Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters











Publication year range
1.
Viruses ; 14(11)2022 11 12.
Article in English | MEDLINE | ID: mdl-36423112

ABSTRACT

Human immunodeficiency virus-1 (HIV-1) is a retrovirus that integrates its reverse-transcribed genome as proviral DNA into the host genome to establish a successful infection. The viral genome integration requires safeguarding the subviral complexes, reverse transcription complex (RTC) and preintegration complex (PIC), in the cytosol from degradation, presumably effectively secured by the capsid surrounding these complexes. An intact capsid, however, is a large structure, which raises concerns about its translocation from cytoplasm to nucleus crossing the nuclear membrane, guarded by complex nuclear pore structures, which do not allow non-specific transport of large molecules. In addition, the generation of new virions requires the export of incompletely processed viral RNA from the nucleus to the cytoplasm, an event conventionally not permitted through mammalian nuclear membranes. HIV-1 has evolved multiple mechanisms involving redundant host pathways by liaison with the cell's nucleocytoplasmic trafficking system, failure of which would lead to the collapse of the infection cycle. This review aims to assemble the current developments in temporal and spatial events governing nucleocytoplasmic transport of HIV-1 factors. Discoveries are anticipated to serve as the foundation for devising host-directed therapies involving selective abolishment of the critical interactomes between viral proteins and their host equivalents.


Subject(s)
HIV Infections , HIV-1 , Humans , Active Transport, Cell Nucleus , Capsid/metabolism , Capsid Proteins/genetics , HIV-1/genetics , HIV-1/metabolism , Virus Integration
2.
Sci Data ; 9(1): 240, 2022 05 27.
Article in English | MEDLINE | ID: mdl-35624179

ABSTRACT

Urothelial carcinoma (UC) of the urinary bladder is a prevalent cancer worldwide. Because histone deacetylases (HDACs) are important factors in cancer, targeting these epigenetic regulators is considered an attractive strategy to develop novel anticancer drugs. Whereas HDAC1 and HDAC2 promote UC, HDAC5 is often downregulated and only weakly expressed in UC cell lines, suggesting a tumor-suppressive function. We studied the effect of stable lentiviral-mediated HDAC5 overexpression in four UC cell lines with different phenotypes (RT112, VM-Cub-1, SW1710, and UM-UC-3, each with vector controls). In particular, comprehensive proteomics and RNA-seq transcriptomics analyses were performed on the four cell line pairs, which are described here. For comparison, the immortalized benign urothelial cell line HBLAK was included. These datasets will be a useful resource for researchers studying UC, and especially the influence of HDAC5 on epithelial-mesenchymal transition (EMT). Moreover, these data will inform studies on HDAC5 as a less studied member of the HDAC family in other cell types and diseases, especially fibrosis.


Subject(s)
Carcinoma, Transitional Cell , Histone Deacetylases , Urinary Bladder Neoplasms , Carcinoma, Transitional Cell/genetics , Carcinoma, Transitional Cell/metabolism , Cell Line, Tumor , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , Proteomics , Transcriptome , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/metabolism
3.
Viruses ; 13(12)2021 12 08.
Article in English | MEDLINE | ID: mdl-34960728

ABSTRACT

Staufen, the RNA-binding family of proteins, affects various steps in the Human Immuno-Deficiency Virus (HIV-1) replication cycle. While our previous study established Staufen-2-HIV-1 Rev interaction and its role in augmenting nucleocytoplasmic export of RRE-containing viral RNA, viral incorporation of Staufen-2 and its effect on viral propagation were unknown. Here, we report that Staufen-2 interacts with HIV-1 Gag and is incorporated into virions and that encapsidated Staufen-2 boosted viral infectivity. Further, Staufen-2 gets co-packaged into virions, possibly by interacting with host factors Staufen-1 or antiviral protein APOBEC3G, which resulted in different outcomes on the infectivity of Staufen-2-encapsidated virions. These observations suggest that encapsidated host factors influence viral population dynamics and infectivity. With the explicit identification of the incorporation of Staufen proteins into HIV-1 and other retroviruses, such as Simian Immunodeficiency Virus (SIV), we propose that packaging of RNA binding proteins, such as Staufen, in budding virions of retroviruses is probably a general phenomenon that can drive or impact the viral population dynamics, infectivity, and evolution.


