Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Viruses ; 10(4)2018 04 10.
Article in English | MEDLINE | ID: mdl-29642583

ABSTRACT

The replication of lentiviruses highly depends on host cellular factors, which defines their species-specific tropism. Cellular restriction factors that can inhibit lentiviral replication were recently identified. Feline immunodeficiency virus (FIV) was found to be sensitive to several feline cellular restriction factors, such as apolipoprotein B mRNA-editing enzyme, catalytic polypeptide-like 3 (APOBEC3) and tetherin, but FIV evolved to counteract them. Here, we describe the molecular mechanisms by which feline APOBEC3 restriction factors inhibit FIV replication and discuss the molecular interaction of APOBEC3 proteins with the viral antagonizing protein Vif. We speculate that feline APOBEC3 proteins could explain some of the observed FIV cross-species transmissions described in wild Felids.


Subject(s)
APOBEC Deaminases/metabolism , Cat Diseases/transmission , Immunodeficiency Virus, Feline/physiology , Lentivirus Infections/veterinary , APOBEC Deaminases/genetics , Animals , Cat Diseases/virology , Cats , Gene Products, vif/deficiency , Gene Products, vif/metabolism , Lentivirus Infections/transmission , Lentivirus Infections/virology , Models, Biological , Species Specificity , Virus Replication
2.
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
3.
J Virol ; 83(11): 5956-60, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19297501

ABSTRACT

APOBEC3G restricts Vif-deficient human immunodeficiency virus type 1 (HIV-1) by deaminating viral cDNA cytosines to uracils. This promutagenic activity is counteracted by HIV-1 Vif, which is a natural APOBEC3G antagonist. However, we previously reported that Vif-deficient HIV-1 could evolve resistance to APOBEC3G by a novel mechanism requiring an A200-to-C/T transition mutation and Vpr inactivation. A pyrimidine at nucleotide 200 in the untranslated leader region contributed to resistance by increasing virus particle production, which resulted in fewer APOBEC3G molecules per particle. Here we show that the A200-to-C/T mutation functions posttranscriptionally by inactivating an upstream start codon, which in turn enables optimal viral mRNA translation from canonical start codons.


Subject(s)
Cytidine Deaminase/metabolism , Gene Products, vif/deficiency , HIV-1/metabolism , Protein Biosynthesis/genetics , APOBEC-3G Deaminase , Base Sequence , Cytidine Deaminase/genetics , Gene Products, vif/genetics , Genotype , HIV-1/genetics , Humans , Kinetics , Molecular Sequence Data
4.
J Gen Virol ; 88(Pt 10): 2793-2799, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17872533

ABSTRACT

This report characterizes lentivirus attenuation associated with a vif mutation by inoculation of newborn kittens with a vif-deleted feline immunodeficiency virus provirus plasmid (FIV-pPPRDeltavif). Virus in peripheral blood, antiviral antibody or CD4 T-cell count alterations were not detected in kittens inoculated with FIV-pPPRDeltavif plasmid, with the exception of one kitten that demonstrated FIV Gag antibody production at 42 weeks after inoculation. In contrast, wild-type FIV-pPPR-infected kittens were viraemic, seropositive and exhibited a decrease in the CD4 T-cell subset in peripheral blood. Interestingly, FIV-specific T-cell proliferative responses detected at 32 and 36 weeks after infection were comparable for both FIV-pPPRDeltavif- and wild-type FIV-pPPR-inoculated kittens and suggested the possibility of a discreet tissue reservoir supporting sustained FIV-pPPRDeltavif expression or replication. Overall, these findings confirmed that the severe virus attenuation for both replication and pathogenicity exhibited by a vif-deleted FIV mutant is similar for both neonatal and adult hosts.


