Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
1.
Trends Genet ; 32(1): 16-28, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26608778

ABSTRACT

Information warfare is not limited to the cyber world because it is waged within our cells as well. The unique AID (activation-induced cytidine deaminase)/APOBEC (apolipoprotein B mRNA editing enzyme, catalytic polypeptide) family comprises proteins that alter DNA sequences by converting deoxycytidines to deoxyuridines through deamination. This C-to-U DNA editing enables them to inhibit parasitic viruses and retrotransposons by disrupting their genomic content. In addition to attacking genomic invaders, APOBECs can target their host genome, which can be beneficial by initiating processes that create antibody diversity needed for the immune system or by accelerating the rate of evolution. AID can also alter gene regulation by removing epigenetic modifications from genomic DNA. However, when uncontrolled, these powerful agents of change can threaten genome stability and eventually lead to cancer.


Subject(s)
APOBEC Deaminases/metabolism , DNA/metabolism , Epigenesis, Genetic , Immunity, Innate , Retroelements , APOBEC Deaminases/genetics , APOBEC Deaminases/immunology , Animals , Cytidine Deaminase/metabolism , Cytosine Deaminase/immunology , Cytosine Deaminase/metabolism , Evolution, Molecular , Genome , Humans
2.
Gastroenterology ; 155(1): 180-193.e6, 2018 07.
Article in English | MEDLINE | ID: mdl-29550589

ABSTRACT

BACKGROUND & AIMS: Strategies to develop virus-specific T cells against hepatic viral infections have been hindered by safety concerns. We engineered nonlytic human T cells to suppress replication of hepatitis B virus (HBV) and hepatitis C virus (HCV) without overt hepatotoxicity and investigated their antiviral activity. METHODS: We electroporated resting T cells or T cells activated by anti-CD3 with mRNAs encoding HBV or HCV-specific T-cell receptors (TCRs) to create 2 populations of TCR-reprogrammed T cells. We tested their ability to suppress HBV or HCV replication without lysis in 2-dimensional and 3-dimensional cultures of HepG2.2.15 cells and HBV-infected HepG2-hNTCP cells. We also injected TCR-reprogrammed resting and activated T cells into HBV-infected urokinase-type plasminogen activator/severe combined immunodeficiency disease/interleukin 2γ mice with humanized livers and measured levels of intrahepatic and serological viral parameters and serum alanine aminotransferase. Livers were collected for analysis of gene expression patterns to determine effects of the TCR-reprogrammed T cells. RESULTS: TCR-reprogrammed resting T cells produced comparable levels of interferon gamma but lower levels of perforin and granzyme than activated T cells and did not lyse HCV- or HBV-infected hepatoma cells. Although T-cell secretion of interferon gamma was required to inhibit HCV replication, the HBV-specific TCR-reprogrammed resting T cells reduced HBV replication also through intracellular activation of apolipoprotein B mRNA editing enzyme, catalytic polypeptide 3 (APOBEC3). The mechanism of APOBEC3 intracellular activation involved temporal expression of lymphotoxin-ß receptor ligands on resting T cells after TCR-mediated antigen recognition and activation of lymphotoxin-ß receptor in infected cells. CONCLUSIONS: We developed TCR-reprogrammed nonlytic T cells capable of activating APOBEC3 in hepatoma cells and in HBV-infected human hepatocytes in mice, limiting viral infection. These cells with limited hepatotoxicity might be developed for treatment of chronic HBV infection.


Subject(s)
Cytosine Deaminase/immunology , Hepacivirus/immunology , Hepatitis B virus/immunology , Hepatitis B, Chronic/therapy , Liver/metabolism , Lymphocyte Activation/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , APOBEC Deaminases , Animals , Cytidine Deaminase , Electroporation , Hep G2 Cells , Hepatocytes , Humans , Interferon-gamma/immunology , Mice , Mice, SCID , RNA, Messenger , RNA, Viral , Receptors, Antigen, T-Cell/genetics
3.
Biochem Biophys Res Commun ; 503(3): 2124-2131, 2018 09 10.
Article in English | MEDLINE | ID: mdl-30100060

ABSTRACT

OBJECTIVE: This study aimed to evaluate functions of APOBEC3F gene in biological process of hepatocellular carcinoma (HCC) and anti-tumor mechanisms of bufalin. METHODS: Effect of APOBEC3F and bufalin on cell proliferation and migration abilities were evaluated by CCK-8, wounding healing tests and transwell assays in SK-Hep1 and Bel-7404 cells. Bioinformatic analysis were also used to compare APOBEC3F expression levels, detect coexpressed genes and enrichment of pathways. RESULTS: APOBEC3F was overexpressed in tumor tissues compared to adjacent tissues in HCC patients. And, APOBEC3F promotes cell proliferation and migration in SK-Hep1 and Bel-7404 cells. Bufalin inhibits cell proliferation and migration and reduces APOBEC3F expression. GO and KEGG enrichment of APOBEC3F-coexpressed genes revealed that APOBEC3F might active intestinal immune network for IgA production signaling pathway, leading to malignant biological behaviors of HCC cells. Additionally, siAPOBEC3F could decrease pIgR, CCR9, CCR10 and CXCR4 protein levels. And, bufalin inhibits the pIgR, CCR9, CCR10 and CXCR4 protein expressions. CONCLUSIONS: Bufalin inhibits cell proliferation and migration of HCC cells via APOBEC3F induced intestinal immune network for IgA production signaling pathway.


