Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 117
Filter
1.
Viruses ; 13(12)2021 12 06.
Article in English | MEDLINE | ID: mdl-34960718

ABSTRACT

Equine infectious anemia virus (EIAV) is a lentivirus similar to HIV that infects horses. Clinical and experimental studies demonstrating immune control of EIAV infection hold promise for efforts to produce an HIV vaccine. Antibody infusions have been shown to block both wild-type and mutant virus infection, but the mutant sometimes escapes. Using these data, we develop a mathematical model that describes the interactions between antibodies and both wild-type and mutant virus populations, in the context of continual virus mutation. The aim of this work is to determine whether repeated vaccinations through antibody infusions can reduce both the wild-type and mutant strains of the virus below one viral particle, and if so, to examine the vaccination period and number of infusions that ensure eradication. The antibody infusions are modelled using impulsive differential equations, a technique that offers insight into repeated vaccination by approximating the time-to-peak by an instantaneous change. We use impulsive theory to determine the maximal vaccination intervals that would be required to reduce the wild-type and mutant virus levels below one particle per horse. We show that seven boosts of the antibody vaccine are sufficient to eradicate both the wild-type and the mutant strains. In the case of a mutant virus infection that is given infusions of antibodies targeting wild-type virus (i.e., simulation of a heterologous infection), seven infusions were likewise sufficient to eradicate infection, based upon the data set. However, if the period between infusions was sufficiently increased, both the wild-type and mutant virus would eventually persist in the form of a periodic orbit. These results suggest a route forward to design antibody-based vaccine strategies to control viruses subject to mutant escape.


Subject(s)
Antibodies, Viral/immunology , Broadly Neutralizing Antibodies/immunology , Equine Infectious Anemia/therapy , Equine Infectious Anemia/virology , Immunization, Passive , Infectious Anemia Virus, Equine/genetics , Infectious Anemia Virus, Equine/immunology , Animals , Antibodies, Viral/administration & dosage , Broadly Neutralizing Antibodies/administration & dosage , Horses , Infectious Anemia Virus, Equine/physiology , Models, Biological , Mutation , Viral Load
2.
Emerg Microbes Infect ; 9(1): 1309-1320, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32525460

ABSTRACT

Lentiviruses harbour high genetic variability for efficient evasion from host immunity. An attenuated equine infectious anaemia (EIA) vaccine was developed decades ago in China and presented remarkably robust protection against EIA. The vaccine was recently proven to have high genomic diversity, particular in env. However, how and to what extent the high env diversity relates to immune protection remains unclear. In this study, we compared immune protections and responses of three groups of horses stimulated by the high-diversity vaccine EIAV_HD, a single molecular clone of the vaccine EIAV_LD with low env diversity, as well as a constructed vaccine strain EIAV_MD with moderate env diversity. The disparity of virus-host interactions between three env diversity-varied groups (5 horses in each group) was evaluated using clinical manifestation, pathological scores, and env-specific antibody. We found the highest titres of env antibodies (Abs) or neutralizing Abs (nAbs) in the EIAV_HD group, followed by the EIAV_MD group, and the lowest titres in the EIAV_LD group (P<0.05). The occurrence of disease/death was different between EIAV_HD group (1/0), EIAV_MD (2/2), and EIAV_LD group (4/2). A similar env diversity-related linear relationship was observed in the clinical manifestations and pathological changes. This diversity-dependent disparity in changes between the three groups was more distinct after immunosuppression, suggesting that env diversity plays an important role in protection under low host immunocompetence. In summary, inoculation with vaccines with higher genetic diversity could present broader and more efficient protection. Our findings strongly suggest that an abundance of Env antigens are required for efficient protection against lentiviruses.


Subject(s)
Equine Infectious Anemia/prevention & control , Gene Products, env/immunology , Infectious Anemia Virus, Equine/physiology , Polymorphism, Single Nucleotide , Viral Vaccines/administration & dosage , Animals , Antibodies, Neutralizing/metabolism , Antibodies, Viral/metabolism , Equine Infectious Anemia/immunology , Gene Products, env/genetics , High-Throughput Nucleotide Sequencing , Horses , Vaccines, Attenuated , Viral Vaccines/immunology , Viral Vaccines/pharmacology , Virus Replication/drug effects
3.
PLoS Pathog ; 16(1): e1008277, 2020 01.
Article in English | MEDLINE | ID: mdl-31986188