Subject(s)
Cats/virology , Feline Acquired Immunodeficiency Syndrome/prevention & control , Gene Deletion , Gene Products, vif/deficiency , Gene Products, vif/genetics , Immunodeficiency Virus, Feline/genetics , Animals , Animals, Newborn , Antibodies, Viral/blood , CD4-Positive T-Lymphocytes/immunology , Feline Acquired Immunodeficiency Syndrome/immunology , Genome, Viral , Immunodeficiency Virus, Feline/physiology , Mutation , Virus Replication
5.
J Virol ; 81(13): 7099-110, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17428871

ABSTRACT

Encapsidation of host restriction factor APOBEC3G (A3G) into vif-deficient human immunodeficiency virus type 1 (HIV-1) blocks virus replication at least partly by C-to-U deamination of viral minus-strand DNA, resulting in G-to-A hypermutation. A3G may also inhibit HIV-1 replication by reducing viral DNA synthesis and inducing viral DNA degradation. To gain further insight into the mechanisms of viral inhibition, we examined the metabolism of A3G-exposed viral DNA. We observed that an overall 35-fold decrease in viral infectivity was accompanied by a five- to sevenfold reduction in viral DNA synthesis. Wild-type A3G induced an additional fivefold decrease in the amount of viral DNA that was integrated into the host cell genome and similarly reduced the efficiency with which HIV-1 preintegration complexes (PICs) integrated into a target DNA in vitro. The A3G C-terminal catalytic domain was required for both of these antiviral activities. Southern blotting analysis of PICs showed that A3G reduced the efficiency and specificity of primer tRNA processing and removal, resulting in viral DNA ends that are inefficient substrates for integration and plus-strand DNA transfer. However, the decrease in plus-strand DNA transfer did not account for all of the observed decrease in viral DNA synthesis associated with A3G. These novel observations suggest that HIV-1 cDNA produced in the presence of A3G exhibits defects in primer tRNA processing, plus-strand DNA transfer, and integration.


Subject(s)
DNA, Complementary/metabolism , DNA, Viral/metabolism , HIV-1/physiology , Nucleoside Deaminases/metabolism , Repressor Proteins/metabolism , Virus Integration/physiology , Virus Replication/physiology , APOBEC-3G Deaminase , Catalytic Domain/genetics , Cell Line , Cytidine Deaminase , DNA, Complementary/genetics , Gene Products, vif/deficiency , Gene Products, vif/metabolism , Genome, Human , Humans , RNA Processing, Post-Transcriptional/physiology , RNA, Transfer/metabolism , vif Gene Products, Human Immunodeficiency Virus
6.
Virology ; 351(1): 80-91, 2006 Jul 20.
Article in English | MEDLINE | ID: mdl-16631224

ABSTRACT

Reverse transcription (RTn) in HIV-infected cells occurs in a nucleoprotein complex termed the reverse transcription complex (RTC). RTCs containing RT activity and integrase (IN) were shown to be heterogeneous in size and density on sucrose velocity and equilibrium gradients. WT and Vif-deficient (Deltavif) RTCs produced by infection with virus from permissive cells displayed similar sedimentation characteristics, while RTCs from Deltavif virus produced in non-permissive cells demonstrated a reduction in the major RTC form and more of the RTn products in rapidly sedimenting structures. APOBEC3G derived from virions did not co-sediment with RTCs, but RTCs from Deltavif infections showed elevated levels of mutations in RTn products, consistent with APOBEC3G and other mutational mechanisms. The most mutated transcripts were present within rapidly sedimenting RTCs. Thus, virus without functional vif, produced from non-permissive cells, forms abnormal RTCs that contain increased mutation of RTC-associated RTn products in newly infected target cells.


Subject(s)
Gene Products, vif/deficiency , HIV Reverse Transcriptase/chemistry , HIV Reverse Transcriptase/metabolism , Mutation/genetics , Reverse Transcription , APOBEC-3G Deaminase , Cell Line , Cell-Free System , Cytidine Deaminase , Gene Products, vif/metabolism , Humans , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Mutagenesis , Nucleoside Deaminases/metabolism , Repressor Proteins/metabolism
8.
Nature ; 424(6944): 94-8, 2003 Jul 03.
Article in English | MEDLINE | ID: mdl-12808465

ABSTRACT

High mutation frequency during reverse transcription has a principal role in the genetic variation of primate lentiviral populations. It is the main driving force for the generation of drug resistance and the escape from immune surveillance. G to A hypermutation is one of the characteristics of primate lentiviruses, as well as other retroviruses, during replication in vivo and in cell culture. The molecular mechanisms of this process, however, remain to be clarified. Here, we demonstrate that CEM15 (also known as apolipoprotein B mRNA editing enzyme, catalytic polypeptide-like 3G; APOBEC3G), an endogenous inhibitor of human immunodeficiency virus type 1 (HIV-1) replication, is a cytidine deaminase and is able to induce G to A hypermutation in newly synthesized viral DNA. This effect can be counteracted by the HIV-1 virion infectivity factor (Vif). It seems that this viral DNA mutator is a viral defence mechanism in host cells that may induce either lethal hypermutation or instability of the incoming nascent viral reverse transcripts, which could account for the Vif-defective phenotype. Importantly, the accumulation of CEM15-mediated non-lethal hypermutation in the replicating viral genome could potently contribute to the genetic variation of primate lentiviral populations.


Subject(s)
DNA, Viral/biosynthesis , DNA, Viral/genetics , HIV-1/genetics , Mutagenesis/genetics , Proteins/metabolism , APOBEC-3G Deaminase , Cell Line , Cytidine Deaminase , Gene Products, vif/deficiency , Gene Products, vif/genetics , Gene Products, vif/metabolism , HIV-1/growth & development , HIV-1/physiology , Humans , Nucleoside Deaminases , Point Mutation/genetics , Proteins/genetics , Repressor Proteins , Serial Passage , Transcription, Genetic/genetics , Virus Replication , vif Gene Products, Human Immunodeficiency Virus
9.
Nature ; 424(6944): 99-103, 2003 Jul 03.
Article in English | MEDLINE | ID: mdl-12808466

ABSTRACT

Viral replication usually requires that innate intracellular lines of defence be overcome, a task usually accomplished by specialized viral gene products. The virion infectivity factor (Vif) protein of human immunodeficiency virus (HIV) is required during the late stages of viral production to counter the antiviral activity of APOBEC3G (apolipoprotein B mRNA-editing enzyme, catalytic polypeptide-like 3G; also known as CEM15), a protein expressed notably in human T lymphocytes. When produced in the presence of APOBEC3G, vif-defective virus is non-infectious. APOBEC3G is closely related to APOBEC1, the central component of an RNA-editing complex that deaminates a cytosine residue in apoB messenger RNA. APOBEC family members also have potent DNA mutator activity through dC deamination; however, whether the editing potential of APOBEC3G has any relevance to HIV inhibition is unknown. Here, we demonstrate that it does, as APOBEC3G exerts its antiviral effect during reverse transcription to trigger G-to-A hypermutation in the nascent retroviral DNA. We also find that APOBEC3G can act on a broad range of retroviruses in addition to HIV, suggesting that hypermutation by editing is a general innate defence mechanism against this important group of pathogens.


Subject(s)
DNA, Viral/biosynthesis , DNA, Viral/genetics , HIV-1/genetics , Mutagenesis/genetics , Proteins/metabolism , Transcription, Genetic/genetics , APOBEC-3G Deaminase , Antiviral Agents/metabolism , Base Sequence , Cell Line , Cytidine Deaminase , Gene Products, vif/deficiency , Gene Products, vif/genetics , Gene Products, vif/metabolism , HIV-1/physiology , Humans , Molecular Sequence Data , Nucleoside Deaminases , Point Mutation/genetics , Proteins/genetics , RNA Editing/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Repressor Proteins , Viral Load , Virus Replication , vif Gene Products, Human Immunodeficiency Virus
10.
Cell ; 113(6): 803-9, 2003 Jun 13.
Article in English | MEDLINE | ID: mdl-12809610

ABSTRACT

CEM15/APOBEC3G is a cellular protein required for resistance to infection by virion infectivity factor (Vif)-deficient human immunodeficiency virus (HIV). Here, using a murine leukemia virus (MLV)-based system, we provide evidence that CEM15/APOBEC3G is a DNA deaminase that is incorporated into virions during viral production and subsequently triggers massive deamination of deoxycytidine to deoxyuridine within the retroviral minus (first)-strand cDNA, thus providing a probable trigger for viral destruction. Furthermore, HIV Vif can protect MLV from this CEM15/APOBEC3G-dependent restriction. These findings imply that targeted DNA deamination is a major strategy of innate immunity to retroviruses and likely also contributes to the sequence variation observed in many viruses (including HIV).


Subject(s)
DNA, Complementary/metabolism , Eukaryotic Cells/metabolism , HIV Infections/immunology , HIV/metabolism , Immunity, Innate/genetics , Proteins/metabolism , APOBEC-3G Deaminase , Animals , Base Sequence/genetics , Cell Line , Cytidine Deaminase , DNA Replication/genetics , DNA, Complementary/genetics , Deamination , Gene Products, vif/deficiency , Gene Products, vif/genetics , HIV/genetics , HIV/pathogenicity , HIV Infections/genetics , HIV Infections/metabolism , Humans , Leukemia Virus, Murine/genetics , Leukemia Virus, Murine/metabolism , Mice , Molecular Sequence Data , Mutation/genetics , Nucleoside Deaminases , Proteins/genetics , RNA, Viral/biosynthesis , RNA, Viral/genetics , Repressor Proteins , vif Gene Products, Human Immunodeficiency Virus
11.
J Virol ; 77(10): 5810-20, 2003 May.
Article in English | MEDLINE | ID: mdl-12719574

ABSTRACT

Replication of human immunodeficiency virus type 1 (HIV-1) in primary blood lymphocytes, certain T-cell lines (nonpermissive cells), and most likely in vivo is highly dependent on the virally encoded Vif protein. Evidence suggests that Vif acts late in the viral life cycle during assembly, budding, and/or maturation to counteract the antiviral activity of the CEM15 protein and possibly other antiviral factors. Because HIV-1 virions produced in the absence of Vif are severely restricted at a postentry, preintegration step of infection, it is presumed that such virions differ from wild-type virions in some way. In the present study, we established a protocol for producing large quantities of vif-deficient HIV-1 (HIV-1/Delta vif) from an acute infection of nonpermissive T cells and performed a thorough examination of the defect in these virions. Aside from the expected lack of Vif, we observed no apparent abnormalities in the packaging, modification, processing, or function of proteins in Delta vif virions. In addition, we found no consistent defect in the ability of Delta vif virions to perform intravirion reverse transcription under a variety of assay conditions, suggesting that the reverse transcription complexes in these particles can behave normally under cell-free conditions. Consistent with this finding, neither the placement of the primer tRNA3Lys nor its ability to promote reverse transcription in an in vitro assay was affected by a lack of Vif. Based on the inability of this comprehensive analysis to uncover molecular defects in Delta vif virions, we speculate that such defects are likely to be subtle and/or rare.


Subject(s)
Gene Products, vif/deficiency , HIV-1/genetics , HIV-1/pathogenicity , Virion/metabolism , Cell Line , Gene Products, vif/genetics , Gene Products, vif/metabolism , HIV Infections/virology , Humans , RNA, Transfer, Lys/metabolism , T-Lymphocytes/virology , Transcription, Genetic , Virion/genetics , Virus Replication , vif Gene Products, Human Immunodeficiency Virus
12.
Virology ; 282(1): 77-86, 2001 Mar 30.
Article in English | MEDLINE | ID: mdl-11259192