Subject(s)
Bufanolides/pharmacology , Carcinoma, Hepatocellular/drug therapy , Cell Movement/drug effects , Cytosine Deaminase/biosynthesis , Immunoglobulin A/immunology , Intestinal Mucosa/drug effects , Liver Neoplasms/drug therapy , Signal Transduction/drug effects , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cytosine Deaminase/immunology , Cytosine Deaminase/metabolism , Dose-Response Relationship, Drug , Humans , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Signal Transduction/immunology , Structure-Activity Relationship
4.
J Immunol ; 195(10): 4565-70, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26546688

ABSTRACT

Apolipoprotein B editing complex 3 family members are cytidine deaminases that play important roles in intrinsic responses to infection by retroviruses and have been implicated in the control of other viruses, such as parvoviruses, herpesviruses, papillomaviruses, hepatitis B virus, and retrotransposons. Although their direct effect on modification of viral DNA has been clearly demonstrated, whether they play additional roles in innate and adaptive immunity to viruses is less clear. We review the data regarding the various steps in the innate and adaptive immune response to virus infection in which apolipoprotein B editing complex 3 proteins have been implicated.


Subject(s)
Cytosine Deaminase/immunology , DNA, Viral/immunology , HIV Infections/immunology , HIV-1/immunology , Virus Diseases/immunology , APOBEC Deaminases , Adaptive Immunity/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cytidine Deaminase , Cytosine Deaminase/genetics , DNA, Viral/genetics , HIV Infections/virology , Hepatitis B/immunology , Herpesviridae Infections/immunology , Herpesviridae Infections/virology , Humans , Papillomavirus Infections/immunology , Parvoviridae Infections/immunology , Parvoviridae Infections/virology
5.
Adv Exp Med Biol ; 962: 415-431, 2017.
Article in English | MEDLINE | ID: mdl-28299671

ABSTRACT

In order to achieve a persistent infection, viruses must overcome the host immune system. Host restriction factors dominantly block virus transmission, but are subject to down regulation by viral accessory proteins. HIV encodes several accessory factors that overcome different cellular restriction factors. For example, the HIV-1 protein Vif down regulates the human APOBEC3 family of restriction factors by targeting them for proteolysis by the ubiquitin-proteasome pathway. Recently, this function was shown to require the transcription cofactor CBFß, which acts as a template to assist in Vif folding and allow for assembly of an APOBEC3-targeting E3 ligase complex. In uninfected cells, CBFß is an essential binding partner of RUNX transcription factors. By binding CBFß, Vif has also been shown to perturb transcription of genes regulated by the RUNX proteins, including restrictive APOBEC3 family members. Here we review how the link between CBFß and Vif supports transcriptional and post-transcriptional repression of innate immunity. The ability of a single viral protein to coopt multiple host pathways is an economical strategy for a pathogen with limited protein coding capacity to achieve a productive infection.


Subject(s)
Core Binding Factor beta Subunit/metabolism , HIV Infections/metabolism , HIV Infections/virology , HIV-1/metabolism , vif Gene Products, Human Immunodeficiency Virus/metabolism , Animals , Core Binding Factor beta Subunit/immunology , Cytosine Deaminase/immunology , Cytosine Deaminase/metabolism , HIV Infections/immunology , HIV-1/immunology , Host-Pathogen Interactions/immunology , Host-Pathogen Interactions/physiology , Humans , Immunity, Innate/immunology , Transcription, Genetic/immunology , Transcription, Genetic/physiology , vif Gene Products, Human Immunodeficiency Virus/immunology
6.
Eur J Immunol ; 45(3): 695-704, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25501566

ABSTRACT

In general, a long-lasting immune response to viruses is achieved when they are infectious and replication competent. In the mouse, the neutralizing antibody response to Friend murine leukemia virus is contributed by an allelic form of the enzyme Apobec3 (abbreviated A3). This is counterintuitive because A3 directly controls viremia before the onset of adaptive antiviral immune responses. It suggests that A3 also affects the antibody response directly. Here, we studied the relative size of cell populations of the adaptive immune system as a function of A3 activity. We created a transgenic mouse that expresses all seven human A3 enzymes and compared it to WT and mouse A3-deficient mice. A3 enzymes decreased the number of marginal zone B cells, but not the number of follicular B or T cells. When mouse A3 was knocked out, the retroelement hitchhiker-1 and sialyl transferases encoded by genes close to it were overexpressed three and two orders of magnitude, respectively. We suggest that A3 shifts the balance, from the fast antibody response mediated by marginal zone B cells with little affinity maturation, to a more sustained germinal center B-cell response, which drives affinity maturation and, thereby, a better neutralizing response.