ABSTRACT

Retrovirus assembly is driven by the multidomain structural protein Gag. Interactions between the capsid domains (CA) of Gag result in Gag multimerization, leading to an immature virus particle that is formed by a protein lattice based on dimeric, trimeric, and hexameric protein contacts. Among retroviruses the inter- and intra-hexamer contacts differ, especially in the N-terminal sub-domain of CA (CANTD). For HIV-1 the cellular molecule inositol hexakisphosphate (IP6) interacts with and stabilizes the immature hexamer, and is required for production of infectious virus particles. We have used in vitro assembly, cryo-electron tomography and subtomogram averaging, atomistic molecular dynamics simulations and mutational analyses to study the HIV-related lentivirus equine infectious anemia virus (EIAV). In particular, we sought to understand the structural conservation of the immature lentivirus lattice and the role of IP6 in EIAV assembly. Similar to HIV-1, IP6 strongly promoted in vitro assembly of EIAV Gag proteins into virus-like particles (VLPs), which took three morphologically highly distinct forms: narrow tubes, wide tubes, and spheres. Structural characterization of these VLPs to sub-4Å resolution unexpectedly showed that all three morphologies are based on an immature lattice with preserved key structural components, highlighting the structural versatility of CA to form immature assemblies. A direct comparison between EIAV and HIV revealed that both lentiviruses maintain similar immature interfaces, which are established by both conserved and non-conserved residues. In both EIAV and HIV-1, IP6 regulates immature assembly via conserved lysine residues within the CACTD and SP. Lastly, we demonstrate that IP6 stimulates in vitro assembly of immature particles of several other retroviruses in the lentivirus genus, suggesting a conserved role for IP6 in lentiviral assembly.


Subject(s)
Equine Infectious Anemia/metabolism , Gene Products, gag/chemistry , Gene Products, gag/metabolism , Infectious Anemia Virus, Equine/physiology , Phytic Acid/metabolism , Virion/physiology , Amino Acid Sequence , Animals , Electron Microscope Tomography , Equine Infectious Anemia/virology , Gene Products, gag/genetics , HIV Infections/metabolism , HIV Infections/virology , HIV-1/genetics , HIV-1/physiology , HIV-1/ultrastructure , Horses , Host-Pathogen Interactions , Infectious Anemia Virus, Equine/chemistry , Infectious Anemia Virus, Equine/genetics , Infectious Anemia Virus, Equine/ultrastructure , Sequence Alignment , Virion/genetics , Virion/ultrastructure , Virus Assembly , gag Gene Products, Human Immunodeficiency Virus/chemistry , gag Gene Products, Human Immunodeficiency Virus/genetics , gag Gene Products, Human Immunodeficiency Virus/metabolism
5.
J Virol ; 92(18)2018 09 15.
Article in English | MEDLINE | ID: mdl-29743377

ABSTRACT

Human myxovirus resistance protein 2 (huMxB) has been shown to be a determinant type I interferon (IFN)-induced host factor involved in the inhibition of human immunodeficiency virus type 1 (HIV-1) as well as many other primate lentiviruses. This blocking occurs after the reverse transcription of viral RNA and ahead of integration into the host DNA, which is closely connected to the ability of the protein to bind the viral capsid. To date, Mx2s derived from nonprimate animals have shown no capacity for HIV-1 suppression. In this study, we examined the restrictive effect of equine Mx2 (eqMx2) on both equine infectious anemia virus (EIAV) and HIV-1 and investigated possible mechanisms for its specific function. We demonstrated that IFN-α/ß upregulates the expression of eqMx2 in equine monocyte-derived macrophages (eMDMs). The overexpression of eqMx2 significantly suppresses the replication of EIAV, HIV-1, and simian immunodeficiency viruses (SIVs) but not that of murine leukemia virus (MLV). The knockdown of eqMx2 transcription weakens the inhibition of EIAV replication by type I interferon. Interestingly, data from immunofluorescence assays suggest that the subcellular localization of eqMx2 changes following virus infection, from being dispersed in the cytoplasm to being accumulated at the nuclear envelope. Furthermore, eqMx2 blocks the nuclear uptake of the proviral genome by binding to the viral capsid. The N-terminally truncated mutant of eqMx2 lost the ability to bind the viral capsid as well as the restriction effect for lentiviruses. These results improve our understanding of the Mx2 protein in nonprimate animals.IMPORTANCE Previous research has shown that the antiviral ability of Mx2s is confined to primates, particularly humans. EIAV has been shown to be insensitive to restriction by human MxB. Here, we describe the function of equine Mx2. This protein plays an important role in the suppression of EIAV, HIV-1, and SIVs. The antiviral activity of eqMx2 depends on its subcellular location as well as its capsid binding capacity. Our results showed that following viral infection, eqMx2 changes its original cytoplasmic location and accumulates at the nuclear envelope, where it binds to the viral capsid and blocks the nuclear entry of reverse-transcribed proviral DNAs. In contrast, huMxB does not bind to the EIAV capsid and shows no EIAV restriction effect. These studies expand our understanding of the function of the equine Mx2 protein.


