Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
Add more filters










Publication year range
1.
J Virol ; 96(24): e0121022, 2022 12 21.
Article in English | MEDLINE | ID: mdl-36448796

ABSTRACT

All lentiviruses encode a post-transcriptional transactivator, Rev, which mediates the export of viral mRNA from the nucleus to the cytoplasm and which is required for viral gene expression and viral replication. In the current study, we demonstrate that equine infectious anemia virus (EIAV), an equine lentivirus, encodes a second post-transcriptional transactivator that we designate Grev. Grev is encoded by a novel transcript with a single splicing event that was identified using reverse transcription-PCR (RT-PCR) and RNA-seq in EIAV-infected horse tissues and cells. Grev is about 18 kDa in size, comprises the first 18 amino acids (aa) of Gag protein together with the last 82 aa of Rev, and was detected in EIAV-infected cells. Similar to Rev, Grev is localized to the nucleus, and both are able to mediate the expression of Mat (a recently identified viral protein of unknown function from EIAV), but Rev can mediate the expression of EIAV Gag/Pol, while Grev cannot. We also demonstrate that Grev, similar to Rev, specifically binds to rev-responsive element 2 (RRE-2, located in the first exon of mat mRNAs) to promote nuclear export of mat mRNA via the chromosome region maintenance 1 (CRM1) pathway. However, unlike Rev, whose function depends on its multimerization, we could not detect multimerization of Grev using coimmunoprecipitation (co-IP) or bimolecular fluorescence complementation (BiFC) assays. Together, these data suggest that EIAV encodes two post-transcriptional transactivators, Rev and Grev, with similar, but not identical, functions. IMPORTANCE Nuclear export of viral transcripts is a crucial step for viral gene expression and viral replication in lentiviruses, and this export is regulated by a post-transcriptional transactivator, Rev, that is shared by all lentiviruses. Here, we report that the equine infectious anemia virus (EIAV) encodes a novel viral protein, Grev, and demonstrated that Grev, like Rev, mediates the expression of the viral protein Mat by binding to the first exon of mat mRNAs via the chromosome region maintenance 1 (CRM1) pathway. Grev is encoded by a single-spliced transcript containing two exons, whereas Rev is encoded by a multiple-spliced transcript containing four exons. Moreover, Rev is able to mediate EIAV Gag/Pol expression by binding to rev-responsive element (RRE) located within the Env-coding region, while Grev cannot. Therefore, the present study demonstrates that EIAV encodes two post-transcriptional regulators, Grev and Rev, suggesting that post-transcriptional regulation patterns in lentivirus are diverse and complex.


Subject(s)
Equine Infectious Anemia , Infectious Anemia Virus, Equine , Trans-Activators , Animals , Equine Infectious Anemia/virology , Exons , Gene Products, rev/genetics , Horses/genetics , Infectious Anemia Virus, Equine/genetics , Infectious Anemia Virus, Equine/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Gene Expression Regulation, Viral/genetics
2.
J Virol ; 96(20): e0054922, 2022 10 26.
Article in English | MEDLINE | ID: mdl-36197105

ABSTRACT

Equine infectious anemia virus (EIAV) and HIV are both members of the Lentivirus genus and are similar in major virological characters. EIAV endangers the horse industry. In addition, EIAV can also be used as a model for HIV research. The maturation of the lentiviral Env protein, which is necessary for viral entry, requires Env to be folded in the endoplasmic reticulum (ER). It is currently unclear how this process is regulated. Mitochondrion-associated endoplasmic reticulum membrane (MAM) is a specialized part of the close connection between the ER and mitochondria, and one of the main functions of MAM is to promote oxidative protein production in the ER. SYNJ2BP is one of the key proteins that make up the MAM, and we found that SYNJ2BP is essential for EIAV replication. We therefore constructed a SYNJ2BP knockout HEK293T cell line in which the number of MAMs is significantly reduced. Moreover, overexpression of SYNJ2BP could increase the number of MAMs. Our study demonstrates that SYNJ2BP can improve the infectivity of the EIAV virus with elevated production of the viral Env protein through increased MAM formation. Interestingly, SYNJ2BP was able to improve the production of not only EIAV Env but also HIV. Further investigation showed that MAMs can provide more ATP and calcium ions, which are essential factors for Env production, to the ER and can also reduce ER stress induced by HIV or EIAV Envs to increase the Env production level in cells. These results may help us to understand the key production mechanisms of lentiviral Env. IMPORTANCE Lentiviral Env proteins, which are rich in disulfide bonds, need to be fully folded in the ER; otherwise, misfolded Env proteins will induce ER stress and be degraded by ER-associated protein degradation (ERAD). To date, it is still unclear about Env production mechanism in the ER. MAM is the structure of closely connection between the ER and mitochondria. MAMs play important roles in the calcium steady state and oxidative stress, especially in the production of oxidative protein. For the first time, we found that SYNJ2BP can promote the production of lentiviral Env proteins by providing the ATP and calcium ions required for oxidative protein production in the ER and by reducing ER stress through facilitating formation of MAMs. These studies shed light on how MAMs improve lentiviral Env production, which will lay the foundation for the study of replication mechanisms in other lentiviruses from the perspective of the cellular organelle microenvironment.


