Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters










Publication year range
1.
PLoS Comput Biol ; 18(4): e1010053, 2022 04.
Article in English | MEDLINE | ID: mdl-35468127

ABSTRACT

In HIV-1-infected individuals, transmitted/founder (TF) virus contributes to establish new infection and expands during the acute phase of infection, while chronic control (CC) virus emerges during the chronic phase of infection. TF viruses are more resistant to interferon-alpha (IFN-α)-mediated antiviral effects than CC virus, however, its virological relevance in infected individuals remains unclear. Here we perform an experimental-mathematical investigation and reveal that IFN-α strongly inhibits cell-to-cell infection by CC virus but only weakly affects that by TF virus. Surprisingly, IFN-α enhances cell-free infection of HIV-1, particularly that of CC virus, in a virus-cell density-dependent manner. We further demonstrate that LY6E, an IFN-stimulated gene, can contribute to the density-dependent enhancement of cell-free HIV-1 infection. Altogether, our findings suggest that the major difference between TF and CC viruses can be explained by their resistance to IFN-α-mediated inhibition of cell-to-cell infection and their sensitivity to IFN-α-mediated enhancement of cell-free infection.


Subject(s)
HIV Infections , HIV-1 , Antiviral Agents , HIV Infections/drug therapy , Humans , Interferon-alpha/pharmacology
2.
PLoS Pathog ; 16(9): e1008812, 2020 09.
Article in English | MEDLINE | ID: mdl-32913367

ABSTRACT

The APOBEC3 deaminases are potent inhibitors of virus replication and barriers to cross-species transmission. For simian immunodeficiency virus (SIV) to transmit to a new primate host, as happened multiple times to seed the ongoing HIV-1 epidemic, the viral infectivity factor (Vif) must be capable of neutralizing the APOBEC3 enzymes of the new host. Although much is known about current interactions of HIV-1 Vif and human APOBEC3s, the evolutionary changes in SIV Vif required for transmission from chimpanzees to gorillas and ultimately to humans are poorly understood. Here, we demonstrate that gorilla APOBEC3G is a factor with the potential to hamper SIV transmission from chimpanzees to gorillas. Gain-of-function experiments using SIVcpzPtt Vif revealed that this barrier could be overcome by a single Vif acidic amino acid substitution (M16E). Moreover, degradation of gorilla APOBEC3F is induced by Vif through a mechanism that is distinct from that of human APOBEC3F. Thus, our findings identify virus adaptations in gorillas that preceded and may have facilitated transmission to humans.


Subject(s)
APOBEC-3G Deaminase/metabolism , Evolution, Molecular , Gene Products, vif/metabolism , Host-Pathogen Interactions , Simian Acquired Immunodeficiency Syndrome/transmission , Simian Immunodeficiency Virus/isolation & purification , Virus Replication , APOBEC-3G Deaminase/chemistry , APOBEC-3G Deaminase/genetics , Amino Acid Sequence , Animals , Gene Products, vif/chemistry , Gene Products, vif/genetics , Gorilla gorilla , Humans , Pan troglodytes , Phylogeny , Protein Conformation , Sequence Homology , Simian Acquired Immunodeficiency Syndrome/virology
3.
J Theor Biol ; 498: 110295, 2020 08 07.
Article in English | MEDLINE | ID: mdl-32335137

ABSTRACT

APOBEC3 proteins inhibit human immunodeficiency virus (HIV)-1 infection by independently impairing viral reverse transcription and inducing G-to-A mutations in viral DNA. An HIV-1-encoded protein, viral infectivity factor (Vif), can counteract these antiviral activities of APOBEC3 proteins. Although previous studies using in vitro cell culture systems have revealed the molecular mechanisms of the antiviral action of APOBEC3 proteins and their antagonism by Vif, it remains unclear how APOBEC3 proteins affect the kinetics of HIV-1 replication in vivo. Here we quantified the time-series of viral load datasets from humanized mice infected with HIV-1 variants in the presence of APOBEC3F, APOBEC3G, or both APOBEC3F/G using a simple mathematical model that accounted for inter-individual variability. Through experimental and mathematical investigation, we formulated and calculated the total antiviral activity of APOBEC3F and APOBEC3G based on the estimated initial growth rates of viral loads in vivo. Interestingly, we quantitatively demonstrated that compared with APOBEC3G, the antiviral activity of APOBEC3F was widely distributed but skewed toward lower activity, although their mean values were similar. We concluded that APOBEC3G markedly and robustly restricted the initial stages of viral growth in vivo. This is the first report to quantitatively elucidate how APOBEC3F and APOBEC3G differ in their anti-HIV-1 modes in vivo.