Subject(s)
HIV Infections/metabolism , HIV-1/physiology , HIV-1/pathogenicity , Nerve Tissue Proteins/metabolism , RNA-Binding Proteins/metabolism , Viral Genome Packaging , Cell Line , Gene Products, gag/genetics , Gene Products, gag/metabolism , HIV Infections/genetics , HIV Infections/virology , HIV-1/genetics , Host-Pathogen Interactions , Humans , Nerve Tissue Proteins/genetics , Protein Binding , RNA-Binding Proteins/genetics , Virulence
4.
Viruses ; 13(3)2021 03 18.
Article in English | MEDLINE | ID: mdl-33803830

ABSTRACT

Non-human primates (NHP) are an important source of viruses that can spillover to humans and, after adaptation, spread through the host population. Whereas HIV-1 and HTLV-1 emerged as retroviral pathogens in humans, a unique class of retroviruses called foamy viruses (FV) with zoonotic potential are occasionally detected in bushmeat hunters or zookeepers. Various FVs are endemic in numerous mammalian natural hosts, such as primates, felines, bovines, and equines, and other animals, but not in humans. They are apathogenic, and significant differences exist between the viral life cycles of FV and other retroviruses. Importantly, FVs replicate in the presence of many well-defined retroviral restriction factors such as TRIM5α, BST2 (Tetherin), MX2, and APOBEC3 (A3). While the interaction of A3s with HIV-1 is well studied, the escape mechanisms of FVs from restriction by A3 is much less explored. Here we review the current knowledge of FV biology, host restriction factors, and FV-host interactions with an emphasis on the consequences of FV regulatory protein Bet binding to A3s and outline crucial open questions for future studies.


Subject(s)
APOBEC Deaminases/metabolism , Host Microbial Interactions , Retroviridae Proteins/metabolism , Spumavirus/genetics , Spumavirus/physiology , Animals , Cell Line , Humans , Mutation , Primates/virology , Retroviridae Infections/immunology , Retroviridae Infections/virology , Retroviridae Proteins/classification , Retroviridae Proteins/genetics , Spumavirus/immunology
5.
Virology ; 557: 1-14, 2021 05.
Article in English | MEDLINE | ID: mdl-33581610

ABSTRACT

The human APOBEC3A (A3A) polynucleotide cytidine deaminase has been shown to have antiviral activity against HTLV-1 but not HIV-1, when expressed in the virus producer cell. In viral target cells, high levels of endogenous A3A activity have been associated with the restriction of HIV-1 during infection. Here we demonstrate that A3A derived from both target cells and producer cells can block the infection of Moloney-MLV (MLV) and related AKV-derived strains of MLV in a deaminase-dependent mode. Furthermore, glycosylated Gag (glycoGag) of MLV inhibits the encapsidation of human A3A, but target cell A3A was not affected by glycoGag and exerted deamination of viral DNA. Importantly, our results clearly indicate that poor glycoGag expression in MLV gag-pol packaging constructs as compared to abundant levels in full-length amphotropic MLV makes these viral vectors sensitive to A3A-mediated restriction. This raises the possibility of acquiring A3A-induced mutations in retroviral gene therapy applications.


Subject(s)
Cytidine Deaminase/genetics , Cytidine Deaminase/metabolism , Gene Products, gag/metabolism , Proteins/genetics , Proteins/metabolism , Animals , DNA, Viral , Gene Products, gag/genetics , Glycosylation , HEK293 Cells , Humans , Interferon-beta/pharmacology , Leukemia Virus, Murine , Macrophages/drug effects , Macrophages/virology , Mice
6.
Appetite ; 159: 105050, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33249010

ABSTRACT

Previous paradigms used to examine attentional distraction by task-irrelevant food words and food images were not suited for the investigation of involuntary and automatic attentional capture. In the current experiments we adapted a well-established visual-search paradigm (with eye tracking) to investigate involuntary attentional capture by food and drink rewards. We first used a satiety procedure to manipulate relative preference for different food and drink outcomes (potato chips and water in Experiment 1 and popcorn and chocolate Smarties in Experiment 2). Participants then performed the visual-search task where a coloured distractor signalled on each trial which of the two food and drink rewards was available for successful identification of the target. The signalled reward was cancelled, however, if any eye gaze was registered on the distractor. Participants were therefore motivated to try and control the automatic orienting of attention towards cues signalling valuable outcomes, in order to earn those outcomes. In both experiments we found that attention was more often captured by the distractor signalling the valuable (non-sated) outcome, replicating previous studies using this paradigm with monetary rewards. We also found that those scoring high on eating restraint (as measured with the Dutch Eating Behavior Questionnaire) were better at controlling reflexive orienting of attention to desirable food rewards. This paradigm offers a novel approach for understanding how reflexive attention and control relate to conflicts in everyday life around distracting food cues, and the moderating role of dietary restraint.