Subject(s)
HIV Infections , Infectious Anemia Virus, Equine , Horses , Humans , Animals , Gene Products, env/metabolism , Calcium/metabolism , HEK293 Cells , Infectious Anemia Virus, Equine/genetics , Infectious Anemia Virus, Equine/metabolism , Endoplasmic Reticulum/metabolism , Mitochondria/metabolism , HIV Infections/metabolism , Adenosine Triphosphate/metabolism , Disulfides/metabolism , Membrane Proteins/metabolism
3.
J Virol ; 96(18): e0098622, 2022 09 28.
Article in English | MEDLINE | ID: mdl-36069548

ABSTRACT

All lentiviruses encode the accessory protein Rev, whose main biological function is to mediate the nuclear export of unspliced and incompletely spliced viral transcripts by binding to a viral cis-acting element (termed the Rev-responsive element, RRE) within the env-encoding region. Equine infectious anemia virus (EIAV) is a member of the lentivirus genus in the Retroviridae family and is considered an important model for the study of lentivirus pathogenesis. Here, we identified a novel transcript from the EIAV genome that encoded a viral protein, named Mat, with an unknown function. The transcript mat was fully spliced and comprised parts of the coding regions of MA and TM. Interestingly, the expression of Mat depended on Rev and the chromosome region maintenance 1 (CRM1) pathway. Rev could specifically bind to Mat mRNA to promote its nuclear export. We further identified that the first exon of Mat mRNA, which was located within the Gag-encoding region, acted as an unreported RRE. Altogether, we identified a novel fully spliced transcript mat with an unusual RRE, which interacted with Rev for nuclear export through the CRM1 pathway. These findings updated the EIAV genome structure, highlighted the diversification of posttranscriptional regulation patterns in EIAV, and may help to expand the understanding of gene transcription and expression of lentivirus. IMPORTANCE In lentiviruses, the nuclear export of viral transcripts is an important step in controlling viral gene expression. Generally, the unspliced and incompletely spliced transcripts are exported via the CRM1-dependent export pathway in a process mediated by the viral Rev protein by binding to the Rev-responsive element (RRE) located within the Env-coding region. However, the completely spliced transcripts are exported via an endogenous cellular pathway, which was Rev independent. Here, we identified a novel fully spliced transcript from EIAV and demonstrated that it encoded a viral protein, termed Mat. Interestingly, we determined that the expression of Mat depended on Rev and identified that the first exon of Mat mRNA could specifically bind to Rev and be exported to the cytoplasm, which suggested that the first exon of Mat mRNA was a second RRE of EIAV. These findings provided important insights into the Rev-dependent nuclear export of completely spliced transcripts in lentiviruses.


Subject(s)
Gene Products, rev , Infectious Anemia Virus, Equine , Lentiviruses, Equine , Animals , Gene Products, rev/genetics , Horses , Infectious Anemia Virus, Equine/metabolism , RNA Splicing , RNA, Messenger/genetics , RNA, Viral/genetics
4.
PLoS Pathog ; 18(2): e1009986, 2022 02.
Article in English | MEDLINE | ID: mdl-35139135

ABSTRACT

The Nrf2/Keap1 axis plays a complex role in viral susceptibility, virus-associated inflammation and immune regulation in host cells. However, whether or how the Nrf2/Keap1 axis is involved in the interactions between equine lentiviruses and their hosts remains unclear. Here, we demonstrate that the Nrf2/Keap1 axis was activated during EIAV infection. Mechanistically, EIAV-Rev competitively binds to Keap1 and releases Nrf2 from Keap1-mediated repression, leading to the accumulation of Nrf2 in the nucleus and promoting Nrf2 responsive genes transcription. Subsequently, we demonstrated that the Nrf2/Keap1 axis represses EIAV replication via two independent molecular mechanisms: directly increasing antioxidant enzymes to promote effective cellular resistance against EIAV infection, and repression of Rev-mediated RNA transport through direct interaction between Keap1 and Rev. Together, these data suggest that activation of the Nrf2/Keap1 axis mediates a passive defensive response to combat EIAV infection. The Nrf2/Keap1 axis could be a potential target for developing strategies for combating EIAV infection.


Subject(s)
Antiviral Agents/pharmacology , Gene Products, rev/metabolism , Infectious Anemia Virus, Equine/metabolism , Kelch-Like ECH-Associated Protein 1/metabolism , Antioxidants/metabolism , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Humans , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Signal Transduction/drug effects
5.
J Virol ; 95(23): e0108721, 2021 11 09.
Article in English | MEDLINE | ID: mdl-34495693

ABSTRACT

Envelope glycoproteins (Envs) of lentiviruses harbor unusually long cytoplasmic tails (CTs). Natural CT truncations always occur in vitro and are accompanied by attenuated virulence, but their effects on viral replication have not been fully elucidated. The Env in equine infectious anemia virus (EIAV) harbors the longest CT in the lentiviral family, and a truncated CT was observed in a live attenuated vaccine. This study demonstrates that CT truncation significantly increased EIAV production, as determined by comparing the virion yields from EIAV infectious clones in the presence and absence of the CT. A significant increase in a cleaved product from the CT-truncated Env precursor, but not the full-length Env, was observed. We further confirmed that the presence of the CT inhibited the cleavage of the Env precursor and found that a functional domain located at the C terminus was responsible for this function. Moreover, CT-truncated Env was mainly localized at the plasma membrane (PM), while full-length Env was mainly localized in the cytoplasm. The CT truncation caused a dramatic reduction in the endocytosis of Env. These results suggest that the CT can modulate the processing and trafficking of EIAV Env and thus regulate EIAV replication. IMPORTANCE The mature lentivirus envelope glycoprotein (Env) is composed of a surface unit (SU) and a transmembrane unit (TM), which are cleaved products of the Env precursor. After mature Env is heterodimerically formed from the cleavage of the Env precursor, it is trafficked to the plasma membrane (PM) for incorporation and virion assembly. Env harbors a long cytoplasmic tail (CT), which has been increasingly found to play multiple roles in the Env biological cycle. Here, we revealed for the first time that the CT of equine infectious anemia virus (EIAV) Env inhibits cleavage of the Env precursor. Simultaneously, the CT promoted Env endocytosis, resulting in weakened Env localization at the PM. We also validated that the CT could significantly decrease EIAV production. These findings suggest that the CT regulates the processing and trafficking of EIAV Env to balance virion production.


