Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
Add more filters










Publication year range
1.
Viruses ; 11(9)2019 09 07.
Article in English | MEDLINE | ID: mdl-31500260

ABSTRACT

Feline immunodeficiency virus (FIV) is a naturally occurring T-cell tropic lentiviral disease of felids with many similarities to HIV/AIDS in humans. Similar to primate lentiviral-host interactions, feline APOBEC3 (A3) has been shown to inhibit FIV infection in a host-specific manner and feline A3 degradation is mediated by FIV Vif. Further, infection of felids with non-native FIV strains results in restricted viral replication in both experimental and naturally occurring infections. However, the link between molecular A3-Vif interactions and A3 biological activity during FIV infection has not been well characterized. We thus examined expression of the feline A3 genes A3Z2, A3Z3 and A3Z2-Z3 during experimental infection of domestic cats with host-adapted domestic cat FIV (referred to as FIV) and non-adapted Puma concolor FIV (referred to as puma lentivirus, PLV). We determined A3 expression in different tissues and blood cells from uninfected, FIV-infected, PLV-infected and FIV/PLV co-infected cats; and in purified blood cell subpopulations from FIV-infected and uninfected cats. Additionally, we evaluated regulation of A3 expression by cytokines, mitogens, and FIV infection in cultured cells. In all feline cells and tissues studied, there was a striking difference in expression between the A3 genes which encode FIV inhibitors, with A3Z3 mRNA abundance exceeding that of A3Z2-Z3 by 300-fold or more. Interferon-alpha treatment of cat T cells resulted in upregulation of A3 expression, while treatment with interferon-gamma enhanced expression in cat cell lines. In cats, secondary lymphoid organs and peripheral blood mononuclear cells (PBMC) had the highest basal A3 expression levels and A3 genes were differentially expressed among blood T cells, B cells, and monocytes. Acute FIV and PLV infection of cats, and FIV infection of primary PBMC resulted in no detectable change in A3 expression with the exception of significantly elevated A3 expression in the thymus, the site of highest FIV replication. We conclude that cat A3 expression is regulated by cytokine treatment but, by and large, lentiviral infection did not appear to alter expression. Differences in A3 expression in different blood cell subsets did not appear to impact FIV viral replication kinetics within these cells. Furthermore, the relative abundance of A3Z3 mRNA compared to A3Z2-Z3 suggests that A3Z3 may be the major active anti-lentiviral APOBEC3 gene product in domestic cats.


Subject(s)
Cytosine Deaminase/immunology , Feline Acquired Immunodeficiency Syndrome/enzymology , Immunodeficiency Virus, Feline/physiology , Lentivirus Infections/veterinary , Animals , B-Lymphocytes/immunology , Cats , Cytosine Deaminase/genetics , Feline Acquired Immunodeficiency Syndrome/genetics , Feline Acquired Immunodeficiency Syndrome/immunology , Feline Acquired Immunodeficiency Syndrome/virology , Host-Pathogen Interactions , Immunodeficiency Virus, Feline/genetics , Lentivirus Infections/enzymology , Lentivirus Infections/genetics , Lentivirus Infections/immunology , T-Lymphocytes/immunology , Virus Replication
2.
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
3.
Trends Mol Med ; 24(5): 507-520, 2018 05.
Article in English | MEDLINE | ID: mdl-29609878

ABSTRACT

The infectivity of HIV depends on overcoming APOBEC3 (A3) innate immunity, predominantly through the expression of the viral protein Vif, which induces A3 degradation in the proteasome. Disruption of the functional interactions of Vif enables A3 mutagenesis of the HIV genome during viral replication, which can result in a broadly neutralizing antiviral effect. Vif function requires self-association along with interactions with A3 proteins, protein chaperones, and factors of the ubiquitination machinery and these are described here as a potential platform for novel antiviral drug discovery. This Review will examine the current state of development of Vif inhibitors that we believe to have therapeutic and functional cure potential.