Subject(s)
Antibody Formation , B-Lymphocytes/immunology , Cytidine Deaminase/immunology , Cytosine Deaminase/immunology , Germinal Center/immunology , APOBEC Deaminases , Animals , Cytidine Deaminase/genetics , Cytosine Deaminase/genetics , Humans , Mice , Mice, Inbred BALB C , Mice, Knockout , Virus Diseases/genetics , Virus Diseases/immunology , Virus Diseases/pathology
7.
J Virol ; 89(4): 2415-24, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25505075

ABSTRACT

UNLABELLED: Members of the APOBEC3 family of cytidine deaminases vary in their proportions of a virion-incorporated enzyme that is localized to mature retrovirus cores. We reported previously that APOBEC3F (A3F) was highly localized into mature human immunodeficiency virus type 1 (HIV-1) cores and identified that L306 in the C-terminal cytidine deaminase (CD) domain contributed to its core localization (C. Song, L. Sutton, M. Johnson, R. D'Aquila, J. Donahue, J Biol Chem 287:16965-16974, 2012, http://dx.doi.org/10.1074/jbc.M111.310839). We have now determined an additional genetic determinant(s) for A3F localization to HIV-1 cores. We found that one pair of leucines in each of A3F's C-terminal and N-terminal CD domains jointly determined the degree of localization of A3F into HIV-1 virion cores. These are A3F L306/L368 (C-terminal domain) and A3F L122/L184 (N-terminal domain). Alterations to one of these specific leucine residues in either of the two A3F CD domains (A3F L368A, L122A, and L184A) decreased core localization and diminished HIV restriction without changing virion packaging. Furthermore, double mutants in these leucine residues in each of A3F's two CD domains (A3F L368A plus L184A or A3F L368A plus L122A) still were packaged into virions but completely lost core localization and anti-HIV activity. HIV virion core localization of A3F is genetically separable from its virion packaging, and anti-HIV activity requires some core localization. IMPORTANCE: Specific leucine-leucine interactions are identified as necessary for A3F's core localization and anti-HIV activity but not for its packaging into virions. Understanding these signals may lead to novel strategies to enhance core localization that may augment effects of A3F against HIV and perhaps of other A3s against retroviruses, parvoviruses, and hepatitis B virus.


Subject(s)
Cytosine Deaminase/analysis , Cytosine Deaminase/genetics , HIV-1/physiology , Virus Assembly , Cell Line , Cytosine Deaminase/immunology , DNA Mutational Analysis , Genes, Reporter , HIV-1/chemistry , HIV-1/immunology , Humans , Luciferases/analysis , Models, Molecular , Mutagenesis, Site-Directed , Mutation, Missense , Staining and Labeling , beta-Galactosidase/analysis
8.
PLoS Pathog ; 10(3): e1004024, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24651717

ABSTRACT

The APOBEC3 deoxycytidine deaminase family functions as host restriction factors that can block replication of Vif (virus infectivity factor) deficient HIV-1 virions to differing degrees by deaminating cytosines to uracils in single-stranded (-)HIV-1 DNA. Upon replication of the (-)DNA to (+)DNA, the HIV-1 reverse transcriptase incorporates adenines opposite the uracils, thereby inducing C/G→T/A mutations that can functionally inactivate HIV-1. Although both APOBEC3F and APOBEC3G are expressed in cell types HIV-1 infects and are suppressed by Vif, there has been no prior biochemical analysis of APOBEC3F, in contrast to APOBEC3G. Using synthetic DNA substrates, we characterized APOBEC3F and found that similar to APOBEC3G; it is a processive enzyme and can deaminate at least two cytosines in a single enzyme-substrate encounter. However, APOBEC3F scanning movement is distinct from APOBEC3G, and relies on jumping rather than both jumping and sliding. APOBEC3F jumping movements were also different from APOBEC3G. The lack of sliding movement from APOBEC3F is due to an ¹9°NPM¹9² motif, since insertion of this motif into APOBEC3G decreases its sliding movements. The APOBEC3G NPM mutant induced significantly less mutations in comparison to wild-type APOBEC3G in an in vitro model HIV-1 replication assay and single-cycle infectivity assay, indicating that differences in DNA scanning were relevant to restriction of HIV-1. Conversely, mutation of the APOBEC3F ¹9¹Pro to ¹9¹Gly enables APOBEC3F sliding movements to occur. Although APOBEC3F ¹9°NGM¹9² could slide, the enzyme did not induce more mutagenesis than wild-type APOBEC3F, demonstrating that the unique jumping mechanism of APOBEC3F abrogates the influence of sliding on mutagenesis. Overall, we demonstrate key differences in the impact of APOBEC3F- and APOBEC3G-induced mutagenesis on HIV-1 that supports a model in which both the processive DNA scanning mechanism and preferred deamination motif (APOBEC3F, 5'TTC; APOBEC3G 5'CCC) influences the mutagenic and gene inactivation potential of an APOBEC3 enzyme.