Subject(s)
Cell Membrane/metabolism , Equine Infectious Anemia/virology , Genes, env/genetics , Infectious Anemia Virus, Equine/metabolism , Virion/metabolism , Animals , Endocytosis , Genome, Viral , HEK293 Cells , HIV-1 , Horses , Humans , Infectious Anemia Virus, Equine/genetics , Vaccines, Attenuated , Viral Envelope Proteins/genetics , Virion/genetics , Virus Replication
6.
J Virol ; 94(7)2020 03 17.
Article in English | MEDLINE | ID: mdl-31941773

ABSTRACT

The cellular protein SERINC5 inhibits the infectivity of diverse retroviruses, and its activity is counteracted by the glycosylated Gag (glycoGag) protein of murine leukemia virus (MLV), the S2 protein of equine infectious anemia virus (EIAV), and the Nef protein of human immunodeficiency virus type 1 (HIV-1). Determining the regions within SERINC5 that provide restrictive activity or Nef sensitivity should inform mechanistic models of the SERINC5/HIV-1 relationship. Here, we report that deletion of the conserved sequence EDTEE, which is located within a cytoplasmic loop of SERINC5 and which is reminiscent of an acidic-cluster membrane trafficking signal, increases the sensitivity of SERINC5 to antagonism by Nef, while it has no effect on the intrinsic activity of the protein as an inhibitor of infectivity. These effects correlated with enhanced removal of the ΔEDTEE mutant relative to that of wild-type SERINC5 from the cell surface and with enhanced exclusion of the mutant protein from virions by Nef. Mutational analysis indicated that the acidic residues, but not the threonine, within the EDTEE motif are important for the relative resistance to Nef. Deletion of the EDTEE sequence did not increase the sensitivity of SERINC5 to antagonism by the glycoGag protein of MLV, suggesting that its virologic role is Nef specific. These results are consistent with the reported mapping of the cytoplasmic loop that contains the EDTEE sequence as a general determinant of Nef responsiveness, but they further indicate that sequences inhibitory to as well as supportive of Nef activity reside in this region. We speculate that the EDTEE motif might have evolved to mediate resistance against retroviruses that use Nef-like proteins to antagonize SERINC5.IMPORTANCE Cellular membrane proteins in the SERINC family, especially SERINC5, inhibit the infectivity of retroviral virions. This inhibition is counteracted by retroviral proteins, specifically, HIV-1 Nef, MLV glycoGag, and EIAV S2. One consequence of such a host-pathogen "arms race" is a compensatory change in the host antiviral protein as it evolves to escape the effects of viral antagonists. This is often reflected in a genetic signature, positive selection, which is conspicuously missing in SERINC5 Here we show that despite this lack of genetic evidence, a sequence in SERINC5 nonetheless provides relative resistance to antagonism by HIV-1 Nef.


Subject(s)
Membrane Proteins/chemistry , nef Gene Products, Human Immunodeficiency Virus/metabolism , Alleles , Amino Acid Motifs , Cytoplasm/metabolism , Gene Deletion , Glycosylation , HEK293 Cells , HIV-1 , HeLa Cells , Humans , Infectious Anemia Virus, Equine/metabolism , Jurkat Cells , Moloney murine leukemia virus/metabolism , Mutation , Protein Domains , nef Gene Products, Human Immunodeficiency Virus/genetics
7.
Virology ; 523: 52-63, 2018 10.
Article in English | MEDLINE | ID: mdl-30081309

ABSTRACT

Human myxovirus resistance protein B (hMXB) is a restriction factor of HIV-1 that also inhibits a variety of retroviruses. However, hMXB is not antiviral against equine infectious anemia virus (EIAV). We show here that equine MX2 (eMX2) potently restricts EIAV in vitro. Additionally, eMX2 inhibits HIV-1 and other lentiviruses, including murine leukemia virus. Previously, it was reported that hMXB repression is reduced in hMXB Δ1-25, but not in GTP-binding mutant K131A and GTP-hydrolysis mutant T151A. In contrast to this phenomenon, our study indicates that eMX2 restriction is not diminished in eMX2 Δ1-25, but is in eMX2 K127A and T147A, which correspond to hMXB K131A and T151A, respectively. Thus, eMX2 may inhibit retroviral replication by a novel mechanism that differs from that of hMXB.