Subject(s)
Capsid Proteins/metabolism , HIV-1/physiology , Infectious Anemia Virus, Equine/physiology , Myxovirus Resistance Proteins/genetics , Virus Replication/genetics , Animals , Capsid Proteins/antagonists & inhibitors , Cytoplasm/physiology , Cytoplasm/ultrastructure , Cytoplasm/virology , HIV-1/genetics , Horses , Infectious Anemia Virus, Equine/genetics , Interferon-alpha/genetics , Leukemia Virus, Murine/physiology , Macrophages/virology , Myxovirus Resistance Proteins/deficiency , Myxovirus Resistance Proteins/metabolism , Nuclear Envelope/metabolism , Nuclear Envelope/ultrastructure , Simian Immunodeficiency Virus/physiology
6.
mBio ; 7(6)2016 11 22.
Article in English | MEDLINE | ID: mdl-27879338

ABSTRACT

Gammaretroviruses, such as murine leukemia viruses (MLVs), encode, in addition to the canonical Gag, Pol, and Env proteins that will form progeny virus particles, a protein called "glycogag" (glycosylated Gag). MLV glycogag contains the entire Gag sequence plus an 88-residue N-terminal extension. It has recently been reported that glycogag, like the Nef protein of HIV-1, counteracts the antiviral effects of the cellular protein Serinc5. We have found, in agreement with prior work, that glycogag strongly enhances the infectivity of MLVs with some Env proteins but not those with others. In contrast, however, glycogag was detrimental to MLVs carrying Ebolavirus glycoprotein. Glycogag could be replaced, with respect to viral infectivity, by the unrelated S2 protein of equine infectious anemia virus. We devised an assay for viral entry in which virus particles deliver the Cre recombinase into cells, leading to the expression of a reporter. Data from this assay showed that both the positive and the negative effects of glycogag and S2 upon MLV infectivity are exerted at the level of virus entry. Moreover, transfection of the virus-producing cells with a Serinc5 expression plasmid reduced the infectivity and entry capability of MLV carrying xenotropic MLV Env, particularly in the absence of glycogag. Conversely, Serinc5 expression abrogated the negative effects of glycogag upon the infectivity and entry capability of MLV carrying Ebolavirus glycoprotein. As Serinc5 may influence cellular phospholipid metabolism, it seems possible that all of these effects on virus entry derive from changes in the lipid composition of viral membranes. IMPORTANCE: Many murine leukemia viruses (MLVs) encode a protein called "glycogag." The function of glycogag is not fully understood, but it can assist HIV-1 replication in the absence of the HIV-1 protein Nef under some circumstances. In turn, Nef counteracts the cellular protein Serinc5. Glycogag enhances the infectivity of MLVs with some but not all MLV Env proteins (which mediate viral entry into the host cell upon binding to cell surface receptors). We now report that glycogag acts by enhancing viral entry and that, like Nef, glycogag antagonizes Serinc5. Surprisingly, the effects of glycogag and Serinc5 upon the entry and infectivity of MLV particles carrying an Ebolavirus glycoprotein are the opposite of those observed with the MLV Env proteins. The unrelated S2 protein of equine infectious anemia virus (EIAV) is functionally analogous to glycogag in our experiments. Thus, three retroviruses (HIV-1, MLV, and EIAV) have independently evolved accessory proteins that counteract Serinc5.


Subject(s)
Ebolavirus/physiology , Host-Pathogen Interactions , Leukemia Virus, Murine/physiology , Membrane Glycoproteins/metabolism , Membrane Proteins/metabolism , Viral Proteins/metabolism , Virus Internalization , Animals , Cells, Cultured , Infectious Anemia Virus, Equine/physiology , Mice
7.
Virology ; 495: 112-21, 2016 08.
Article in English | MEDLINE | ID: mdl-27200480

ABSTRACT

Human schlafen11 is a novel restriction factor for HIV-1 based on bias regarding relative synonymous codon usage (RSCU). Here, we report the cloning of equine schlafen11 (eSLFN11) and the characteristics of its role in restricting the production of equine infectious anemia virus (EIAV), a retrovirus similar to HIV-1. Overexpression of eSLFN11 inhibited EIAV replication, whereas knockdown of endogenous eSLFN11 by siRNA enhanced the release of EIAV from its principal target cell. Notably, although eSLFN11 significantly suppressed expression of viral Gag protein and EIAV release into the culture medium, the levels of intracellular viral early gene proteins Tat and Rev and viral genomic RNA were unaffected. Coincidently, similar altered patterns of codon usage bias were observed for both the early and late genes of EIAV. Therefore, our data suggest that eSLFN11 restricts EIAV production by impairing viral mRNA translation via a mechanism that is similar to that employed by hSLFN11 for HIV-1.


Subject(s)
Codon , Infectious Anemia Virus, Equine/physiology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Replication , Amino Acid Sequence , Animals , Gene Expression Regulation, Viral , Gene Knockdown Techniques , Genome, Viral , HIV-1/physiology , Horses , Humans , Nuclear Proteins/chemistry , RNA, Small Interfering/genetics , RNA, Viral , Transcription, Genetic , Viral Proteins/chemistry , Virus Release
8.
J Virol ; 90(4): 1824-38, 2016 02 15.
Article in English | MEDLINE | ID: mdl-26637458