Subject(s)
Anti-Retroviral Agents/therapeutic use , Cytosine Deaminase/immunology , HIV Infections/drug therapy , HIV-1/drug effects , Immunity, Innate/drug effects , vif Gene Products, Human Immunodeficiency Virus/immunology , APOBEC Deaminases , Cytidine Deaminase , Cytosine Deaminase/metabolism , HIV Infections/immunology , HIV Infections/virology , HIV-1/immunology , HIV-1/physiology , Host-Pathogen Interactions/drug effects , Host-Pathogen Interactions/immunology , Humans , Protein Binding/drug effects , Proteolysis/drug effects , vif Gene Products, Human Immunodeficiency Virus/metabolism
4.
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
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.
Virus Res ; 231: 21-33, 2017 03 02.
Article in English | MEDLINE | ID: mdl-27890631

ABSTRACT

A majority of human papillomavirus (HPV) infections are asymptomatic and self-resolving in the absence of medical interventions. Various innate and adaptive immune responses, as well as physical barriers, have been implicated in controlling early HPV infections. However, if HPV overcomes these host immune defenses and establishes persistence in basal keratinocytes, it becomes very difficult for the host to eliminate the infection. The HPV oncoproteins E5, E6, and E7 are important in regulating host immune responses. These oncoproteins dysregulate gene expression, protein-protein interactions, posttranslational modifications, and cellular trafficking of critical host immune modulators. In addition to the HPV oncoproteins, sequence variation and dinucleotide depletion in papillomavirus genomes has been suggested as an alternative strategy for evasion of host immune defenses. Since anti-HPV host immune responses are also considered to be important for antitumor immunity, immune dysregulation by HPV during virus persistence may contribute to immune suppression essential for HPV-associated cancer progression. Here, we discuss cellular pathways dysregulated by HPV that allow the virus to evade various host immune defenses.


Subject(s)
Genome, Viral , Immune Evasion , Keratinocytes/immunology , NF-kappa B/genetics , Oncogene Proteins, Viral/genetics , Papillomaviridae/genetics , Papillomavirus Infections/virology , APOBEC Deaminases , Adaptive Immunity , Cytidine Deaminase , Cytosine Deaminase/genetics , Cytosine Deaminase/immunology , DNA Methylation , Gene Expression Regulation , Histones/genetics , Histones/immunology , Humans , Immunity, Innate , Keratinocytes/virology , NF-kappa B/immunology , Oncogene Proteins, Viral/immunology , Papillomaviridae/growth & development , Papillomaviridae/pathogenicity , Papillomavirus Infections/genetics , Papillomavirus Infections/immunology , Papillomavirus Infections/pathology , Virion/genetics , Virion/growth & development , Virion/pathogenicity , Virus Replication
7.
Sci Rep ; 6: 34752, 2016 10 13.
Article in English | MEDLINE | ID: mdl-27734899

ABSTRACT

The Bowman-Birk inhibitor (BBI), a soybean-derived protease inhibitor, is known to have anti-inflammatory effect in both in vitro and in vivo systems. Macrophages play a key role in inflammation and immune activation, which is implicated in HIV disease progression. Here, we investigated the effect of BBI on HIV infection of peripheral blood monocyte-derived macrophages. We demonstrated that BBI could potently inhibit HIV replication in macrophages without cytotoxicity. Investigation of the mechanism(s) of BBI action on HIV showed that BBI induced the expression of IFN-ß and multiple IFN stimulated genes (ISGs), including Myxovirus resistance protein 2 (Mx2), 2',5'-oligoadenylate synthetase (OAS-1), Virus inhibitory protein (viperin), ISG15 and ISG56. BBI treatment of macrophages also increased the expression of several known HIV restriction factors, including APOBEC3F, APOBEC3G and tetherin. Furthermore, BBI enhanced the phosphorylation of IRF3, a key regulator of IFN-ß. The inhibition of IFN-ß pathway by the neutralization antibody to type I IFN receptor (Anti-IFNAR) abolished BBI-mediated induction of the anti-HIV factors and inhibition of HIV in macrophages. These findings that BBI could activate IFN-ß-mediated signaling pathway, initialize the intracellular innate immunity in macrophages and potently inhibit HIV at multiple steps of viral replication cycle indicate the necessity to further investigate BBI as an alternative and cost-effective anti-HIV natural product.