Subject(s)
HIV-1/genetics , Host-Pathogen Interactions , Infectious Anemia Virus, Equine/genetics , Myxovirus Resistance Proteins/genetics , Amino Acid Substitution , Animals , Base Sequence , Binding Sites , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Guanosine Triphosphate/metabolism , HIV-1/growth & development , HIV-1/metabolism , Horses , Humans , Infectious Anemia Virus, Equine/growth & development , Infectious Anemia Virus, Equine/metabolism , Lentivirus/genetics , Lentivirus/metabolism , Mutation , Myxovirus Resistance Proteins/metabolism , Protein Binding , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction
8.
J Virol ; 92(8)2018 04 15.
Article in English | MEDLINE | ID: mdl-29386282

ABSTRACT

The equine infectious anemia virus (EIAV) attenuated vaccine was developed by long-term passaging of a field-isolated virulent strain in cross-species hosts, followed by successive cultivation in cells in vitro To explore the molecular mechanism underlying the evolution of the EIAV attenuated vaccine, a systematic study focusing on long-terminal-repeat (LTR) variation in numerous virus strains ranging from virulent EIAV to attenuated EIAV was performed over time both in vitro and in vivo Two hypervariable regions were identified within the U3 region in the enhancer region (EHR) and the negative regulatory element (NRE) and within the R region in the transcription start site (TSS) and the Tat-activating region (TAR). Among these sites, variation in the U3 region resulted in the formation of additional transcription factor binding sites; this variation of the in vitro-adapted strains was consistent with the loss of pathogenicity. Notably, the same LTR variation pattern was observed both in vitro and in vivo Generally, the LTR variation in both the attenuated virus and the virulent strain fluctuated over time in vivo Interestingly, the attenuated-virus-specific LTR variation was also detected in horses infected with the virulent strain, supporting the hypothesis that the evolution of an attenuated virus might have involved branching from EIAV quasispecies. This hypothesis was verified by phylogenetic analysis. The present systematic study examining the molecular evolution of attenuated EIAV from EIAV quasispecies may provide an informative model reflecting the evolution of similar lentiviruses.IMPORTANCE The attenuated EIAV vaccine was the first lentiviral vaccine used to successfully control for equine infectious anemia in China. This vaccine provides an important reference for studying the relationship between EIAV gene variation and changes in biological characteristics. Importantly, the vaccine provides a model for the investigation of lentiviral quasispecies evolution. This study followed the "natural" development of the attenuated EIAV vaccine by use of a systematic analysis of LTR evolution in vitro and in vivo The results revealed that the increase in LTR variation with passaging was accompanied by a decrease in virulence, which indicated that LTR variability might parallel the attenuation of virulence. Interestingly, the attenuated-virus-specific LTR variation was also detected in virulent-strain-infected horses, a finding consistent with those of previous investigations of gp90 and S2 evolution. Therefore, we present a hypothesis that the evolution of the attenuated virus may involve branching from EIAV quasispecies present in vivo.


Subject(s)
Equine Infectious Anemia/genetics , Evolution, Molecular , Infectious Anemia Virus, Equine/genetics , Terminal Repeat Sequences , Animals , Equine Infectious Anemia/metabolism , Horses , Infectious Anemia Virus, Equine/metabolism
9.
Biochem Biophys Res Commun ; 486(3): 712-719, 2017 05 06.
Article in English | MEDLINE | ID: mdl-28342872

ABSTRACT

The conjugation of small ubiquitin-like modifier (SUMO) to the target protein, namely, SUMOylation, is involved in the regulation of many important biological events including host-pathogen interaction. Some viruses have evolved to exploit the host SUMOylation machinery to modify their own protein. Retroviral Gag protein plays critical roles in the viral life cycle. The HIV-1 p6 and the Moloney murine leukemia virus CA have been reported to be conjugated with SUMO. In this study, we report for the first time, to our knowledge, the covalent conjugation of equine infectious anemia virus (EIAV) Gag with SUMO. The C-terminal p9 domain of Gag is a main target for SUMOylation and SUMO is attached to multiple sites of p9, including K30 whose mutation abolished p9 SUMOylation completely. The SUMOylation of p9, but not the p9-K30 mutant, was also detected in equine fibroblastic cells ATCC® CCL-57™. Ubc9 and its C93 residue are indispensable for the SUMOylation of p9. Using confocal microscopy, it is found that EIAV Gag localizes primarily, if not exclusively, in the cytoplasm of the cell and the co-localization of EIAV Gag with Ubc9 was observed. Our findings that EIAV Gag is SUMOylated at p9-K30, together with previous findings on the defects of p9-K30 mutant in viral DNA translocation from cytoplasm to the nucleus, suggests that SUMOylation of Gag may be involved in such functions.


Subject(s)
Gene Products, gag/genetics , Infectious Anemia Virus, Equine/genetics , Lysine/metabolism , SUMO-1 Protein/genetics , Ubiquitin-Conjugating Enzymes/genetics , Amino Acid Sequence , Animals , Cell Line , Escherichia coli/genetics , Escherichia coli/metabolism , Fibroblasts/metabolism , Fibroblasts/virology , Gene Expression Regulation , Gene Products, gag/metabolism , HEK293 Cells , Horses , Host-Pathogen Interactions , Humans , Infectious Anemia Virus, Equine/metabolism , Mutation , Protein Domains , SUMO-1 Protein/metabolism , Signal Transduction , Sumoylation , Ubiquitin-Conjugating Enzymes/metabolism
10.
Virology ; 491: 64-72, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26874586

ABSTRACT

Equine infectious anaemia virus (EIAV) and human immunodeficiency virus (HIV) are members of the lentiviral genus. Similar to HIV gp41, EIAV gp45 is a fusogenic protein that mediates fusion between the viral particle and the host cell membrane. The crystal structure of gp45 reported reveals a different conformation in the here that includes the fusion peptide proximal region (FPPR) and neighboring asparagine-rich layer compared with previous HIV-1 gp41 structures. A complicated hydrogen-bond network containing a cluster of solvent molecules appears to be critical for the stability of the gp45 helical bundle. Interestingly, viral replication was relatively unaffected by site-directed mutagenesis of EIAV, in striking contrast to that of HIV-1. Based on these observations, we speculate that EIAV is more adaptable to emergent mutations, which might be important for the evolution of EIAV as a quasi-species, and could potentially contribute to the success of the EIAV vaccine.