ABSTRACT

UNLABELLED: Gag intracellular assembly and export are very important processes for lentiviruses replication. Previous studies have demonstrated that equine infectious anemia virus (EIAV) matrix (MA) possesses distinct phosphoinositide affinity compared with HIV-1 MA and that phosphoinositide-mediated targeting to peripheral and internal membranes is a critical factor in EIAV assembly and release. In this study, we compared the cellular assembly sites of EIAV and HIV-1. We observed that the assembly of EIAV particles occurred on interior cellular membranes, while HIV-1 was targeted to the plasma membrane (PM) for assembly. Then, we determined that W7 and K9 in the EIAV MA N terminus were essential for Gag assembly and release but did not affect the cellular distribution of Gag. The replacement of EIAV MA with HIV-1 MA directed chimeric Gag to the PM but severely impaired Gag release. MA structural analysis indicated that the EIAV and HIV-1 MAs had similar spatial structures but that helix 1 of the EIAV MA was closer to loop 2. Further investigation indicated that EIAV Gag accumulated in the trans-Golgi network (TGN) but not the early and late endosomes. The 9 N-terminal amino acids of EIAV MA harbored the signal that directed Gag to the TGN membrane system. Additionally, we demonstrated that EIAV particles were transported to the extracellular space by the cellular vesicle system. This type of EIAV export was not associated with multivesicular bodies or microtubule depolymerization but could be inhibited by the actin-depolymerizing drug cytochalasin D, suggesting that dynamic actin depolymerization may be associated with EIAV production. IMPORTANCE: In previous studies, EIAV Gag was reported to localize to both the cell interior and the plasma membrane. Here, we demonstrate that EIAV likely uses the TGN as the assembly site in contrast to HIV-1, which is targeted to the PM for assembly. These distinct assembly features are determined by the MA domain. We also identified two sites in the N terminus of EIAV MA that were important for Gag assembly and release. Furthermore, the observation of EIAV transport by cellular vesicles but not by multivesicular bodies sheds light on the mechanisms underlying EIAV cellular replication.


Subject(s)
Cytoplasmic Vesicles/metabolism , Gene Products, gag/metabolism , Infectious Anemia Virus, Equine/physiology , Viral Matrix Proteins/metabolism , Virus Assembly , trans-Golgi Network/metabolism , HIV-1/physiology , Humans , Protein Transport
9.
Rev Sci Tech ; 34(2): 513-23, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26601453

ABSTRACT

There is no credible evidence that the lentivirus that causes equine infectious anaemia (EIA) replicates in invertebrates. The virus persistently infects its equid hosts and is often present in blood in significant quantities. Blood-feeding arthropods thus have the potential to transfer the virus between hosts, especially if their feeding on the first host is interrupted and immediately continued on a second host. The general details and dynamics of mechanical transmission are included in this paper, as this agent presents an excellent model. Mechanical transmission can be effectively controlled if the dynamics and interactions of the host, virus and vector populations are understood. Efficient transmission is proportional to the amount of agent found in the source material, the environmental survival of the agent, the number of vector feedings, the number of interrupted feedings, vector refeeding, the proximity of infected and naive hosts, host population density, and the length of time during which vectors and hosts are in contact. Establishing firm quantitative risk estimates for EIA is impossible, mainly because the virus content in blood can change exponentially from day to day. The EIA virus can be transmitted by horse flies for at least 30 minutes after feeding on a horse with acute signs of EIA, butthe probability of a horse fly being interrupted and completing its blood feeding on a second host at a distance of 50 m is very low, and the separation of infected and uninfected equids by 200 m breaks transmission. The statements above assume that human interactions are absent or do not contribute to the risk of virus transmission; however, the risk from human interventions, such as the too-often-used procedure of administering > 200 ml of plasma to foals, can easily be more than 10(7) times greater than the risk posed by a single horse fly. Controlling EIA depends upon the identification of persistently infected equids by serological testing because other methods of identifying infective virus orviral genetic material are less accurate or impractical.


Subject(s)
Culicidae/virology , Diptera/virology , Equine Infectious Anemia/transmission , Infectious Anemia Virus, Equine/physiology , Insect Vectors/virology , Animals , Horses , Humans
10.
Viruses ; 7(6): 3241-60, 2015 Jun 19.
Article in English | MEDLINE | ID: mdl-26102582

ABSTRACT

Human immunodeficiency virus (HIV)-1 has a unique integration profile in the human genome relative to murine and avian retroviruses. Equine infectious anemia virus (EIAV) is another well-studied lentivirus that can also be used as a promising retro-transfection vector, but its integration into its native host has not been characterized. In this study, we mapped 477 integration sites of the EIAV strain EIAVFDDV13 in fetal equine dermal (FED) cells during in vitro infection. Published integration sites of EIAV and HIV-1 in the human genome were also analyzed as references. Our results demonstrated that EIAVFDDV13 tended to integrate into genes and AT-rich regions, and it avoided integrating into transcription start sites (TSS), which is consistent with EIAV and HIV-1 integration in the human genome. Notably, the integration of EIAVFDDV13 favored long interspersed elements (LINEs) and DNA transposons in the horse genome, whereas the integration of HIV-1 favored short interspersed elements (SINEs) in the human genome. The chromosomal environment near LINEs or DNA transposons potentially influences viral transcription and may be related to the unique EIAV latency states in equids. The data on EIAV integration in its natural host will facilitate studies on lentiviral infection and lentivirus-based therapeutic vectors.