Subject(s)
HIV Infections , HIV-1 , APOBEC-3G Deaminase , Animals , Antiviral Agents , Cytidine Deaminase , Cytosine Deaminase , Mice
4.
Nat Microbiol ; 4(9): 1532-1544, 2019 09.
Article in English | MEDLINE | ID: mdl-31133753

ABSTRACT

RNA-modulating factors not only regulate multiple steps of cellular RNA metabolism, but also emerge as key effectors of the immune response against invading viral pathogens including human immunodeficiency virus type-1 (HIV-1). However, the cellular RNA-binding proteins involved in the establishment and maintenance of latent HIV-1 reservoirs have not been extensively studied. Here, we screened a panel of 62 cellular RNA-binding proteins and identified NEDD4-binding protein 1 (N4BP1) as a potent interferon-inducible inhibitor of HIV-1 in primary T cells and macrophages. N4BP1 harbours a prototypical PilT N terminus-like RNase domain and inhibits HIV-1 replication by interacting with and degrading viral mRNA species. Following activation of CD4+ T cells, however, N4BP1 undergoes rapid cleavage at Arg 509 by the paracaspase named mucosa-associated lymphoid tissue lymphoma translocation 1 (MALT1). Mutational analyses and knockout studies revealed that MALT1-mediated inactivation of N4BP1 facilitates the reactivation of latent HIV-1 proviruses. Taken together, our findings demonstrate that the RNase N4BP1 is an efficient restriction factor of HIV-1 and suggest that inactivation of N4BP1 by induction of MALT1 activation might facilitate elimination of latent HIV-1 reservoirs.


Subject(s)
HIV Infections/virology , HIV-1/physiology , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/metabolism , Nuclear Proteins/metabolism , RNA-Binding Proteins/metabolism , Virus Activation/genetics , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/virology , Cell Line , Gene Expression/drug effects , HIV Infections/metabolism , Humans , Interferon-alpha/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Macrophages/virology , Mice , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/genetics , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Protein Domains , RNA, Messenger/metabolism , RNA, Viral/metabolism , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , Receptors, Antigen, T-Cell/metabolism , Virus Latency
5.
Sci Rep ; 8(1): 6770, 2018 Apr 25.
Article in English | MEDLINE | ID: mdl-29691441

ABSTRACT

A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has not been fixed in the paper.

6.
J Virol ; 92(4)2018 02 15.
Article in English | MEDLINE | ID: mdl-29212937

ABSTRACT

Human immunodeficiency virus type 1 (HIV-1), the causative agent of AIDS, originated from simian immunodeficiency virus from chimpanzees (SIVcpz), the precursor of the human virus, approximately 100 years ago. This indicates that HIV-1 has emerged through the cross-species transmission of SIVcpz from chimpanzees to humans. However, it remains unclear how SIVcpz has evolved into pandemic HIV-1 in humans. To address this question, we inoculated three SIVcpz strains (MB897, EK505, and MT145), four pandemic HIV-1 strains (NL4-3, NLCSFV3, JRCSF, and AD8), and two nonpandemic HIV-1 strains (YBF30 and DJO0131). Humanized mice infected with SIVcpz strain MB897, a virus phylogenetically similar to pandemic HIV-1, exhibited a peak viral load comparable to that of mice infected with pandemic HIV-1, while peak viral loads of mice infected with SIVcpz strain EK505 or MT145 as well as nonpandemic HIV-1 strains were significantly lower. These results suggest that SIVcpz strain MB897 is preadapted to humans, unlike the other SIVcpz strains. Moreover, viral RNA sequencing of MB897-infected humanized mice identified a nonsynonymous mutation in env, a G413R substitution in gp120. The infectivity of the gp120 G413R mutant of MB897 was significantly higher than that of parental MB897. Furthermore, we demonstrated that the gp120 G413R mutant of MB897 augments the capacity for viral replication in both in vitro cell cultures and humanized mice. Taken together, this is the first experimental investigation to use an animal model to demonstrate a gain-of-function evolution of SIVcpz into pandemic HIV-1.IMPORTANCE From the mid-20th century, humans have been exposed to the menace of infectious viral diseases, such as severe acute respiratory syndrome coronavirus, Ebola virus, and Zika virus. These outbreaks of emerging/reemerging viruses can be triggered by cross-species viral transmission from wild animals to humans, or zoonoses. HIV-1, the causative agent of AIDS, emerged by the cross-species transmission of SIVcpz, the HIV-1 precursor in chimpanzees, around 100 years ago. However, the process by which SIVcpz evolved to become HIV-1 in humans remains unclear. Here, by using a hematopoietic stem cell-transplanted humanized-mouse model, we experimentally recapitulate the evolutionary process of SIVcpz to become HIV-1. We provide evidence suggesting that a strain of SIVcpz, MB897, preadapted to infect humans over other SIVcpz strains. We further demonstrate a gain-of-function evolution of SIVcpz in infected humanized mice. Our study reveals that pandemic HIV-1 has emerged through at least two steps: preadaptation and subsequent gain-of-function mutations.