Subject(s)
Asparagine/metabolism , Equine Infectious Anemia/virology , Infectious Anemia Virus, Equine/metabolism , Peptides/metabolism , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/metabolism , Amino Acid Motifs , Animals , Asparagine/genetics , Crystallization , Horses , Hydrogen Bonding , Infectious Anemia Virus, Equine/chemistry , Infectious Anemia Virus, Equine/genetics , Peptides/chemistry , Peptides/genetics , Protein Structure, Secondary , Viral Envelope Proteins/genetics
11.
Vet Immunol Immunopathol ; 161(3-4): 161-9, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25176006

ABSTRACT

Unlike other lentiviruses, EIAV replication can be controlled in most infected horses leading to an inapparent carrier state free of overt clinical signs which lasts for many years. While the resolution of the initial infection is correlated with the appearance of virus specific cellular immune responses, the precise immune mechanisms responsible for control of the infection are not yet identified. Since the virus undergoes rapid mutation following infection, the immune response must also adapt to meet this challenge. We hypothesize that this adaptation involves peptide-specific recognition shifting from immunodominant variable determinants to conserved immunorecessive determinants following EIAV infection. Forty-four peptides, spanning the entire surface unit protein (gp90) of EIAV, were used to monitor peptide-specific T cell responses in vivo over a six-month period following infection. Peptides were injected intradermally and punch biopsies were collected for real-time PCR analysis to monitor the cellular peptide-specific immune responses in vivo. Similar to the CMI response to HIV infection, peptide-specific T cell recognition patterns changed over time. Early post infection (1 month), immune responses were directed to the peptides in the carboxyl-terminus variable region. By six months post infection, the peptide recognition spanned the entire gp90 sequence. These results indicate that peptide recognition broadens during EIAV infection.


Subject(s)
Epitopes , Equine Infectious Anemia/immunology , Glycoproteins/metabolism , Immunity, Cellular/physiology , Infectious Anemia Virus, Equine/metabolism , Amino Acid Sequence , Animals , Antibodies, Viral/blood , Antibody Affinity , Equine Infectious Anemia/metabolism , Gene Expression Regulation, Viral/immunology , Genetic Variation , Glycoproteins/genetics , Horses , Infectious Anemia Virus, Equine/genetics , Viral Proteins/genetics , Viral Proteins/metabolism
12.
Biochemistry ; 52(32): 5365-71, 2013 Aug 13.
Article in English | MEDLINE | ID: mdl-23906107

ABSTRACT

Capsid (CA) proteins from all retroviruses, including HIV-1, are structurally homologous dual-domain helical proteins. They form a capsid lattice composed of unitary symmetric CA hexamers. X-ray crystallography has shown that within each hexamer a monomeric CA adopts a single conformation, where most helices are parallel to the symmetry axis. In solution, large differences in averaged NMR spin relaxation rates for the two domains were observed, suggesting they are dynamically independent. One relevant question for the capsid assembly remains: whether the interdomain conformer within a hexamer unit needs to be induced or pre-exists within the conformational space of a monomeric CA. The latter seems more consistent with the relaxation data. However, possible CA protein oligomerization and the structure of each domain will affect relaxation measurements and data interpretation. This study, using CA proteins from equine infectious anemia virus (EIAV) as an example, demonstrates a linear extrapolation approach to obtain backbone (15)N spin relaxation time ratios T1/T2 for a monomeric EIAV-CA in the presence of oligomerization equilibrium. The interdomain motion turns out to be limited. The large difference in the domain averaged for a CA monomer is a consequence of the orthogonal distributions of helices in the two domains. The new monomeric interdomain conformation in solution is significantly different from that in CA hexamer. Therefore, if capsid assembly follows a nucleation-propagation process, the interdomain conformational change might be a key step during the nucleation, as the configuration in hexagonal assembly is never formed by diffusion of its two domains in solution.


Subject(s)
Capsid Proteins/chemistry , Infectious Anemia Virus, Equine/genetics , Capsid/metabolism , Capsid Proteins/metabolism , Crystallography, X-Ray , Infectious Anemia Virus, Equine/metabolism , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular , Protein Multimerization , Protein Structure, Tertiary , Retroviridae/genetics , Retroviridae/metabolism
13.
EMBO J ; 32(4): 538-51, 2013 Feb 20.
Article in English | MEDLINE | ID: mdl-23361315

ABSTRACT

The ubiquitylation signal promotes trafficking of endogenous and retroviral transmembrane proteins. The signal is decoded by a large set of ubiquitin (Ub) receptors that tether Ub-binding domains (UBDs) to the trafficking machinery. We developed a structure-based procedure to scan the protein data bank for hidden UBDs. The screen retrieved many of the known UBDs. Intriguingly, new potential UBDs were identified, including the ALIX-V domain. Pull-down, cross-linking and E3-independent ubiquitylation assays biochemically corroborated the in silico findings. Guided by the output model, we designed mutations at the postulated ALIX-V:Ub interface. Biophysical affinity measurements using microscale-thermophoresis of wild-type and mutant proteins revealed some of the interacting residues of the complex. ALIX-V binds mono-Ub with a K(d) of 119 µM. We show that ALIX-V oligomerizes with a Hill coefficient of 5.4 and IC(50) of 27.6 µM and that mono-Ub induces ALIX-V oligomerization. Moreover, we show that ALIX-V preferentially binds K63 di-Ub compared with mono-Ub and K48 di-Ub. Finally, an in vivo functionality assay demonstrates the significance of ALIX-V:Ub interaction in equine infectious anaemia virus budding. These results not only validate the new procedure, but also demonstrate that ALIX-V directly interacts with Ub in vivo and that this interaction can influence retroviral budding.