Subject(s)
Cytidine Deaminase/metabolism , Cytosine Deaminase/metabolism , Gene Silencing/physiology , HIV-1/genetics , APOBEC-3G Deaminase , Amino Acid Motifs , Animals , Anisotropy , Cell Line , Chromatography, Gel , Cytidine Deaminase/immunology , Cytosine Deaminase/immunology , DNA, Single-Stranded/genetics , DNA, Viral/genetics , HIV-1/immunology , Humans , Immunoblotting , Mutagenesis/physiology
9.
J Virol ; 88(24): 14380-95, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25275135

ABSTRACT

UNLABELLED: The APOBEC3 deoxycytidine deaminases can restrict the replication of HIV-1 in cell culture to differing degrees. The effects of APOBEC3 enzymes are largely suppressed by HIV-1 Vif that interacts with host proteins to form a Cullin5-Ring E3 ubiquitin ligase that induces (48)K-linked polyubiquitination (poly-Ub) and proteasomal degradation of APOBEC3 enzymes. Vif variants have differing abilities to induce degradation of APOBEC3 enzymes and the underlying biochemical mechanisms for these differences is not fully understood. We hypothesized that by characterizing the interaction of multiple APOBEC3 enzymes and Vif variants we could identify common features that resulted in Vif-mediated degradation and further define the determinants required for efficient Vif-mediated degradation of APOBEC3 enzymes. We used Vifs from HIV-1 NL4-3 (IIIB) and HXB2 to characterize their induced degradation of and interaction with APOBEC3G, APOBEC3G D128K, APOBEC3H, and APOBEC3B in 293T cells. We quantified the APOBEC3G-Vif and APOBEC3H-Vif interaction strengths in vitro using rotational anisotropy. Our biochemical and cellular analyses of the interactions support a model in which the degradation efficiency of VifIIIB and VifHXB2 correlated with both the binding strength of the APOBEC3-Vif interaction and the APOBEC3-Vif interface, which differs for APOBEC3G and APOBEC3H. Notably, Vif bound to APOBEC3H and APOBEC3B in the natural absence of Vif-induced degradation and the interaction resulted in (63)K-linked poly-Ub of APOBEC3H and APOBEC3B, demonstrating additional functionality of the APOBEC3-Vif interaction apart from induction of proteasomal degradation. IMPORTANCE: APOBEC3 enzymes can potently restrict the replication of HIV-1 in the absence of HIV-1 Vif. Vif suppresses APOBEC3 action by inducing their degradation through a direct interaction with APOBEC3 enzymes and other host proteins. Vif variants from different HIV-1 strains have different effects on APOBEC3 enzymes. We used differing Vif degradation capacities of two Vif variants and various APOBEC3 enzymes with differential sensitivities to Vif to delineate determinants of the APOBEC3-Vif interaction that are required for inducing efficient degradation. Using a combined biochemical and cellular approach we identified that the strength of the APOBEC3-Vif binding interaction and the APOBEC3-Vif interface are determinants for degradation efficiency. Our results highlight the importance of using Vif variants with different degradation potential when delineating mechanisms of Vif-induced APOBEC3 degradation and identify features important for consideration in the development of HIV-1 therapies that disrupt the APOBEC3-Vif interaction.


Subject(s)
Cytosine Deaminase/antagonists & inhibitors , HIV-1/physiology , Host-Pathogen Interactions , vif Gene Products, Human Immunodeficiency Virus/metabolism , APOBEC Deaminases , Cell Line , Cytidine Deaminase , Cytosine Deaminase/immunology , Cytosine Deaminase/metabolism , HIV-1/immunology , Humans , Protein Binding , Proteolysis , vif Gene Products, Human Immunodeficiency Virus/immunology
10.
Arch Virol ; 160(8): 1901-8, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26016442

ABSTRACT

Pigs are considered to be suitable xenotransplantation organ donors. However, the risk of pathogen transmission from pigs to humans is a major concern in the transplantation of porcine tissues. The porcine endogenous retroviruses (PERVs) PERV-A, PERV-A/C, and PERV-B can infect human cells, but PERV-C is an ecotropic virus infecting only pig cells. Thus, several strategies have been proposed to reduce PERV transmission in xenograft recipients. Human APOBEC3G (huA3G) is a single-strand DNA cytosine deaminase, which inactivates the coding capacity of the virus by deamination of cDNA cytosines to uracils. This reaction occurs within the (-) DNA strand during reverse transcription, resulting in a G-to-A mutation in the (+) strand. While recent data have shown that PERV-B is severely inhibited by huA3G and porcine A3Z2-Z3 (poA3F) in a pseudotype assay, little is known about PERV-C. Here, we compare the antiretroviral activities of huA3G, huA3F and poA3Z2-Z3 against PERV-C. Our data show that APOBEC3 was packaged into PERV-C particles and inhibited PERV-C replication in a dose-dependent manner. PERV-C infectivity was strongly inhibited by poA3Z2-Z3, but it did not markedly reduce PERV-B infectivity. This suggests that PERV-C Gag interacts efficiently with poA3Z2-Z3. In addition, we constructed stably huA3G- and poA3Z2-Z3-expressing 293-PERV-PK-CIRCE cells (human 293 cells infected with PK15-derived PERVs) to examine whether PERV is resistant to poA3Z2-Z3 in a virus-spreading assay. The stably expressed huA3G and poA3Z2-Z3 were more packaging-competent than transiently expressed APOBEC3 proteins. These results suggest that poA3Z2-Z3 can inhibit PERV replication in a pseudotype assay as well as in a virus-spreading assay.