Subject(s)
Evolution, Molecular , HIV-1/genetics , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/genetics , Zoonoses/transmission , Animals , Animals, Wild/virology , Disease Models, Animal , Female , HEK293 Cells , Humans , Male , Mice , Mice, Knockout , Pan troglodytes/virology , Phylogeny , RNA, Viral/genetics , Viral Load , Virus Replication
8.
Sci Rep ; 7(1): 6913, 2017 07 31.
Article in English | MEDLINE | ID: mdl-28761140

ABSTRACT

Combination anti-retroviral therapy (cART) has drastically improved the clinical outcome of HIV-1 infection. Nonetheless, despite effective cART, HIV-1 persists indefinitely in infected individuals. Clonal expansion of HIV-1-infected cells in peripheral blood has been reported recently. cART is effective in stopping the retroviral replication cycle, but not in inhibiting clonal expansion of the infected host cells. Thus, the proliferation of HIV-1-infected cells may play a role in viral persistence, but little is known about the kinetics of the generation, the tissue distribution or the underlying mechanism of clonal expansion in vivo. Here we analyzed the clonality of HIV-1-infected cells using high-throughput integration site analysis in a hematopoietic stem cell-transplanted humanized mouse model. Clonally expanded, HIV-1-infected cells were detectable at two weeks post infection, their abundance increased with time, and certain clones were present in multiple organs. Expansion of HIV-1-infected clones was significantly more frequent when the provirus was integrated near host genes in specific gene ontological classes, including cell activation and chromatin regulation. These results identify potential drivers of clonal expansion of HIV-1-infected cells in vivo.


Subject(s)
Clone Cells/virology , HIV Infections/genetics , HIV Infections/virology , HIV-1/physiology , Sequence Analysis, RNA/methods , Animals , CD4-Positive T-Lymphocytes/metabolism , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Gene Regulatory Networks , HIV Infections/immunology , High-Throughput Nucleotide Sequencing/methods , Humans , Jurkat Cells , Lymphocyte Activation , Mice , RNA, Viral/analysis , Tissue Distribution , Viral Load , Virus Integration , Virus Latency
9.
PLoS Pathog ; 13(5): e1006348, 2017 May.
Article in English | MEDLINE | ID: mdl-28475648

ABSTRACT

APOBEC3 (A3) family proteins are DNA cytosine deaminases recognized for contributing to HIV-1 restriction and mutation. Prior studies have demonstrated that A3D, A3F, and A3G enzymes elicit a robust anti-HIV-1 effect in cell cultures and in humanized mouse models. Human A3H is polymorphic and can be categorized into three phenotypes: stable, intermediate, and unstable. However, the anti-viral effect of endogenous A3H in vivo has yet to be examined. Here we utilize a hematopoietic stem cell-transplanted humanized mouse model and demonstrate that stable A3H robustly affects HIV-1 fitness in vivo. In contrast, the selection pressure mediated by intermediate A3H is relaxed. Intriguingly, viral genomic RNA sequencing reveled that HIV-1 frequently adapts to better counteract stable A3H during replication in humanized mice. Molecular phylogenetic analyses and mathematical modeling suggest that stable A3H may be a critical factor in human-to-human viral transmission. Taken together, this study provides evidence that stable variants of A3H impose selective pressure on HIV-1.