Subject(s)
Infectious Anemia Virus, Equine/metabolism , Multienzyme Complexes , Mutation , Ubiquitin-Protein Ligases , Virus Release/physiology , Animals , Humans , Infectious Anemia Virus, Equine/genetics , Mice , Models, Biological , Multienzyme Complexes/chemistry , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , Protein Structure, Tertiary , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
14.
J Virol ; 86(8): 4182-93, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22345468

ABSTRACT

We recently reported that human immunodeficiency virus type 1 (HIV-1) carrying PTAP and LYPX(n)L L domains ceased budding when the nucleocapsid (NC) domain was mutated, suggesting a role for NC in HIV-1 release. Here we investigated whether NC involvement in virus release is a property specific to HIV-1 or a general requirement of retroviruses. Specifically, we examined a possible role for NC in the budding of retroviruses relying on divergent L domains and structurally homologous NC domains that harbor diverse protein sequences. We found that NC is critical for the release of viruses utilizing the PTAP motif whether it functions within its native Gag in simian immunodeficiency virus cpzGAB2 (SIVcpzGAB2) or SIVsmmE543 or when it is transplanted into the heterologous Gag protein of equine infectious anemia virus (EIAV). In both cases, virus release was severely diminished even though NC mutant Gag proteins retained the ability to assemble spherical particles. Moreover, budding-defective NC mutants, which displayed particles tethered to the plasma membrane, were triggered to release virus when access to the cell endocytic sorting complex required for transport pathway was restored (i.e., in trans expression of Nedd4.2s). We also examined the role of NC in the budding of EIAV, a retrovirus relying exclusively on the (L)YPX(n)L-type L domain. We found that EIAV late budding defects were rescued by overexpression of the isolated Alix Bro1 domain (Bro1). Bro1-mediated rescue of EIAV release required the wild-type NC. EIAV NC mutants lost interactions with Bro1 and failed to produce viruses despite retaining the ability to self-assemble. Together, our studies establish a role for NC in the budding of retroviruses harboring divergent L domains and evolutionarily diverse NC sequences, suggesting the utilization of a common conserved mechanism and/or cellular factor rather than a specific motif.


Subject(s)
Nucleocapsid/chemistry , Nucleocapsid/metabolism , Retroviridae/metabolism , Virus Release , Amino Acid Motifs , Amino Acid Sequence , Animals , Cell Line , Endosomal Sorting Complexes Required for Transport/metabolism , Gene Products, gag/genetics , Humans , Infectious Anemia Virus, Equine/genetics , Infectious Anemia Virus, Equine/metabolism , Molecular Sequence Data , Mutation , Protein Binding , Protein Structure, Tertiary , Retroviridae/classification , Retroviridae/genetics , Sequence Alignment , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/metabolism , Virus Release/genetics
15.
Traffic ; 12(4): 438-51, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21176037

ABSTRACT

Phosphatidylinositol 4,5-biphosphate [PI(4,5)P(2) ], the predominant phosphoinositide (PI) on the plasma membrane, binds the matrix (MA) protein of human immunodeficiency virus type 1 (HIV-1) and equine infectious anemia virus (EIAV) with similar affinities in vitro. Interaction with PI(4,5)P(2) is critical for HIV-1 assembly on the plasma membrane. EIAV has been shown to localize in internal compartments; hence, the significance of its interaction with PI(4,5)P(2) is unclear. We therefore investigated the binding in vitro of other PIs to EIAV MA and whether intracellular association with compartments bearing these PIs was important for assembly and release of virus-like particles (VLPs) formed by Gag. In vitro, EIAV MA bound phosphatidylinositol 3-phosphate [PI(3)P] with higher affinity than PI(4,5)P(2) as revealed by nuclear magnetic resonance (NMR) spectra upon lipid titration. Gag was detected on the plasma membrane and in compartments enriched in phosphatidylinositol 3,5-biphosphate [PI(3,5)P(2) ]. Treatment of cells with YM201636, a kinase inhibitor that blocks production of PI(3,5)P(2) from PI(3)P, caused Gag to colocalize with aberrant compartments and inhibited VLP release. In contrast to HIV-1, release of EIAV VLPs was not significantly diminished by coexpression with 5-phosphatase IV, an enzyme that specifically depletes PI(4,5)P(2) from the plasma membrane. However, coexpression with synaptojanin 2, a phosphatase with broader specificity, diminished VLP production. PI-binding pocket mutations caused striking budding defects, as revealed by electron microscopy. One of the mutations also modified Gag-Gag interaction, as suggested by altered bimolecular fluorescence complementation. We conclude that PI-mediated targeting to peripheral and internal membranes is a critical factor in EIAV assembly and release.