Subject(s)
Anti-HIV Agents/pharmacology , HIV-1/drug effects , Host-Pathogen Interactions/immunology , Interferon-beta/antagonists & inhibitors , Macrophages/immunology , Trypsin Inhibitor, Bowman-Birk Soybean/pharmacology , Virus Replication/drug effects , 2',5'-Oligoadenylate Synthetase/genetics , 2',5'-Oligoadenylate Synthetase/immunology , APOBEC-3G Deaminase/genetics , APOBEC-3G Deaminase/immunology , Adaptor Proteins, Signal Transducing , Antibodies, Neutralizing/pharmacology , Cells, Cultured , Cytokines/genetics , Cytokines/immunology , Cytosine Deaminase/genetics , Cytosine Deaminase/immunology , Gene Expression Regulation , HIV-1/genetics , HIV-1/growth & development , Host-Pathogen Interactions/genetics , Humans , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/immunology , Interferon-beta/genetics , Interferon-beta/immunology , Macrophages/metabolism , Macrophages/virology , Myxovirus Resistance Proteins/genetics , Myxovirus Resistance Proteins/immunology , Oxidoreductases Acting on CH-CH Group Donors , Proteins/genetics , Proteins/immunology , RNA-Binding Proteins , Receptor, Interferon alpha-beta/antagonists & inhibitors , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/immunology , Signal Transduction , Transcription Factors/genetics , Transcription Factors/immunology , Ubiquitins/genetics , Ubiquitins/immunology
8.
Immunobiology ; 221(2): 282-90, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26563749

ABSTRACT

Given the multifactorial nature of action of type I interferon (IFN) in HIV-1 infection and the need to firmly establish the action of key components of IFN pathways, we compared the IFN stimulated gene (ISG)15 expression with that of other well-characterized ISGs, evaluating its relationship with immunosuppressive factors regulating T-cell response in HIV-1 patients. PBMC from 225 subjects were included: healthy donors (n=30), naïve (n=93) and HAART treated HIV-1 subjects (n=102). Levels of ISG15-mRNA, ISG56-mRNA, APOBEC3G/3F-mRNA, TRAIL-mRNA, IDO-mRNA, proviral load andISG15 (rs15842 and rs1921) SNPs were evaluated by using TaqMan assays. We found that ISG15, ISG56, APOBEC3G/3F levels were increased in untreated HIV-1 patients compared to healthy donors, being ISG15 the highest ISG expressed. The amount of ISG15 correlated with viral load and with CD4+ T cell counts whereas no relationship was found between all ISGs analyzed and proviral load or HIV-1 tropism. ISG15 expression was reduced following long-term antiretroviral therapy. In addition, ISG15 levels were correlated with those of TRAIL and IDO in HIV-1 viremic patients. Lastly, ISG15 SNPs had no influence on ISG15 levels. We demonstrates that ISG15 is elevated in viremic HIV-1 patients and is associated with high TRAIL and IDO levels.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cytokines/genetics , HIV Infections/genetics , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , RNA, Messenger/genetics , TNF-Related Apoptosis-Inducing Ligand/genetics , Ubiquitins/genetics , Viremia/genetics , APOBEC-3G Deaminase , Adaptor Proteins, Signal Transducing , Adult , Antiretroviral Therapy, Highly Active , CD4 Lymphocyte Count , CD4-Positive T-Lymphocytes/virology , Case-Control Studies , Cytidine Deaminase/genetics , Cytidine Deaminase/immunology , Cytokines/immunology , Cytosine Deaminase/genetics , Cytosine Deaminase/immunology , Female , Gene Expression Regulation , HIV Infections/drug therapy , HIV Infections/immunology , HIV Infections/virology , HIV-1/drug effects , HIV-1/immunology , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Male , Middle Aged , Polymorphism, Single Nucleotide , RNA, Messenger/immunology , RNA-Binding Proteins , Signal Transduction , TNF-Related Apoptosis-Inducing Ligand/immunology , Transcription Factors/genetics , Transcription Factors/immunology , Ubiquitins/immunology , Viral Load/drug effects , Viral Tropism , Viremia/drug therapy , Viremia/immunology , Viremia/virology
9.
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
10.
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
11.
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
12.
Trends Mol Med ; 21(5): 274-84, 2015 May.
Article in English | MEDLINE | ID: mdl-25820175