Subject(s)
Aminohydrolases/genetics , Cytosine Deaminase/genetics , HIV Infections/virology , HIV-1/physiology , vif Gene Products, Human Immunodeficiency Virus/genetics , APOBEC Deaminases , Aminohydrolases/metabolism , Animals , Cytidine Deaminase , Cytosine Deaminase/metabolism , Disease Models, Animal , HEK293 Cells , HIV Infections/transmission , HIV-1/genetics , Humans , Mice , Mice, Knockout , Models, Genetic , Mutation , Phylogeny , RNA, Viral/chemistry , RNA, Viral/genetics , Sequence Analysis, RNA , Virus Replication
10.
J Virol ; 91(11)2017 06 01.
Article in English | MEDLINE | ID: mdl-28331087

ABSTRACT

The interplay between viral and host proteins has been well studied to elucidate virus-host interactions and their relevance to virulence. Mammalian genes encode apolipoprotein B mRNA-editing enzyme catalytic polypeptide-like 3 (APOBEC3) proteins, which act as intrinsic restriction factors against lentiviruses. To overcome APOBEC3-mediated antiviral actions, lentiviruses have evolutionarily acquired an accessory protein, viral infectivity factor (Vif), and Vif degrades host APOBEC3 proteins via a ubiquitin/proteasome-dependent pathway. Although the Vif-APOBEC3 interaction and its evolutionary significance, particularly those of primate lentiviruses (including HIV) and primates (including humans), have been well investigated, those of nonprimate lentiviruses and nonprimates are poorly understood. Moreover, the factors that determine lentiviral pathogenicity remain unclear. Here, we focus on feline immunodeficiency virus (FIV), a pathogenic lentivirus in domestic cats, and the interaction between FIV Vif and feline APOBEC3 in terms of viral virulence and evolution. We reveal the significantly reduced diversity of FIV subtype B compared to that of other subtypes, which may associate with the low pathogenicity of this subtype. We also demonstrate that FIV subtype B Vif is less active with regard to feline APOBEC3 degradation. More intriguingly, we further reveal that FIV protease cleaves feline APOBEC3 in released virions. Taken together, our findings provide evidence that a lentivirus encodes two types of anti-APOBEC3 factors, Vif and viral protease.IMPORTANCE During the history of mammalian evolution, mammals coevolved with retroviruses, including lentiviruses. All pathogenic lentiviruses, excluding equine infectious anemia virus, have acquired the vif gene via evolution to combat APOBEC3 proteins, which are intrinsic restriction factors against exogenous lentiviruses. Here we demonstrate that FIV, a pathogenic lentivirus in domestic cats, antagonizes feline APOBEC3 proteins by both Vif and a viral protease. Furthermore, the Vif proteins of an FIV subtype (subtype B) have attenuated their anti-APOBEC3 activity through evolution. Our findings can be a clue to elucidate the complicated evolutionary processes by which lentiviruses adapt to mammals.


Subject(s)
APOBEC Deaminases/antagonists & inhibitors , Aspartic Acid Endopeptidases/metabolism , Gene Products, vif/metabolism , Immunodeficiency Virus, Feline/genetics , APOBEC Deaminases/metabolism , Animals , Aspartic Acid Endopeptidases/genetics , Cats , Evolution, Molecular , Gene Products, vif/genetics , Host-Pathogen Interactions , Immunodeficiency Virus, Feline/metabolism , Immunodeficiency Virus, Feline/pathogenicity , Virulence
11.
Sci Rep ; 6: 33988, 2016 Sep 26.
Article in English | MEDLINE | ID: mdl-27665724

ABSTRACT

Mammals have co-evolved with lentiviruses for a long time. As evidence, viral infectivity factor (Vif), encoded by lentiviruses, antagonizes the anti-viral action of cellular APOBEC3 of their hosts. Here, we address the co-evolutionary dynamics of bovine APOBEC3 and the following two bovine lentiviruses: bovine immunodeficiency virus (BIV) and Jembrana disease virus (JDV). We determined the sequences of three APOBEC3 genes of bovids belonging to the genera Bos and Bison and showed that bovine APOBEC3Z3 is under a strong positive selection. We found that APOBEC3Z3 of gaur, a bovid in the genus Bos, acquired resistance to JDV Vif-mediated degradation after diverging from the other bovids through conversion of the structural composition of the loop 1 domain. Interestingly, the resistance of gaur APOBEC3Z3 can be attributed to the positive selection of residue 62. This study provides the first evidence, suggesting that a co-evolutionary arms race between bovids and lentiviruses occurred in Asia.