Subject(s)
Cytidine Deaminase/immunology , Cytosine Deaminase/immunology , Endogenous Retroviruses/immunology , Retroviridae Infections/enzymology , Retroviridae/immunology , Swine/immunology , Zoonoses/enzymology , APOBEC-3G Deaminase , Animals , Cytidine Deaminase/genetics , Cytosine Deaminase/genetics , Endogenous Retroviruses/classification , Endogenous Retroviruses/genetics , Endogenous Retroviruses/physiology , Host-Pathogen Interactions , Humans , Retroviridae/classification , Retroviridae/genetics , Retroviridae/physiology , Retroviridae Infections/genetics , Retroviridae Infections/immunology , Retroviridae Infections/virology , Swine/genetics , Swine/virology , Transplantation, Heterologous , Zoonoses/genetics , Zoonoses/immunology , Zoonoses/virology
11.
Nat Rev Immunol ; 4(11): 868-77, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15516966

ABSTRACT

A powerful mechanism of vertebrate innate immunity has been discovered in the past year, in which APOBEC proteins inhibit retroviruses by deaminating cytosine residues in nascent retroviral cDNA. To thwart this cellular defence, HIV encodes Vif, a small protein that mediates APOBEC degradation. Therefore, the balance between APOBECs and Vif might be a crucial determinant of the outcome of retroviral infection. Vertebrates have up to 11 different APOBEC proteins, with primates having the most. APOBEC proteins include AID, a probable DNA mutator that is responsible for immunoglobulin-gene diversification, and APOBEC1, an RNA editor with antiretroviral activities. This APOBEC abundance might help to tip the balance in favour of cellular defences.


Subject(s)
Cytidine Deaminase/immunology , Retroviridae/immunology , APOBEC-1 Deaminase , APOBEC-3G Deaminase , Cytidine Deaminase/metabolism , Cytosine Deaminase/immunology , Cytosine Deaminase/metabolism , DNA/metabolism , Evolution, Molecular , Gene Products, vif/metabolism , Humans , Immunoglobulins/genetics , Nucleoside Deaminases , Phylogeny , Proteins/immunology , Proteins/metabolism , Repressor Proteins , Retroviridae/metabolism
12.
J Virol ; 87(11): 6073-80, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23536679

ABSTRACT

APOBEC3 proteins mediate potent antiretroviral activity by hypermutating the retroviral genome during reverse transcription. To counteract APOBEC3 and gain a replicative advantage, lentiviruses such as human immunodeficiency virus type 1 (HIV-1) and simian immunodeficiency virus (SIV) have evolved the Vif protein, which targets APOBEC3 proteins for proteasomal degradation. However, the proteasome plays a critical role in the generation of T cell peptide epitopes. Whether Vif-mediated destruction of APOBEC3 proteins leads to the generation and presentation of APOBEC3-derived T cell epitopes on the surfaces of lentivirus-infected cells remains unknown. Here, using peptides derived from multiple Vif-sensitive APOBEC3 proteins, we identified APOBEC3-specific T cell responses in both HIV-1-infected patients and SIV-infected rhesus macaques. These results raise the possibility that these T cell responses may be part of the larger antiretroviral immune response.


Subject(s)
CD8-Positive T-Lymphocytes/virology , Cytidine Deaminase/immunology , Cytosine Deaminase/immunology , HIV Infections/enzymology , HIV-1/physiology , Simian Acquired Immunodeficiency Syndrome/enzymology , Simian Immunodeficiency Virus/physiology , APOBEC-3G Deaminase , Adult , Animals , CD8-Positive T-Lymphocytes/immunology , Cytidine Deaminase/genetics , Cytosine Deaminase/genetics , Female , Gene Products, vif/genetics , Gene Products, vif/immunology , HIV Infections/immunology , HIV Infections/virology , HIV-1/genetics , HIV-1/immunology , Humans , Macaca mulatta , Male , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/immunology
13.
Curr Top Microbiol Immunol ; 371: 1-27, 2013.
Article in English | MEDLINE | ID: mdl-23686230

ABSTRACT

The ability to regulate and even target mutagenesis is an extremely valuable cellular asset. Enzyme-catalyzed DNA cytosine deamination is a molecular strategy employed by vertebrates to promote antibody diversity and defend against foreign nucleic acids. Ten years ago, a family of cellular enzymes was first described with several proving capable of deaminating DNA and inhibiting HIV-1 replication. Ensuing studies on the apolipoprotein B mRNA-editing enzyme catalytic polypeptide-like 3 (APOBEC3) restriction factors have uncovered a broad-spectrum innate defense network that suppresses the replication of numerous endogenous and exogenous DNA-based parasites. Although many viruses possess equally elaborate counter-defense mechanisms, the APOBEC3 enzymes offer a tantalizing possibility of leveraging innate immunity to fend off viral infection. Here, we focus on mechanisms of retroelement restriction by the APOBEC3 family of restriction enzymes, and we consider the therapeutic benefits, as well as the possible pathological consequences, of arming cells with active DNA deaminases.