Subject(s)
Gene Products, gag/metabolism , Infectious Anemia Virus, Equine/metabolism , Phosphatidylinositol Phosphates/metabolism , Acid Anhydride Hydrolases/metabolism , Aminopyridines/pharmacology , Animals , Antiviral Agents/pharmacology , COS Cells , Cell Membrane/metabolism , Cells, Cultured , Chlorocebus aethiops , Gene Products, gag/genetics , HIV-1/genetics , HIV-1/metabolism , HIV-1/physiology , Heterocyclic Compounds, 3-Ring/pharmacology , Horses , Humans , Infectious Anemia Virus, Equine/genetics , Infectious Anemia Virus, Equine/physiology , Mutation , Phosphatidylinositol Phosphates/antagonists & inhibitors , Phosphatidylinositol Phosphates/biosynthesis , Protein Binding/physiology , Protein Transport , Transfection , Virus Assembly/drug effects , Virus Assembly/physiology
16.
Virology ; 405(2): 414-23, 2010 Sep 30.
Article in English | MEDLINE | ID: mdl-20619429

ABSTRACT

In human cells, endogenous TRIM5alpha strongly inhibits N-tropic strains of murine leukemia virus (N-MLV) but does not target the closely related B-MLV. We have used a shRNA-based loss-of-function screen to isolate factors other than TRIM5alpha involved in the restriction of N-MLV. In one of the isolated clones, the shRNA expressed was found to target the murine double minute-2 mRNA. Knocking down MDM2 increased N-MLV and EIAV infection of human cells by 2- to 5-fold while having little effect on B-MLV. Similarly, knocking down MDM2 in African green monkey cells diminished the restriction of both N-MLV and HIV-1. Dual knockdown experiments showed that MDM2 was involved in the restriction mediated by TRIM5alpha. Moreover, MDM2 knockdown decreased the sensitivity of N-MLV infection to treatment with MG132 and As(2)O(3), two known TRIM5alpha pharmacological inhibitors. Altogether, our data suggest that MDM2 is a general but nonessential modulator of TRIM5alpha-mediated antiretroviral functions.


Subject(s)
Carrier Proteins/metabolism , Infectious Anemia Virus, Equine/pathogenicity , Leukemia Virus, Murine/pathogenicity , Proto-Oncogene Proteins c-mdm2/metabolism , RNA Interference , Animals , Antiviral Restriction Factors , Arsenic Trioxide , Arsenicals/pharmacology , Carrier Proteins/genetics , Cell Line , HIV-1/genetics , HIV-1/metabolism , HIV-1/pathogenicity , Humans , Infectious Anemia Virus, Equine/genetics , Infectious Anemia Virus, Equine/metabolism , Leukemia Virus, Murine/genetics , Leukemia Virus, Murine/metabolism , Leupeptins/pharmacology , Mice , Oxides/pharmacology , Proteasome Endopeptidase Complex , Proto-Oncogene Proteins c-mdm2/genetics , Tripartite Motif Proteins , Ubiquitin-Protein Ligases
17.
BMC Struct Biol ; 9: 74, 2009 Dec 17.
Article in English | MEDLINE | ID: mdl-20015412

ABSTRACT

BACKGROUND: The equine infection anemia virus (EIAV) p9 Gag protein contains the late (L-) domain required for efficient virus release of nascent virions from the cell membrane of infected cell. RESULTS: In the present study the p9 protein and N- and C-terminal fragments (residues 1-21 and 22-51, respectively) were chemically synthesized and used for structural analyses. Circular dichroism and 1H-NMR spectroscopy provide the first molecular insight into the secondary structure and folding of this 51-amino acid protein under different solution conditions. Qualitative 1H-chemical shift and NOE data indicate that in a pure aqueous environment p9 favors an unstructured state. In its most structured state under hydrophobic conditions, p9 adopts a stable helical structure within the C-terminus. Quantitative NOE data further revealed that this alpha-helix extends from Ser-27 to Ser-48, while the N-terminal residues remain unstructured. The structural elements identified for p9 differ substantially from that of the functional homologous HIV-1 p6 protein. CONCLUSIONS: These structural differences are discussed in the context of the different types of L-domains regulating distinct cellular pathways in virus budding. EIAV p9 mediates virus release by recruiting the ALG2-interacting protein X (ALIX) via the YPDL-motif to the site of virus budding, the counterpart of the YPXnL-motif found in p6. However, p6 contains an additional PTAP L-domain that promotes HIV-1 release by binding to the tumor susceptibility gene 101 (Tsg101). The notion that structures found in p9 differ form that of p6 further support the idea that different mechanisms regulate binding of ALIX to primary versus secondary L-domains types.


Subject(s)
Calcium-Binding Proteins/chemistry , Gene Products, gag/chemistry , HIV-1/chemistry , Infectious Anemia Virus, Equine/chemistry , gag Gene Products, Human Immunodeficiency Virus/chemistry , Amino Acid Sequence , Binding Sites , Calcium-Binding Proteins/metabolism , Circular Dichroism , Gene Products, gag/metabolism , HIV-1/metabolism , Infectious Anemia Virus, Equine/metabolism , Models, Molecular , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Protein Structure, Secondary , Protein Structure, Tertiary , Structural Homology, Protein , gag Gene Products, Human Immunodeficiency Virus/metabolism
18.
PLoS One ; 4(1): e4178, 2009.
Article in English | MEDLINE | ID: mdl-19137065