12.
Microbiol Immunol ; 60(7): 483-96, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27278725

ABSTRACT

The range of genome-editing tools has recently been expanded. In particular, an RNA-guided genome-editing tool, the clustered regularly interspaced short palindromic repeat (CRISPR)-associated 9 (Cas9) system, has many applications for human diseases. In this study, guide RNA (gRNA) to target gag, pol and a long terminal repeat of HIV-1 was designed and used to generate gRNA-expressing lentiviral vectors. An HIV-1-specific gRNA and Cas9 were stably dually transduced into a highly HIV-1-susceptible human T-cell line and the inhibitory ability of the anti-HIV-1 CRISPR/Cas9 lentiviral vector assessed. Although clear inhibition of the early phase of HIV-1 infection was observed, as evaluated by a VSV-G-pseudotyped HIV-1 reporter system, the anti-HIV-1 potency in multiple rounds of wild type (WT) viral replication was insufficient, either because of generation of resistant viruses or overcoming of the activity of the WT virus. Thus, there are potential difficulties that must be addressed when considering anti-HIV-1 treatment with the CRISPR/Cas9 system alone.


Subject(s)
CRISPR-Cas Systems , HIV-1/genetics , Virus Replication , Cell Line , Cells, Cultured , Gene Editing , Gene Targeting , Genes, Viral , HIV Infections/virology , Humans , Mutation , RNA, Guide, Kinetoplastida , Virus Integration
13.
Microbiol Immunol ; 60(6): 427-36, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27193350

ABSTRACT

Mammals have co-evolved with retroviruses, including lentiviruses, over a long period. Evidence supporting this contention is that viral infectivity factor (Vif) encoded by lentiviruses antagonizes the anti-viral action of cellular apolipoprotein B mRNA editing enzyme catalytic polypeptide-like 3 (APOBEC3) of the host. To orchestrate E3 ubiquitin ligase complex for APOBEC3 degradation, Vifs utilize mammalian proteins such as core-binding factor beta (CBFB; for primate lentiviruses) or cyclophilin A (CYPA; for Maedi-Visna virus [MVV]). However, the co-evolutionary relationship between lentiviral Vif and the mammalian proteins associated with Vif-mediated APOBEC3 degradation is poorly understood. Moreover, it is unclear whether Vif proteins of small ruminant lentiviruses (SRLVs), including MVV and caprine arthritis encephalitis virus (CAEV), commonly utilize CYPA to degrade the APOBEC3 of their hosts. In this study, molecular phylogenetic and protein homology modeling revealed that Vif co-factors are evolutionarily and structurally conserved. It was also found that not only MVV but also CAEV Vifs degrade APOBEC3 of both sheep and goats and that CAEV Vifs interact with CYPA. These findings suggest that lentiviral Vifs chose evolutionarily and structurally stable proteins as their partners (e.g., CBFB or CYPA) for APOBEC3 degradation and, particularly, that SRLV Vifs evolved to utilize CYPA as their co-factor in degradation of ovine and caprine APOBEC3.


Subject(s)
Arthritis-Encephalitis Virus, Caprine/genetics , Cyclophilin A/genetics , Cyclophilin A/metabolism , Cytidine Deaminase/metabolism , Gene Products, vif/genetics , Gene Products, vif/metabolism , Animals , Arthritis-Encephalitis Virus, Caprine/metabolism , Cells, Cultured , Core Binding Factor beta Subunit/genetics , Core Binding Factor beta Subunit/metabolism , Cyclophilins/genetics , Cyclophilins/metabolism , Cytidine Deaminase/genetics , Evolution, Molecular , Goats , HEK293 Cells , Host-Pathogen Interactions , Humans , Interleukin-2/genetics , Phylogeny , Sheep
14.
Retrovirology ; 13: 23, 2016 Apr 18.
Article in English | MEDLINE | ID: mdl-27086687