Subject(s)
Cytosine Deaminase/immunology , Immunity, Innate , Animals , Antibody Diversity/physiology , HIV-1/physiology , Humans , Mutagenesis/physiology
14.
Retrovirology ; 10: 76, 2013 Jul 24.
Article in English | MEDLINE | ID: mdl-23880220

ABSTRACT

BACKGROUND: APOBEC3 (A3) proteins restrict viral replication by cytidine deamination of viral DNA genomes and impairing reverse transcription and integration. To escape this restriction, lentiviruses have evolved the viral infectivity factor (Vif), which binds A3 proteins and targets them for proteolytic degradation. In contrast, foamy viruses (FVs) encode Bet proteins that allow replication in the presence of A3, apparently by A3 binding and/or sequestration, thus preventing A3 packaging into virions and subsequent restriction. Due to a long-lasting FV-host coevolution, Bet proteins mainly counteract restriction by A3s from their cognate or highly related host species. RESULTS: Through bioinformatics, we identified conserved motifs in Bet, all localized in the bel2 exon. In line with the localization of these conserved motifs within bel2, this part of feline FV (FFV) Bet has been shown to be essential for feline A3 (feA3) inactivation and feA3 protein binding. To study the function of the Bet motifs in detail, we analyzed the ability of targeted deletion, substitution, and chimeric FFV-PFV (prototype FV) Bet mutants to physically bind and/or inactivate feA3. Binding of Bet to feA3Z2b is sensitive to mutations in the first three conserved motifs and N- and C-terminal deletions and substitutions across almost the complete bel2 coding sequence. In contrast, the Bel1 (also designated Tas) domain of Bet is dispensable for basal feA3Z2b inactivation and binding but mainly increases the steady state level of Bet. Studies with PFV Bel1 and full-length FFV Bel2 chimeras confirmed the importance of Bel2 for A3 inactivation indicating that Bel1 is dispensable for basal feA3Z2b inactivation and binding but increases Bet stability. Moreover, the bel1/tas exon may be required for expression of a fully functional Bet protein from a spliced transcript. CONCLUSIONS: We show that the Bel2 domain of FV Bet is essential for the inactivation of APOBEC3 cytidine deaminase restriction factors. The Bel1/Tas domain increases protein stability and can be exchanged by related sequence. Since feA3 binding and inactivation by Bet are highly correlated, the data support the view that FV Bet prevents A3-mediated restriction of viral replication by creating strong complexes with these proteins.


Subject(s)
Cytosine Deaminase/immunology , Cytosine Deaminase/metabolism , Protein Interaction Domains and Motifs , Retroviridae Proteins/immunology , Retroviridae Proteins/metabolism , Spumavirus/physiology , Animals , Cats , Cell Line , Protein Binding , Spumavirus/immunology
15.
J Virol ; 86(21): 11712-24, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22915799

ABSTRACT

The APOBEC3 cytidine deaminases play a critical role in host-mediated defense against exogenous viruses, most notably, human immunodeficiency virus type-1 (HIV-1) and endogenous transposable elements. APOBEC3G and APOBEC3F interact with numerous proteins that regulate cellular RNA metabolism, including components of the RNA-induced silencing complex (RISC), and colocalize with a subset of these proteins to mRNA processing bodies (P bodies), which are sites of mRNA translational repression and decay. We sought to determine the role of P bodies and associated proteins in HIV-1 replication and APOBEC3 antiviral activity. While we established a positive correlation between APOBEC3 protein incorporation into virions and localization to P bodies, depletion of the P-body components DDX6 or Lsm1 did not affect HIV-1 replication, APOBEC3 packaging into virions or APOBEC3 protein mediated inhibition of HIV-1 infectivity. In addition, neither HIV-1 genomic RNA nor Gag colocalized with P-body proteins. However, simultaneous depletion of multiple Argonaute family members, the effector proteins of RISC, could modestly increase viral infectivity. Because some APOBEC3 proteins interact with several Argonaute proteins, we also tested whether they could modulate microRNA (miRNA) activity. We found no evidence for the specific regulation of miRNA function by the APOBEC3 proteins, though more general effects on transfected gene expression were observed. In sum, our results indicate that P bodies and certain associated proteins do not regulate HIV-1 replication or APOBEC3 protein antiviral activity. Localization to P bodies may therefore provide a means of sequestering APOBEC3 enzymatic activity away from cellular DNA or may be linked to as yet unidentified cellular functions.