ABSTRACT

The APOBEC3 cytosine deaminases play key roles in innate immunity through their ability to mutagenize viral DNA and restrict viral replication. Recent advances in cancer genomics, together with biochemical characterization of the APOBEC3 enzymes, have now implicated at least two family members in somatic mutagenesis during tumor development. We review the evidence linking these enzymes to carcinogenesis and highlight key questions, including the potential mechanisms that misdirect APOBEC3 activity to the host genome, the links to viral infection, and the association between a common APOBEC3 polymorphism and cancer risk.


Subject(s)
Carcinogenesis/genetics , Cytosine Deaminase/genetics , Neoplasms/genetics , Retroviridae/genetics , Animals , Carcinogenesis/immunology , Cytosine Deaminase/immunology , Humans , Immunity, Innate/genetics , Immunity, Innate/immunology , Mutagenesis/genetics , Mutagenesis/immunology , Neoplasms/immunology , Neoplasms/virology , Retroviridae/immunology , Risk , Virus Replication/genetics , Virus Replication/immunology
13.
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
14.
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
15.
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
16.
Structure ; 22(5): 668-84, 2014 May 06.
Article in English | MEDLINE | ID: mdl-24657093

ABSTRACT

Human APOBEC3 (A3) proteins are host-encoded intrinsic restriction factors that inhibit the replication of many retroviral pathogens. Restriction is believed to occur as a result of the DNA cytosine deaminase activity of the A3 proteins; this activity converts cytosines into uracils in single-stranded DNA retroviral replication intermediates. A3 proteins are also equipped with deamination-independent means to restrict retroviruses that work cooperatively with deamination-dependent restriction pathways. A3 proteins substantially bolster the intrinsic immune system by providing a powerful block to the transmission of retroviral pathogens; however, most retroviruses are able to subvert this replicative restriction in their natural host. HIV-1, for instance, evades A3 proteins through the activity of its accessory protein Vif. Here, we summarize data from recent A3 structural and functional studies to provide perspectives into the interactions between cellular A3 proteins and HIV-1 macromolecules throughout the viral replication cycle.


Subject(s)
Cytosine Deaminase/chemistry , Cytosine Deaminase/metabolism , HIV-1/pathogenicity , vif Gene Products, Human Immunodeficiency Virus/metabolism , APOBEC Deaminases , Amino Acid Sequence , Binding Sites , Cytidine Deaminase , Cytosine Deaminase/genetics , Cytosine Deaminase/immunology , DNA, Single-Stranded/metabolism , Host-Pathogen Interactions , Humans , Models, Molecular , Molecular Sequence Data , Nucleocapsid/metabolism , Sequence Homology, Amino Acid , Structure-Activity Relationship , Substrate Specificity , Virus Replication
17.
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
18.
AIDS ; 28(4): 487-91, 2014 Feb 20.
Article in English | MEDLINE | ID: mdl-24401644