ABSTRACT

BACKGROUND: Tetherin is an intrinsic anti-viral factor impairing the release of nascent HIV-1 particles from infected cells. Vpu, an HIV-1 accessory protein, antagonizes the anti-viral action of tetherin. Although previous studies using in vitro cell culture systems have revealed the molecular mechanisms of the anti-viral action of tetherin and the antagonizing action of Vpu against tetherin, it still remains unclear how Vpu affects the kinetics of HIV-1 replication in vivo. RESULTS: To quantitatively assess the role of Vpu in viral replication in vivo, we analyzed time courses of experimental data with viral load and target cell levels in the peripheral blood of humanized mice infected with wild-type and vpu-deficient HIV-1. Our recently developed mathematical model describes the acute phase of this infection reasonably, and allowed us to estimate several parameters characterizing HIV-1 infection in mice. Using a technique of Bayesian parameter estimation, we estimate distributions of the basic reproduction number of wild-type and vpu-deficient HIV-1. This reveals that Vpu markedly increases the rate of viral replication in vivo. CONCLUSIONS: Combining experiments with mathematical modeling, we provide an estimate for the contribution of Vpu to viral replication in humanized mice.


Subject(s)
HIV-1/physiology , Human Immunodeficiency Virus Proteins/metabolism , Viral Regulatory and Accessory Proteins/metabolism , Virus Replication , Animals , Humans , Mice , Mice, SCID , Models, Theoretical
15.
Microbiol Immunol ; 60(4): 272-9, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26935128

ABSTRACT

How host-virus co-evolutionary relationships manifest is one of the most intriguing issues in virology. To address this topic, the mammal-lentivirus relationship can be considered as an interplay of cellular and viral proteins, particularly apolipoprotein B mRNA editing enzyme catalytic polypeptide-like 3 (APOBEC3) and viral infectivity factor (Vif). APOBEC3s enzymatically restrict lentivirus replication, whereas Vif antagonizes the host anti-viral action mediated by APOBEC3. In this study, the focus was on the interplay between feline APOBEC3 proteins and two feline immunodeficiency viruses in cats and pumas. To our knowledge, this study provides the first evidence of non-primate lentiviral Vif being incapable of counteracting a natural host's anti-viral activity mediated via APOBEC3 protein.


Subject(s)
Cytosine Deaminase/metabolism , Gene Products, vif/metabolism , Immunodeficiency Virus, Feline/metabolism , APOBEC Deaminases , Animals , Cats , Cytidine Deaminase , Cytosine Deaminase/genetics , Evolution, Molecular , Gene Products, vif/genetics , Gene Products, vif/immunology , Host-Pathogen Interactions , Immunity, Innate , Immunodeficiency Virus, Feline/genetics , Immunodeficiency Virus, Feline/immunology , Puma , Species Specificity , Virus Diseases/veterinary , Virus Replication
16.
J Virol ; 90(1): 474-85, 2016 01 01.
Article in English | MEDLINE | ID: mdl-26491161