Subject(s)
Chromosomes/virology , DNA, Viral/analysis , Genetic Loci , Infectious Anemia Virus, Equine/physiology , Proviruses/genetics , Virus Integration , Animals , Cells, Cultured , DNA, Viral/genetics , Epithelial Cells/virology , Genome , HIV-1/genetics , HIV-1/physiology , Horses , Humans , Infectious Anemia Virus, Equine/genetics
11.
Virology ; 476: 364-371, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25589239

ABSTRACT

Adenosine deaminases that act on RNA (ADARs) have been reported to be functional on various viruses. ADAR1 may exhibit antiviral or proviral activity depending on the type of virus. Human immunodeficiency virus (HIV)-1 is the most well-studied lentivirus with respect to its interaction with ADAR1, and variable results have been reported. In this study, we demonstrated that equine ADAR1 (eADAR1) was a positive regulator of equine infectious anemia virus (EIAV), another lentivirus of the Retroviridae family. First, eADAR1 significantly promoted EIAV replication, and the enhancement of viral protein expression was associated with the long terminal repeat (LTR) and Rev response element (RRE) regions. Second, the RNA binding domain 1 of eADAR1 was essential only for enhancing LTR-mediated gene expression. Third, in contrast with APOBEC proteins, which have been shown to reduce lentiviral infectivity, eADAR1 increased the EIAV infectivity. This study indicated that eADAR1 was proviral rather than antiviral for EIAV.


Subject(s)
Adenosine Deaminase/metabolism , Equine Infectious Anemia/enzymology , Infectious Anemia Virus, Equine/physiology , Infectious Anemia Virus, Equine/pathogenicity , RNA, Double-Stranded/metabolism , RNA, Viral/metabolism , Virus Replication , Adenosine Deaminase/genetics , Animals , Cell Line , Equine Infectious Anemia/genetics , Equine Infectious Anemia/virology , Horses , Host-Pathogen Interactions , Infectious Anemia Virus, Equine/genetics , Protein Structure, Tertiary , RNA, Double-Stranded/genetics , RNA, Viral/chemistry , RNA, Viral/genetics , Terminal Repeat Sequences , Virulence
12.
Retrovirology ; 11: 115, 2014 Dec 23.
Article in English | MEDLINE | ID: mdl-25533001

ABSTRACT

BACKGROUND: The lentiviral Rev protein mediates nuclear export of intron-containing viral RNAs that encode structural proteins or serve as the viral genome. Following translation, HIV-1 Rev localizes to the nucleus and binds its cognate sequence, termed the Rev-responsive element (RRE), in incompletely spliced viral RNA. Rev subsequently multimerizes along the viral RNA and associates with the cellular Crm1 export machinery to translocate the RNA-protein complex to the cytoplasm. Equine infectious anemia virus (EIAV) Rev is functionally homologous to HIV-1 Rev, but shares very little sequence similarity and differs in domain organization. EIAV Rev also contains a bipartite RNA binding domain comprising two short arginine-rich motifs (designated ARM-1 and ARM-2) spaced 79 residues apart in the amino acid sequence. To gain insight into the topology of the bipartite RNA binding domain, a computational approach was used to model the tertiary structure of EIAV Rev. RESULTS: The tertiary structure of EIAV Rev was modeled using several protein structure prediction and model quality assessment servers. Two types of structures were predicted: an elongated structure with an extended central alpha helix, and a globular structure with a central bundle of helices. Assessment of models on the basis of biophysical properties indicated they were of average quality. In almost all models, ARM-1 and ARM-2 were spatially separated by >15 Å, suggesting that they do not form a single RNA binding interface on the monomer. A highly conserved canonical coiled-coil motif was identified in the central region of EIAV Rev, suggesting that an RNA binding interface could be formed through dimerization of Rev and juxtaposition of ARM-1 and ARM-2. In support of this, purified Rev protein migrated as a dimer in Blue native gels, and mutation of a residue predicted to form a key coiled-coil contact disrupted dimerization and abrogated RNA binding. In contrast, mutation of residues outside the predicted coiled-coil interface had no effect on dimerization or RNA binding. CONCLUSIONS: Our results suggest that EIAV Rev binding to the RRE requires dimerization via a coiled-coil motif to juxtapose two RNA binding motifs, ARM-1 and ARM-2.


Subject(s)
Gene Products, rev/chemistry , Gene Products, rev/metabolism , Infectious Anemia Virus, Equine/physiology , Protein Multimerization , RNA, Viral/metabolism , Models, Molecular , Protein Binding , Protein Conformation
13.
Vet Res ; 45: 82, 2014 Aug 09.
Article in English | MEDLINE | ID: mdl-25106750