ABSTRACT

OBJECTIVE: Previous studies on HIV quasispecies have revealed HIV compartmentalization in various tissues within an infected individual. Such HIV variation is a result of a combination of factors including high replication and mutation rates, recombination, and APOBEC3-host selective pressure. METHODS: To evaluate the differential impact of APOBEC3 editing in HIV-1 compartments, we analyzed the level of G-to-A hypermutation in HIV-1 protease and reverse transcriptase sequences among 30 HAART-treated patients for whom peripheral blood mononuclear cells and body tissues or fluids [cerebral spinal fluid (CSF), rectal tissue, or renal tissue] were collected on the same day. RESULTS: APOBEC3-mediated hypermutation was identified in 36% (11/30) of participants in at least one viral reservoir. HIV hypermutated sequences were often observed in viral sanctuaries (total n = 10; CSF, n = 6; renal tissue, n = 1; rectal tissue n = 3) compared with peripheral blood (total n = 4). Accordingly, APOBEC3 editing generated more G-to-A drug resistance mutations in sanctuaries: three patients' CSF (i.e. G73S in protease; M184I, M230I in reverse transcriptase) and two other patients' rectal tissues (M184I, M230I in reverse transcriptase) while such mutations were absent from paired peripheral blood mononuclear cells. CONCLUSION: APOBEC3-induced mutations observed in peripheral blood underestimate the overall proportion of hypermutated viruses in anatomical compartments. The resulting mutations may favor escape to antiretrovirals in these compartments in conjunction with a lower penetration of drugs in some sanctuaries. On the other side, because hypermutated sequences often harbor inactivating mutations, our results suggest that accumulation of defective viruses may be more dominant in sanctuaries than in peripheral blood of patients on effective HAART.


Subject(s)
Cytosine Deaminase/immunology , HIV-1/immunology , HIV-1/physiology , Mutation , Virus Replication , APOBEC Deaminases , Body Fluids/virology , Cytidine Deaminase , Evolution, Molecular , HIV-1/classification , HIV-1/genetics , Humans , Leukocytes, Mononuclear/virology , RNA, Viral/genetics , Selection, Genetic
19.
J Mol Biol ; 426(6): 1220-45, 2014 Mar 20.
Article in English | MEDLINE | ID: mdl-24189052

ABSTRACT

Several members of the APOBEC3 family of cellular restriction factors provide intrinsic immunity to the host against viral infection. Specifically, APOBEC3DE, APOBEC3F, APOBEC3G, and APOBEC3H haplotypes II, V, and VII provide protection against HIV-1Δvif through hypermutation of the viral genome, inhibition of reverse transcription, and inhibition of viral DNA integration into the host genome. HIV-1 counteracts APOBEC3 proteins by encoding the viral protein Vif, which contains distinct domains that specifically interact with these APOBEC3 proteins to ensure their proteasomal degradation, allowing virus replication to proceed. Here, we review our current understanding of APOBEC3 structure, editing and non-editing mechanisms of APOBEC3-mediated restriction, Vif-APOBEC3 interactions that trigger APOBEC3 degradation, and the contribution of APOBEC3 proteins to restriction and control of HIV-1 replication in infected patients.


Subject(s)
Cytosine Deaminase/immunology , HIV Infections/genetics , HIV/physiology , Immunity, Innate/immunology , Virus Replication/immunology , vif Gene Products, Human Immunodeficiency Virus/metabolism , APOBEC Deaminases , Cytidine Deaminase , DNA, Viral/genetics , HIV Infections/drug therapy , HIV Infections/virology , Humans
20.
Viruses ; 5(8): 1934-47, 2013 Jul 30.
Article in English | MEDLINE | ID: mdl-23903287

ABSTRACT

It is becoming increasingly clear that organisms have developed a variety of mechanisms to fight against viral infection. The viruses have developed means of counteracting these defences in various ways. The APOBEC3 proteins are a mammalian-specific family of nucleic acid cytidine deaminases that block retroviral infection. These inhibitors are counteracted by the Vif proteins encoded by most lentiviruses. In this paper, we will review the interaction of the lentiviral Vif proteins with the APOBEC3 proteins, with an emphasis on sheep APOBEC3 and maedi-visna virus (MVV) Vif.


Subject(s)
Cytosine Deaminase/immunology , Cytosine Deaminase/metabolism , Gene Products, vif/metabolism , Host-Pathogen Interactions , Virulence Factors/metabolism , Visna-maedi virus/immunology , Animals , Sheep , Sheep Diseases/immunology , Sheep Diseases/virology
SELECTION OF CITATIONS
SEARCH DETAIL