ABSTRACT

UNLABELLED: Apolipoprotein B mRNA-editing enzyme catalytic polypeptide-like 3 (APOBEC3; A3) DNA cytosine deaminases can be incorporated into progeny virions and inhibit lentiviral replication. On the other hand, viral infectivity factor (Vif) of lentiviruses antagonizes A3-mediated antiviral activities by degrading A3 proteins. It is known that domestic cat (Felis catus) APOBEC3Z3 (A3Z3), the ortholog of human APOBEC3H, potently suppresses the infectivity of vif-defective feline immunodeficiency virus (FIV). Although a recent report has shown that domestic cat encodes 7 haplotypes (hap I to hap VII) of A3Z3, the relevance of A3Z3 polymorphism in domestic cats with FIV Vif has not yet been addressed. In this study, we demonstrated that these feline A3Z3 variants suppress vif-defective FIV infectivity. We also revealed that codon 65 of feline A3Z3 is a positively selected site and that A3Z3 hap V is subject to positive selection during evolution. It is particularly noteworthy that feline A3Z3 hap V is resistant to FIV Vif-mediated degradation and still inhibits vif-proficient viral infection. Moreover, the side chain size, but not the hydrophobicity, of the amino acid at position 65 determines the resistance to FIV Vif-mediated degradation. Furthermore, phylogenetic analyses have led to the inference that feline A3Z3 hap V emerged approximately 60,000 years ago. Taken together, these findings suggest that feline A3Z3 hap V may have been selected for escape from an ancestral FIV. This is the first evidence for an evolutionary "arms race" between the domestic cat and its cognate lentivirus. IMPORTANCE: Gene diversity and selective pressure are intriguing topics in the field of evolutionary biology. A direct interaction between a cellular protein and a viral protein can precipitate an evolutionary arms race between host and virus. One example is primate APOBEC3G, which potently restricts the replication of primate lentiviruses (e.g., human immunodeficiency virus type 1 [HIV-1] and simian immunodeficiency virus [SIV]) if its activity is not counteracted by the viral Vif protein. Here we investigate the ability of 7 naturally occurring variants of feline APOBEC3, APOBEC3Z3 (A3Z3), to inhibit FIV replication. Interestingly, one feline A3Z3 variant is dominant, restrictive, and naturally resistant to FIV Vif-mediated degradation. Phylogenetic analyses revealed that the ancestral change that generated this variant could have been caused by positive Darwinian selection, presumably due to an ancestral FIV infection. The experimental-phylogenetic investigation sheds light on the evolutionary history of the domestic cat, which was likely influenced by lentiviral infection.


Subject(s)
Cytidine Deaminase/metabolism , Host-Pathogen Interactions , Immunity, Innate , Immunodeficiency Virus, Feline/immunology , Immunodeficiency Virus, Feline/physiology , Virus Replication , Animals , Cats , Cytidine Deaminase/genetics , Evolution, Molecular , Gene Products, vif/deficiency , Selection, Genetic
17.
Sci Rep ; 5: 16021, 2015 Nov 04.
Article in English | MEDLINE | ID: mdl-26531727

ABSTRACT

Human immunodeficiency virus, a primate lentivirus (PLV), causes AIDS in humans, whereas most PLVs are less or not pathogenic in monkeys. These notions suggest that the co-evolutionary process of PLVs and their hosts associates with viral pathogenicity, and therefore, that elucidating the history of virus-host co-evolution is one of the most intriguing topics in the field of virology. To address this, recent studies have focused on the interplay between intrinsic anti-viral proteins, such as tetherin, and viral antagonists. Through an experimental-phylogenetic approach, here we investigate the co-evolutionary interplay between tribe Cercopithecini tetherin and viral antagonists, Nef and Vpu. We reveal that tribe Cercopithecini tetherins are positively selected, possibly triggered by ancient Nef-like factor(s). We reconstruct the ancestral sequence of tribe Cercopithecini tetherin and demonstrate that all Nef proteins are capable of antagonizing ancestral Cercopithecini tetherin. Further, we consider the significance of evolutionary arms race between tribe Cercopithecini and their PLVs.


Subject(s)
CD4 Antigens/metabolism , Cercopithecinae/virology , Human Immunodeficiency Virus Proteins/metabolism , Simian Immunodeficiency Virus/pathogenicity , Viral Regulatory and Accessory Proteins/metabolism , nef Gene Products, Human Immunodeficiency Virus/metabolism , Animals , Biological Evolution , Cell Line , HEK293 Cells , HIV-1/genetics , Humans , Selection, Genetic , Simian Immunodeficiency Virus/genetics
18.
Elife ; 42015 Oct 06.
Article in English | MEDLINE | ID: mdl-26441404

ABSTRACT

Cell-to-cell viral infection, in which viruses spread through contact of infected cell with surrounding uninfected cells, has been considered as a critical mode of virus infection. However, since it is technically difficult to experimentally discriminate the two modes of viral infection, namely cell-free infection and cell-to-cell infection, the quantitative information that underlies cell-to-cell infection has yet to be elucidated, and its impact on virus spread remains unclear. To address this fundamental question in virology, we quantitatively analyzed the dynamics of cell-to-cell and cell-free human immunodeficiency virus type 1 (HIV-1) infections through experimental-mathematical investigation. Our analyses demonstrated that the cell-to-cell infection mode accounts for approximately 60% of viral infection, and this infection mode shortens the generation time of viruses by 0.9 times and increases the viral fitness by 3.9 times. Our results suggest that even a complete block of the cell-free infection would provide only a limited impact on HIV-1 spread.