ABSTRACT

The Chinese attenuated equine infectious anemia virus (EIAV) vaccine has successfully protected millions of equine animals from EIA disease in China. Given that the induction of immune protection results from the interactions between viruses and hosts, a better understanding of the characteristics of vaccine strain infection and host responses would be useful for elucidating the mechanism of the induction of immune protection by the Chinese attenuated EIAV strain. In this study, we demonstrate in equine monocyte-derived macrophages (eMDM) that EIAVFDDV13, a Chinese attenuated EIAV strain, induced a strong resistance to subsequent infection by a pathogenic strain, EIAVUK3. Further experiments indicate that the expression of the soluble EIAV receptor sELR1, Toll-like receptor 3 (TLR3) and interferon ß (IFNß) was up-regulated in eMDM infected with EIAVFDDV13 compared with eMDM infected with EIAVUK3. Stimulating eMDM with poly I:C resulted in similar resistance to EIAV infection as induced by EIAVFDDV13 and was correlated with enhanced TLR3, sELR1 and IFNß expression. The knock down of TLR3 mRNA significantly impaired poly I:C-stimulated resistance to EIAV, greatly reducing the expression of sELR1 and IFNß and lowered the level of infection resistance induced by EIAVFDDV13. These results indicate that the induction of restraining infection by EIAVFDDV13 in macrophages is partially mediated through the up-regulated expression of the soluble viral receptor and IFNß, and that the TLR3 pathway activation plays an important role in the development of an EIAV-resistant intracellular environment.


Subject(s)
Equine Infectious Anemia/immunology , Gene Expression Regulation , Horse Diseases/immunology , Infectious Anemia Virus, Equine/physiology , Viral Vaccines/immunology , Animals , Disease Resistance , Enzyme-Linked Immunosorbent Assay/veterinary , Equine Infectious Anemia/genetics , Equine Infectious Anemia/virology , Horse Diseases/genetics , Horse Diseases/virology , Horses , Infectious Anemia Virus, Equine/genetics , Interferon-beta/genetics , Interferon-beta/metabolism , Macrophages/immunology , Real-Time Polymerase Chain Reaction/veterinary , Receptors, Virus/genetics , Receptors, Virus/metabolism , Reverse Transcriptase Polymerase Chain Reaction/veterinary , Toll-Like Receptor 3/genetics , Toll-Like Receptor 3/metabolism
14.
J Virol ; 88(21): 12296-310, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25122784

ABSTRACT

UNLABELLED: Viperin is an endoplasmic reticulum (ER)-associated multifunctional protein that regulates virus replication and possesses broad antiviral activity. In many cases, viperin interferes with the trafficking and budding of viral structural proteins by distorting the membrane transportation system. The lentivirus equine infectious anemia virus (EIAV) has been studied extensively. In this study, we examined the restrictive effect of equine viperin (eViperin) on EIAV replication and investigated the possible molecular basis of this restriction to obtain insights into the effect of this cellular factor on retroviruses. We demonstrated that EIAV infection of primary equine monocyte-derived macrophages (eMDMs) upregulated the expression of eViperin. The overexpression of eViperin significantly inhibited the replication of EIAV in eMDMs, and knockdown of eViperin transcription enhanced the replication of EIAV in eMDMs by approximately 45.8%. Further experiments indicated that eViperin restricts EIAV at multiple steps of viral replication. The overexpression of eViperin inhibited EIAV Gag release. Both the α-helix domain and radical S-adenosylmethionine (SAM) domain were required for this activity. However, the essential motifs in SAM were different from those reported for the inhibition of HIV-1 Gag by human viperin. Furthermore, eViperin disrupted the synthesis of both EIAV Env and receptor, which consequently inhibited viral production and entry, respectively, and this disruption was dependent on the eViperin α-helix domain. Using immunofluorescence assays and electron microscopy, we demonstrated that the α-helix domain is responsible for the distortion of the endoplasmic reticulum (ER). Finally, EIAV did not exhibit counteracting eViperin at the protein level. IMPORTANCE: In previous studies, viperin was indicated as restricting virus replications primarily by the inhibition of virus budding. Here, we show that viperin may have multiple antiviral mechanisms, including the reduction of EIAV Gag budding and Env expression, and these activities are dependent on different viperin domains. We especially demonstrate that the overexpression of viperin inhibits EIAV entry by decreasing the level of virus receptor. Therefore, viperin restriction of viruses is determined largely by the dependence of virus on the cellular membrane transportation system.


Subject(s)
Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/virology , Host-Pathogen Interactions , Infectious Anemia Virus, Equine/immunology , Infectious Anemia Virus, Equine/physiology , Viral Proteins/metabolism , Virus Replication , Animals , Cells, Cultured , Endoplasmic Reticulum/ultrastructure , Fluorescent Antibody Technique , HIV-1 , Horses , Macrophages/immunology , Macrophages/virology , Microscopy, Electron , Virus Release
15.
Retrovirology ; 11: 26, 2014 Mar 21.
Article in English | MEDLINE | ID: mdl-24656154