ABSTRACT

Rev is an essential regulatory protein in the equine infectious anemia virus (EIAV) and other lentiviruses, including HIV-1. It binds incompletely spliced viral mRNAs and shuttles them from the nucleus to the cytoplasm, a critical prerequisite for the production of viral structural proteins and genomic RNA. Despite its important role in production of infectious virus, the development of antiviral therapies directed against Rev has been hampered by the lack of an experimentally-determined structure of the full length protein. We have used a combined computational and biochemical approach to generate and evaluate a structural model of the Rev protein. The modeled EIAV Rev (ERev) structure includes a total of 6 helices, four of which form an anti-parallel four-helix bundle. The first helix contains the leucine-rich nuclear export signal (NES). An arginine-rich RNA binding motif, RRDRW, is located in a solvent-exposed loop region. An ERLE motif required for Rev activity is predicted to be buried in the core of modeled structure where it plays an essential role in stabilization of the Rev fold. This structural model is supported by existing genetic and functional data as well as by targeted mutagenesis of residues predicted to be essential for overall structural integrity. Our predicted structure should increase understanding of structure-function relationships in Rev and may provide a basis for the design of new therapies for lentiviral diseases.


Subject(s)
Gene Products, rev/chemistry , Infectious Anemia Virus, Equine/chemistry , Protein Structure, Tertiary , Amino Acid Sequence , Animals , Binding Sites , Gene Products, rev/genetics , Gene Products, rev/metabolism , Horse Diseases , Horses/microbiology , Infectious Anemia Virus, Equine/genetics , Infectious Anemia Virus, Equine/metabolism , Models, Structural , Molecular Sequence Data , Mutagenesis , Protein Structure, Tertiary/genetics , Structure-Activity Relationship
19.
Traffic ; 9(11): 1972-83, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18817521

ABSTRACT

Retroviral Gag polyprotein precursors are both necessary and sufficient for the assembly and release of virus-like particles (VLPs) from infected cells. It is well established that small Gag-encoded motifs, known as late domains, promote particle release by interacting with components of the cellular endosomal sorting and ubiquitination machinery. The Gag proteins of a number of different retroviruses are ubiquitinated; however, the role of Gag ubiquitination in particle egress remains undefined. In this study, we investigated this question by using a panel of equine infectious anemia virus (EIAV) Gag derivatives bearing the wild-type EIAV late domain, heterologous retroviral late domains or no late domain. Ubiquitin was fused in cis to the C-termini of these Gag polyproteins, and the effects on VLP budding were measured. Remarkably, fusion of ubiquitin to EIAV Gag lacking a late domain (EIAV/DeltaYPDL-Ub) largely rescued VLP release. We also determined the effects of ubiquitin fusion on the sensitivity of particle release to budding inhibitors and to depletion of key endosomal sorting factors. Ubiquitin fusion rendered EIAV/DeltaYPDL-Ub sensitive to depletion of cellular endosomal sorting factors Tsg101 and Alix and to overexpression of dominant-negative fragments of Tsg101 and Alix. These findings demonstrate that ubiquitin can functionally compensate for the absence of a retroviral late domain and provide insights into the host-cell machinery engaged by ubiquitin during particle egress.


Subject(s)
Gene Products, gag/metabolism , Infectious Anemia Virus, Equine/metabolism , Ubiquitin/metabolism , Cysteine Proteinase Inhibitors/pharmacology , Infectious Anemia Virus, Equine/drug effects , Infectious Anemia Virus, Equine/growth & development , Proteasome Inhibitors
20.
J Virol ; 82(19): 9425-32, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18667522

ABSTRACT

Wild-type strains of equine infectious anemia virus (EIAV) prevent superinfection of previously infected cells. A variant strain of virus that spontaneously arose during passage, EIAV(vMA-1c), can circumvent this mechanism in some cells, such as equine dermis (ED) cells, but not in others, such as equine endothelial cells. EIAV(vMA-1c) superinfection of ED cells results in a buildup of unintegrated viral DNA and rapid killing of the cell monolayer. Here, we examined the mechanism of resistance that is used by EIAV to prevent superinfection and explored the means by which EIAV(vMA-1c) overcomes this restriction. We found that the cellular receptor used by EIAV, equine lentivirus receptor 1 (ELR1), remains on the surface of cells chronically infected with EIAV, suggesting that wild-type EIAV interferes with superinfection by masking ELR1. The addition of soluble wild-type SU protein to the medium during infection blocked infection by wild-type strains of virus, implicating SU as the viral protein responsible for interfering with virion entry into previously infected cells. Additionally, interference of wild-type EIAV binding to ELR1 by the addition of either anti-ELR1 antibodies or the ELR1 ectodomain prevented entry of the wild-type strains of EIAV into two permissive cell populations. Many of these same interference treatments prevented EIAV(vMA-1c) infection of endothelial cells but only modestly affected the ability of EIAV(vMA-1c) to enter and kill previously infected ED cells. These findings indicate that EIAV(vMA-1c) retains the ability to use ELR1 for entry and suggest that this virus can interact with an additional, unidentified receptor to superinfect ED cells.


Subject(s)
Dermis/virology , Infectious Anemia Virus, Equine/genetics , Receptors, Virus/metabolism , Animals , Cell Line , Culture Media/metabolism , DNA, Viral/genetics , Fibroblasts/metabolism , Horses , Humans , Hydrogen-Ion Concentration , Infectious Anemia Virus, Equine/metabolism , Kinetics , Membrane Glycoproteins/metabolism , Phenotype , RNA, Small Interfering/metabolism , Viral Envelope Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...