Subject(s)
Cytosine Deaminase/immunology , Cytosine Deaminase/metabolism , HIV-1/immunology , HIV-1/physiology , Virus Replication , APOBEC Deaminases , Cell Line , Cytidine Deaminase , DEAD-box RNA Helicases/deficiency , DEAD-box RNA Helicases/metabolism , Humans , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/metabolism , RNA Stability , RNA-Binding Proteins/metabolism
16.
Retrovirology ; 9: 35, 2012 Apr 30.
Article in English | MEDLINE | ID: mdl-22546055

ABSTRACT

The enzyme APOBEC3G (A3G) mutates the human immunodeficiency virus (HIV) genome by converting deoxycytidine (dC) to deoxyuridine (dU) on minus strand viral DNA during reverse transcription. A3G restricts viral propagation by degrading or incapacitating the coding ability of the HIV genome. Thus, this enzyme has been perceived as an innate immune barrier to viral replication whilst adaptive immunity responses escalate to effective levels. The discovery of A3G less than a decade ago led to the promise of new anti-viral therapies based on manipulation of its cellular expression and/or activity. The rationale for therapeutic approaches has been solidified by demonstration of the effectiveness of A3G in diminishing viral replication in cell culture systems of HIV infection, reports of its mutational footprint in virions from patients, and recognition of its unusually robust enzymatic potential in biochemical studies in vitro. Despite its effectiveness in various experimental systems, numerous recent studies have shown that the ability of A3G to combat HIV in the physiological setting is severely limited. In fact, it has become apparent that its mutational activity may actually enhance viral fitness by accelerating HIV evolution towards the evasion of both anti-viral drugs and the immune system. This body of work suggests that the role of A3G in HIV infection is more complex than heretofore appreciated and supports the hypothesis that HIV has evolved to exploit the action of this host factor. Here we present an overview of recent data that bring to light historical overestimation of A3G's standing as a strictly anti-viral agent. We discuss the limitations of experimental systems used to assess its activities as well as caveats in data interpretation.


Subject(s)
Cytosine Deaminase/metabolism , Gene Expression Regulation, Viral , HIV Infections/drug therapy , HIV Infections/immunology , HIV/pathogenicity , APOBEC Deaminases , Adaptation, Biological , Adaptive Immunity , Animals , Anti-HIV Agents/pharmacology , Cytidine Deaminase , Cytosine Deaminase/genetics , Cytosine Deaminase/immunology , Drug Resistance, Viral , Evolution, Molecular , Genome, Viral , HIV/immunology , HIV/physiology , HIV Infections/virology , Humans , Immune Evasion , Mutation , Virus Replication
17.
J Virol ; 85(10): 4888-97, 2011 May.
Article in English | MEDLINE | ID: mdl-21325415

ABSTRACT

Xenotropic murine leukemia virus-related virus (XMRV) is a gammaretrovirus recently isolated from human prostate cancer and peripheral blood mononuclear cells (PBMCs) of patients with chronic fatigue syndrome (CFS). We and others have shown that host restriction factors APOBEC3G (A3G) and APOBEC3F (A3F), which are expressed in human PBMCs, inhibit XMRV in transient-transfection assays involving a single cycle of viral replication. However, the recovery of infectious XMRV from human PBMCs suggested that XMRV can replicate in these cells despite the expression of APOBEC3 proteins. To determine whether XMRV can replicate and spread in cultured PBMCs even though it can be inhibited by A3G/A3F, we infected phytohemagglutinin-activated human PBMCs and A3G/A3F-positive and -negative cell lines (CEM and CEM-SS, respectively) with different amounts of XMRV and monitored virus production by using quantitative real-time PCR. We found that XMRV efficiently replicated in CEM-SS cells and viral production increased by >4,000-fold, but there was only a modest increase in viral production from CEM cells (<14-fold) and a decrease in activated PBMCs, indicating little or no replication and spread of XMRV. However, infectious XMRV could be recovered from the infected PBMCs by cocultivation with a canine indicator cell line, and we observed hypermutation of XMRV genomes in PBMCs. Thus, PBMCs can potentially act as a source of infectious XMRV for spread to cells that express low levels of host restriction factors. Overall, these results suggest that hypermutation of XMRV in human PBMCs constitutes one of the blocks to replication and spread of XMRV. Furthermore, hypermutation of XMRV proviruses at GG dinucleotides may be a useful and reliable indicator of human PBMC infection.


Subject(s)
Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/virology , Virus Replication , Xenotropic murine leukemia virus-related virus/immunology , Xenotropic murine leukemia virus-related virus/pathogenicity , APOBEC-3G Deaminase , Cells, Cultured , Cytidine Deaminase/genetics , Cytidine Deaminase/immunology , Cytosine Deaminase/genetics , Cytosine Deaminase/immunology , Humans
18.
J Virol ; 85(17): 9139-46, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21697498