ABSTRACT

Lentiviral vectors based on human immunodeficiency virus type 1 (HIV-1) possess the ability to deliver exogenous genes to both dividing and nondividing cells and to subsequently establish a stable provirus in these target cells, which can allow long-term expression of the transferred gene. Herein we describe a stable packaging cell line that is devoid of HIV-1 tat, vif, vpr, vpu, and nef. In order to avoid any risk of cytotoxicity associated with constitutive expression of HIV-1 protease or the VSV-G envelope protein, transcription of the packaging and envelope constructs was tightly controlled by employing the ecdysone-inducible system. Using this cell line, we have been able to consistently generate concentrated pseudotyped vector virus stocks with titers in the range of 10(8) IU/ml, which can efficiently transduce actively dividing and growth-arrested cells in vitro. This novel packaging cell line for lentiviral vectors facilitates the production of high-titer virus stocks in the absence of replication-competent virus and provides us with an important tool for use in future gene transfer studies.


Subject(s)
Cell Line , Genetic Vectors , HIV-1/genetics , Lentivirus/genetics , Membrane Glycoproteins , Ecdysone/pharmacology , Gene Products, tat/deficiency , Gene Products, tat/genetics , Gene Products, vif/deficiency , Gene Products, vif/genetics , Gene Products, vpr/deficiency , Gene Products, vpr/genetics , Green Fluorescent Proteins , Human Immunodeficiency Virus Proteins , Humans , Luminescent Proteins/genetics , Transduction, Genetic , Transfection , Viral Envelope Proteins/genetics , Viral Proteins/genetics , Viral Regulatory and Accessory Proteins/deficiency , Viral Regulatory and Accessory Proteins/genetics , Virus Replication , tat Gene Products, Human Immunodeficiency Virus , vif Gene Products, Human Immunodeficiency Virus , vpr Gene Products, Human Immunodeficiency Virus
13.
Viral Immunol ; 14(4): 339-48, 2001.
Article in English | MEDLINE | ID: mdl-11792063

ABSTRACT

A versatile DNA vaccine (pdIV3) was constructed by replacing the integrase, vif, vpx, and vpr genes of a pathogenic simian immunodeficiency virus (SIV) molecular clone with a linker containing unique cloning sites. The 5' long terminal repeat (LTR) is truncated and transcription is controlled by a cytomegalovirus (CMV) promoter. The construct expresses Gag and Env in vitro and noninfectious virus particles are produced from transfected cells. The ability of pdIV3 to promote cellular and humoral immune responses, along with the flexibility of the linker design to allow insertion of immunostimulatory genes in future constructs, makes this a useful base vector for immunization against primate lentiviruses. We present the construction of a retroviral plasmid designed to serve as a template for the development of safe and effective vaccines against primate immunodeficiency retroviruses. This vaccine component should facilitate the simultaneous induction of cellular and humoral immune responses that protect primates against infection with SIV and human immunodeficiency virus (HIV) and the development of acquired immune deficiency syndrome (AIDS). This plasmid could induce the appropriate immune response required to attack both cell-free and cell-associated viruses. The lack of infectivity, the inability to integrate, and the SIV origin make this construct a safe alternative to attenuated vaccines based on HIV. In addition, we intend to develop this construct as an immunotherapeutic approach to lower the viremia in AIDS patients.


Subject(s)
Defective Viruses/immunology , Genetic Vectors/immunology , Simian Immunodeficiency Virus/immunology , Vaccines, DNA/immunology , Viral Vaccines/immunology , Animals , Antibodies, Viral/biosynthesis , COS Cells , Chlorocebus aethiops , Defective Viruses/isolation & purification , Gene Products, vif/deficiency , Gene Products, vif/genetics , Integrases/deficiency , Integrases/genetics , Leukocytes, Mononuclear/immunology , Lymphocyte Activation , Plasmids , Rabbits , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/isolation & purification , Viral Regulatory and Accessory Proteins/deficiency , Viral Regulatory and Accessory Proteins/genetics , Viral Vaccines/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...