ABSTRACT

BACKGROUND: The equine infectious anemia virus (EIAV) is a lentivirus of the Retrovirus family, which causes persistent infection in horses often characterized by recurrent episodes of high fever. It has a similar morphology and life cycle to the human immunodeficiency virus (HIV). Its transmembrane glycoprotein, gp45 (analogous to gp41 in HIV), mediates membrane fusion during the infection. However, the post-fusion conformation of EIAV gp45 has not yet been determined. EIAV is the first member of the lentiviruses for which an effective vaccine has been successfully developed. The attenuated vaccine strain, FDDV, has been produced from a pathogenic strain by a series of passages in donkey dermal cells. We have previously reported that a V/I505T mutation in gp45, in combination with other mutations in gp90, may potentially contribute to the success of the vaccine strain. To this end, we now report on our structural and biochemical studies of the gp45 protein from both wide type and vaccine strain, providing a valuable structural model for the advancement of the EIAV vaccine. RESULTS: We resolved crystal structures of the ecto-domain of gp45 from both the wild-type EIAV and the vaccine strain FDDV. We found that the V/I505T mutation in gp45 was located in a highly conserved d position within the heptad repeat, which protruded into a 3-fold symmetry axis within the six-helix bundle. Our crystal structure analyses revealed a shift of a hydrophobic to hydrophilic interaction due to this specific mutation, and further biochemical and virological studies confirmed that the mutation reduced the overall stability of the six-helix bundle in post-fusion conformation. Moreover, we found that altering the temperatures drastically affected the viral infectivity. CONCLUSIONS: Our high-resolution crystal structures of gp45 exhibited high conservation between the gp45/gp41 structures of lentiviruses. In addition, a hydrophobic to hydrophilic interaction change in the EIAV vaccine strain was found to modulate the stability and thermal-sensitivity of the overall gp45 structure. Our observations suggest that lowering the stability of the six-helix bundle (post-fusion), which may stabilizes the pre-fusion conformation, might be one of the reasons of acquired dominance for FDDV in viral attenuation.


Subject(s)
Infectious Anemia Virus, Equine/physiology , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Mutation, Missense , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/metabolism , Animals , Crystallography, X-Ray , Horses , Infectious Anemia Virus, Equine/chemistry , Infectious Anemia Virus, Equine/genetics , Mutant Proteins/genetics , Protein Conformation , Protein Stability/radiation effects , Temperature , Viral Envelope Proteins/genetics , Viral Vaccines/chemistry , Viral Vaccines/genetics
16.
J Theor Biol ; 343: 1-8, 2014 Feb 21.
Article in English | MEDLINE | ID: mdl-24252283

ABSTRACT

We develop a mathematical model for the interaction between two competing equine infectious anemia virus strains and neutralizing antibodies. We predict that elimination of one or both virus strains depends on the initial antibody levels, the strength of antibody mediated neutralization, and the persistence of antibody over time. We further show that the ability of a subdominant, neutralization resistant virus to dominate the infection transiently or permanently is dependent on the antibody-mediated neutralization effect. Finally, we determine conditions for persistence of both virus strains. We fit our models to virus titers from horses (foals) with severe combined immunodeficiency to estimate virus-host parameters and to validate analytical results.


Subject(s)
Equine Infectious Anemia/virology , Horses/virology , Host-Pathogen Interactions , Infectious Anemia Virus, Equine/physiology , Severe Combined Immunodeficiency/veterinary , Severe Combined Immunodeficiency/virology , Animals , Computer Simulation , Models, Biological , Mutation/genetics , Numerical Analysis, Computer-Assisted , RNA, Viral/metabolism
17.
J Virol ; 88(2): 1259-70, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24227834

ABSTRACT

Human tetherin is a host restriction factor that inhibits replication of enveloped viruses by blocking viral release. Tetherin has an unusual topology that includes an N-terminal cytoplasmic tail, a single transmembrane domain, an extracellular domain, and a C-terminal glycosylphosphatidylinositol anchor. Tetherin is not well conserved across species, so it inhibits viral replication in a species-specific manner. Thus, studies of tetherin activities from different species provide an important tool for understanding its antiviral mechanism. Here, we report cloning of equine tetherin and characterization of its antiviral activity. Equine tetherin shares 53%, 40%, 36%, and 34% amino acid sequence identity with feline, human, simian, and murine tetherins, respectively. Like the feline tetherin, equine tetherin has a shorter N-terminal domain than human tetherin. Equine tetherin is localized on the cell surface and strongly blocks human immunodeficiency virus type 1 (HIV-1), simian immunodeficiency virus (SIV), and equine infectious anemia virus (EIAV) release from virus-producing cells. The antiviral activity of equine tetherin is neutralized by EIAV envelope protein, but not by the HIV-1 accessory protein Vpu, which is a human tetherin antagonist, and EIAV envelope protein does not counteract human tetherin. These results shed new light on our understanding of the species-specific tetherin antiviral mechanism.