Subject(s)
HIV Infections/virology , HIV-1/physiology , Virus Internalization , Virus Release , Humans , Jurkat Cells , Models, Theoretical
19.
Sci Rep ; 5: 14040, 2015 Sep 14.
Article in English | MEDLINE | ID: mdl-26364986

ABSTRACT

During 2001-2002 and 2008-2011, two epidemic outbreaks of infectious hemorrhagic disease have been found in Japanese macaques (Macaca fuscata) in Kyoto University Primate Research Institute, Japan. Following investigations revealed that the causative agent was simian retrovirus type 4 (SRV-4). SRV-4 was isolated by using human cell lines, which indicates that human cells are potently susceptible to SRV-4 infection. These raise a possibility of zoonotic infection of pathogenic SRV-4 from Japanese macaques into humans. To explore the possibility of zoonotic infection of SRV-4 to humans, here we use a human hematopoietic stem cell-transplanted humanized mouse model. Eight out of the twelve SRV-4-inoculated humanized mice were infected with SRV-4. Importantly, 3 out of the 8 infected mice exhibited anemia and hemophagocytosis, and an infected mouse died. To address the possibility that SRV-4 adapts humanized mouse and acquires higher pathogenicity, the virus was isolated from an infected mice exhibited severe anemia was further inoculated into another 6 humanized mice. However, no infected mice exhibited any illness. Taken together, our findings demonstrate that the zoonotic SRV-4 infection from Japanese macaques to humans is technically possible under experimental condition. However, such zoonotic infection may not occur in the real society.


Subject(s)
Retroviridae Infections/transmission , Retroviruses, Simian/pathogenicity , Zoonoses/transmission , APOBEC Deaminases , Animals , Cell Line , Cytidine Deaminase , Cytokines/metabolism , Cytosine Deaminase/metabolism , Female , HEK293 Cells , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/virology , Humans , Japan , Male , Mice , Mice, Inbred NOD , Mice, Transgenic , Models, Animal , Polymerase Chain Reaction , RNA, Viral/analysis , Retroviridae Infections/pathology , Retroviridae Infections/virology , Retroviruses, Simian/genetics , Retroviruses, Simian/isolation & purification , Transplantation, Heterologous , Zoonoses/virology
20.
Sci Rep ; 5: 12256, 2015 Jul 17.
Article in English | MEDLINE | ID: mdl-26184634

ABSTRACT

Among the four groups of HIV-1 (M, N, O, and P), HIV-1M alone is pandemic and has rapidly expanded across the world. However, why HIV-1M has caused a devastating pandemic while the other groups remain contained is unclear. Interestingly, only HIV-1M Vpu, a viral protein, can robustly counteract human tetherin, which tethers budding virions. Therefore, we hypothesize that this property of HIV-1M Vpu facilitates human-to-human viral transmission. Adopting a multilayered experimental-mathematical approach, we demonstrate that HIV-1M Vpu confers a 2.38-fold increase in the prevalence of HIV-1 transmission. When Vpu activity is lost, protected human populations emerge (i.e., intrinsic herd immunity develops) through the anti-viral effect of tetherin. We also reveal that all Vpus of transmitted/founder HIV-1M viruses maintain anti-tetherin activity. These findings indicate that tetherin plays the role of a host restriction factor, providing 'intrinsic herd immunity', whereas Vpu has evolved in HIV-1M as a tetherin antagonist.


Subject(s)
Antigens, CD/metabolism , HIV Infections/immunology , HIV Infections/metabolism , HIV-1/immunology , Human Immunodeficiency Virus Proteins/immunology , Immunity, Herd , Viral Regulatory and Accessory Proteins/immunology , Algorithms , Amino Acid Motifs , Cell Line , Conserved Sequence , GPI-Linked Proteins/metabolism , HIV Infections/epidemiology , HIV Infections/transmission , HIV Infections/virology , HIV-1/metabolism , Human Immunodeficiency Virus Proteins/chemistry , Human Immunodeficiency Virus Proteins/metabolism , Humans , Models, Theoretical , Pandemics , Position-Specific Scoring Matrices , Protein Binding , Viral Load , Viral Regulatory and Accessory Proteins/chemistry , Viral Regulatory and Accessory Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...