ABSTRACT

It is known that the human immune proteins APOBEC3G and -F (hA3G/F) can inhibit Vif-deficient HIV by G-to-A mutation; however, the roles of these enzymes in the evolution of HIV are debated. We argue that if evolutionary pressure from hA3G/F exists there should be evidence of their imprint on the HIV genome in the form of (i) underrepresentation of hA3G/F target motifs (e.g., TGGG [targeted position is underlined]) and overrepresentation of product motifs (e.g., TAGG) and/or (ii) an increase in the ratio of nonsynonymous to synonymous (NS/S) G-to-A changes among hA3G/F target motifs and a decrease of NS/S A-to-G changes among hA3G/F product motifs. To test the first hypothesis, we studied the representation of hA3G/F target and product motifs in 1,932 complete HIV-1 genomes using Markov models. We found that the highly targeted motifs are not underrepresented and their product motifs are not overrepresented. To test the second hypothesis, we determined the NS/S G↔A changes among the hA3G/F target and product motifs in 1,540 complete sets of nine HIV-1 genes. The NS/S changes did not show an increasing/decreasing trend within the target/product motifs, but the NS/S changes within the motif AG was exceptionally low. We observed the same pattern by analyzing 740 human genes. Given that hA3G/F do not act on the human genome, this suggests a small NS/S change within AG has arisen by other mechanisms. We therefore find no evidence of an evolutionary footprint of hA3G/F. We postulate several mechanisms to explain why the HIV-1 genome does not contain the hA3G/F footprint.


Subject(s)
Cytosine Deaminase/immunology , Evolution, Molecular , Genome, Viral , HIV-1/genetics , HIV-1/immunology , RNA, Viral/genetics , APOBEC Deaminases , Computational Biology/methods , Cytidine Deaminase , HIV Infections/immunology , HIV Infections/virology , Humans , Selection, Genetic
19.
J Virol ; 85(21): 11220-34, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21835787

ABSTRACT

Successful intracellular pathogens must evade or neutralize the innate immune defenses of their host cells and render the cellular environment permissive for replication. For example, to replicate efficiently in CD4(+) T lymphocytes, human immunodeficiency virus type 1 (HIV-1) encodes a protein called viral infectivity factor (Vif) that promotes pathogenesis by triggering the degradation of the retrovirus restriction factor APOBEC3G. Other APOBEC3 proteins have been implicated in HIV-1 restriction, but the relevant repertoire remains ambiguous. Here we present the first comprehensive analysis of the complete, seven-member human and rhesus APOBEC3 families in HIV-1 restriction. In addition to APOBEC3G, we find that three other human APOBEC3 proteins, APOBEC3D, APOBEC3F, and APOBEC3H, are all potent HIV-1 restriction factors. These four proteins are expressed in CD4(+) T lymphocytes, are packaged into and restrict Vif-deficient HIV-1 when stably expressed in T cells, mutate proviral DNA, and are counteracted by HIV-1 Vif. Furthermore, APOBEC3D, APOBEC3F, APOBEC3G, and APOBEC3H of the rhesus macaque also are packaged into and restrict Vif-deficient HIV-1 when stably expressed in T cells, and they are all neutralized by the simian immunodeficiency virus Vif protein. On the other hand, neither human nor rhesus APOBEC3A, APOBEC3B, nor APOBEC3C had a significant impact on HIV-1 replication. These data strongly implicate a combination of four APOBEC3 proteins--APOBEC3D, APOBEC3F, APOBEC3G, and APOBEC3H--in HIV-1 restriction.


Subject(s)
Cytosine Deaminase/immunology , HIV-1/immunology , HIV-1/pathogenicity , Virulence Factors/deficiency , vif Gene Products, Human Immunodeficiency Virus/deficiency , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , Cells, Cultured , Cytosine Deaminase/metabolism , Humans , Macaca mulatta , Molecular Sequence Data , Sequence Analysis, DNA , Simian Immunodeficiency Virus/immunology , Simian Immunodeficiency Virus/pathogenicity
20.
J Virol ; 85(21): 11361-71, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21835794

ABSTRACT

The Apobec3 family of cytidine deaminases can inhibit the replication of retroviruses and retrotransposons. Human and chimpanzee genomes encode seven Apobec3 paralogs; of these, Apobec3DE has the greatest sequence divergence between humans and chimpanzees. Here we show that even though human and chimpanzee Apobec3DEs are very divergent, the two orthologs similarly restrict long terminal repeat (LTR) and non-LTR retrotransposons (MusD and Alu, respectively). However, chimpanzee Apobec3DE also potently restricts two lentiviruses, human immunodeficiency virus type 1 (HIV-1) and the simian immunodeficiency virus (SIV) that infects African green monkeys (SIVagmTAN), unlike human Apobec3DE, which has poor antiviral activity against these same viruses. This difference between human and chimpanzee Apobec3DE in the ability to restrict retroviruses is not due to different levels of Apobec3DE protein incorporation into virions but rather to the ability of Apobec3DE to deaminate the viral genome in target cells. We further show that Apobec3DE rapidly evolved in chimpanzee ancestors approximately 2 to 6 million years ago and that this evolution drove the increased breadth of chimpanzee Apobec3DE antiviral activity to its current high activity against some lentiviruses. Despite a difference in target specificities between human and chimpanzee Apobec3DE, Apobec3DE is likely to currently play a role in host defense against retroelements in both species.


Subject(s)
Cytosine Deaminase/immunology , Cytosine Deaminase/metabolism , Retroelements/immunology , Retroviridae/immunology , Animals , Cell Line , Cluster Analysis , Humans , Molecular Sequence Data , Pan troglodytes , Phylogeny , Recombination, Genetic , Sequence Analysis, DNA , Sequence Homology , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL