467- to 496-amino-acid (aa) region reported previously, aa 135 to 146 were responsible for the N-P colocalization. The residues crucial for N-P interaction were further investigated by introducing alanine substitutions into the untagged N protein. Alanine scanning in the region of aa 135 to 146 has revealed that there are distinct regions essential for the interaction of N-P and the function of N. This is the first study to visualize Nipah viral proteins in live cells and to assess the essential domain of N protein for the interaction with P protein.


Subject(s)
Nipah Virus/physiology , Nucleocapsid Proteins/physiology , Amino Acid Sequence , Amino Acid Substitution , Animals , Binding Sites/genetics , COS Cells , Cell Line , Chlorocebus aethiops , Cricetinae , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Molecular Sequence Data , Mutagenesis, Site-Directed , Nipah Virus/genetics , Nucleocapsid Proteins/chemistry , Nucleocapsid Proteins/genetics , Phosphoproteins/physiology , Protein Interaction Domains and Motifs , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transfection , Vero Cells , Virus Replication/physiology , Red Fluorescent Protein
20.
J Virol ; 84(19): 10276-88, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20660183

ABSTRACT

The coronavirus nucleocapsid (N) protein plays an essential role in virion assembly via interactions with the large, positive-strand RNA viral genome and the carboxy-terminal endodomain of the membrane protein (M). To learn about the functions of N protein domains in the coronavirus mouse hepatitis virus (MHV), we replaced the MHV N gene with its counterpart from the closely related bovine coronavirus (BCoV). The resulting viral mutant was severely defective, even though individual domains of the N protein responsible for N-RNA, N-M, or N-N interactions were completely interchangeable between BCoV and MHV. The lesion in the BCoV N substitution mutant could be compensated for by reverting mutations in the central, serine- and arginine-rich (SR) domain of the N protein. Surprisingly, a second class of reverting mutations were mapped to the amino terminus of a replicase subunit, nonstructural protein 3 (nsp3). A similarly defective MHV N mutant bearing an insertion of the SR region from the severe acute respiratory syndrome coronavirus N protein was rescued by the same two classes of reverting mutations. Our genetic results were corroborated by the demonstration that the expressed amino-terminal segment of nsp3 bound selectively to N protein from infected cells, and this interaction was RNA independent. Moreover, we found a direct correlation between the N-nsp3 interaction and the ability of N protein to stimulate the infectivity of transfected MHV genomic RNA (gRNA). Our results suggest a role for this previously unknown N-nsp3 interaction in the localization of genomic RNA to the replicase complex at an early stage of infection.


Subject(s)
Coronavirus, Bovine/physiology , DNA-Directed RNA Polymerases/physiology , Murine hepatitis virus/physiology , Nucleocapsid Proteins/physiology , RNA, Viral/genetics , RNA-Dependent RNA Polymerase/physiology , Amino Acid Sequence , Animals , Cattle , Coronavirus Nucleocapsid Proteins , Coronavirus, Bovine/genetics , Coronavirus, Bovine/pathogenicity , DNA-Directed RNA Polymerases/genetics , Genome, Viral , Humans , Mice , Molecular Sequence Data , Murine hepatitis virus/genetics , Murine hepatitis virus/pathogenicity , Mutation , Nucleocapsid Proteins/chemistry , Nucleocapsid Proteins/genetics , Protein Structure, Tertiary , RNA-Dependent RNA Polymerase/genetics , Recombination, Genetic , Severe acute respiratory syndrome-related coronavirus/genetics , Severe acute respiratory syndrome-related coronavirus/pathogenicity , Severe acute respiratory syndrome-related coronavirus/physiology , Sequence Homology, Amino Acid , Transfection , Virulence/genetics , Virulence/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...