Subject(s)
Antigens, CD/metabolism , Equine Infectious Anemia/metabolism , Infectious Anemia Virus, Equine/physiology , Viral Envelope Proteins/metabolism , Virus Release , Amino Acid Sequence , Animals , Antigens, CD/chemistry , Antigens, CD/genetics , Base Sequence , Cats , Cell Line , Cloning, Molecular , Equine Infectious Anemia/genetics , Equine Infectious Anemia/virology , Horses , Humans , Infectious Anemia Virus, Equine/genetics , Mice , Molecular Sequence Data , Retroviridae/genetics , Retroviridae/physiology , Sequence Alignment , Viral Envelope Proteins/genetics
18.
Vet Res ; 44: 113, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24289102

ABSTRACT

EIA (Equine Infectious Anemia) is a blood-borne disease primarily transmitted by haematophagous insects or needle punctures. Other routes of transmission have been poorly explored. We evaluated the potential of EIAV (Equine Infectious Anemia Virus) to induce pulmonary lesions in naturally infected equids. Lungs from 77 EIAV seropositive horses have been collected in Romania and France. Three types of lesions have been scored on paraffin-embedded lungs: lymphocyte infiltration, bronchiolar inflammation, and thickness of the alveolar septa. Expression of the p26 EIAV capsid (CA) protein has been evaluated by immunostaining. Compared to EIAV-negative horses, 52% of the EIAV-positive horses displayed a mild inflammation around the bronchioles, 22% had a moderate inflammation with inflammatory cells inside the wall and epithelial bronchiolar hyperplasia and 6.5% had a moderate to severe inflammation, with destruction of the bronchiolar epithelium and accumulation of smooth muscle cells within the pulmonary parenchyma. Changes in the thickness of the alveolar septa were also present. Expression of EIAV capsid has been evidenced in macrophages, endothelial as well as in alveolar and bronchiolar epithelial cells, as determined by their morphology and localization. To summarize, we found lesions of interstitial lung disease similar to that observed during other lentiviral infections such as FIV in cats, SRLV in sheep and goats or HIV in children. The presence of EIAV capsid in lung epithelial cells suggests that EIAV might be responsible for the broncho-interstitial damages observed.


Subject(s)
Epithelial Cells/pathology , Equine Infectious Anemia/pathology , Horse Diseases/pathology , Lung Diseases, Interstitial/veterinary , Lung/pathology , Viral Core Proteins/genetics , Animals , Blotting, Western/veterinary , Epithelial Cells/virology , Equine Infectious Anemia/genetics , Equine Infectious Anemia/virology , Female , France , Horse Diseases/genetics , Horse Diseases/virology , Horses , Infectious Anemia Virus, Equine/physiology , Lung/virology , Lung Diseases, Interstitial/genetics , Lung Diseases, Interstitial/pathology , Lung Diseases, Interstitial/virology , Male , Microscopy, Fluorescence/veterinary , Romania , Viral Core Proteins/metabolism
19.
Vet Microbiol ; 167(1-2): 181-204, 2013 Nov 29.
Article in English | MEDLINE | ID: mdl-24183747

ABSTRACT

A detailed description of equine infectious anemia virus and host responses to it are presented. Current control and eradication of the infection are discussed with suggestions for improvements to increase their effectiveness.


Subject(s)
Equine Infectious Anemia/virology , Infectious Anemia Virus, Equine/physiology , Animals , Equine Infectious Anemia/diagnosis , Equine Infectious Anemia/immunology , Equine Infectious Anemia/pathology , Equine Infectious Anemia/transmission , Horses , Host-Pathogen Interactions , Infectious Anemia Virus, Equine/classification , Infectious Anemia Virus, Equine/genetics , Phylogeny , Viral Proteins/genetics , Viral Proteins/metabolism
20.
Retrovirology ; 10: 104, 2013 Oct 09.
Article in English | MEDLINE | ID: mdl-24107264

ABSTRACT

BACKGROUND: Retroviruses and many other enveloped viruses usurp the cellular ESCRT pathway to bud from cells. However, the stepwise process of ESCRT-mediated virus budding can be challenging to analyze in retroviruses like HIV-1 that recruit multiple different ESCRT factors to initiate budding. RESULTS: In this study, we characterized the ESCRT factor requirements for budding of Equine Infectious Anemia Virus (EIAV), whose only known direct ESCRT protein interaction is with ALIX. siRNA depletion of endogenous ESCRT proteins and "rescue" experiments with exogenous siRNA-resistant wild type and mutant constructs revealed budding requirements for the following ESCRT proteins: ALIX, CHMP4B, CHMP2A and VPS4A or VPS4B. EIAV budding was inhibited by point mutations that abrogate the direct interactions between ALIX:CHMP4B, CHMP4B:CHMP2A, and CHMP2A:VPS4A/B, indicating that each of these interactions is required for EIAV budding. Unexpectedly, CHMP4B depletion led to formation of multi-lobed and long tubular EIAV virions. CONCLUSIONS: We conclude that EIAV budding requires an ESCRT protein network that comprises EIAV Gag-ALIX-CHMP4B-CHMP2A-VPS4 interactions. Our experiments also suggest that CHMP4B recruitment/polymerization helps control Gag polymerization and/or processing to ensure that ESCRT factor assembly and membrane fission occur at the proper stage of virion assembly. These studies help establish EIAV as a streamlined model system for dissecting the stepwise processes of lentivirus assembly and ESCRT-mediated budding.


Subject(s)
Endosomal Sorting Complexes Required for Transport/metabolism , Host-Pathogen Interactions , Infectious Anemia Virus, Equine/physiology , Virus Release , Cell Line , Humans , Protein Binding , Protein Interaction Mapping
SELECTION OF CITATIONS
SEARCH DETAIL
...