Subject(s)
Food , Reward , Cues , Fixation, Ocular , Humans , Reaction Time , Satiation
7.
J Mol Biol ; 432(23): 6200-6227, 2020 11 20.
Article in English | MEDLINE | ID: mdl-33068636

ABSTRACT

APOBEC3 deaminases (A3s) provide mammals with an anti-retroviral barrier by catalyzing dC-to-dU deamination on viral ssDNA. Within primates, A3s have undergone a complex evolution via gene duplications, fusions, arms race, and selection. Human APOBEC3C (hA3C) efficiently restricts the replication of viral infectivity factor (vif)-deficient Simian immunodeficiency virus (SIVΔvif), but for unknown reasons, it inhibits HIV-1Δvif only weakly. In catarrhines (Old World monkeys and apes), the A3C loop 1 displays the conserved amino acid pair WE, while the corresponding consensus sequence in A3F and A3D is the largely divergent pair RK, which is also the inferred ancestral sequence for the last common ancestor of A3C and of the C-terminal domains of A3D and A3F in primates. Here, we report that modifying the WE residues in hA3C loop 1 to RK leads to stronger interactions with substrate ssDNA, facilitating catalytic function, which results in a drastic increase in both deamination activity and in the ability to restrict HIV-1 and LINE-1 replication. Conversely, the modification hA3F_WE resulted only in a marginal decrease in HIV-1Δvif inhibition. We propose that the two series of ancestral gene duplications that generated A3C, A3D-CTD and A3F-CTD allowed neo/subfunctionalization: A3F-CTD maintained the ancestral RK residues in loop 1, while diversifying selection resulted in the RK â†’ WE modification in Old World anthropoids' A3C, possibly allowing for novel substrate specificity and function.


Subject(s)
Cytidine Deaminase/genetics , HIV Infections/genetics , HIV-1/genetics , vif Gene Products, Human Immunodeficiency Virus/genetics , Antiviral Agents/metabolism , DNA, Single-Stranded/genetics , HIV Infections/therapy , HIV Infections/virology , HIV-1/pathogenicity , Humans , Mutagenesis, Site-Directed , Protein Binding/genetics
8.
Cell Host Microbe ; 28(2): 306-312.e6, 2020 08 12.
Article in English | MEDLINE | ID: mdl-32533923

ABSTRACT

Epitranscriptomic RNA modifications, including methylation of adenine and cytidine residues, are now recognized as key regulators of both cellular and viral mRNA function. Moreover, acetylation of the N4 position of cytidine (ac4C) was recently reported to increase the translation and stability of cellular mRNAs. Here, we show that ac4C and N-acetyltransferase 10 (NAT10), the enzyme that adds ac4C to RNAs, have been subverted by human immunodeficiency virus 1 (HIV-1) to increase viral gene expression. HIV-1 transcripts are modified with ac4C at multiple discrete sites, and silent mutagenesis of these ac4C sites led to decreased HIV-1 gene expression. Similarly, loss of ac4C from viral transcripts due to depletion of NAT10 inhibited HIV-1 replication by reducing viral RNA stability. Interestingly, the NAT10 inhibitor remodelin could inhibit HIV-1 replication at concentrations that have no effect on cell viability, thus identifying ac4C addition as a potential target for antiviral drug development.


Subject(s)
Gene Expression Regulation, Viral/genetics , Gene Expression/drug effects , HIV-1/genetics , RNA Stability/drug effects , RNA, Viral/genetics , Acetylation/drug effects , Cell Line , Cytidine/metabolism , Female , Gene Expression Regulation, Viral/drug effects , HEK293 Cells , HIV-1/growth & development , Humans , Hydrazones/pharmacology , Male , N-Terminal Acetyltransferases/metabolism , RNA Stability/genetics , Thiazoles/pharmacology , Virus Replication/physiology
9.
Int J Mol Sci ; 20(9)2019 Apr 30.
Article in English | MEDLINE | ID: mdl-31052182

ABSTRACT

Class I histone deacetylases (HDACs) generally promote cell proliferation and tumorigenesis, whereas class IIA HDACs like HDAC4 and HDAC5 may promote or impede cancer development in a tissue-dependent manner. In urothelial carcinoma (UC), HDAC5 is often downregulated. Accordingly, HDAC5 was weakly expressed in UC cell lines suggesting a possible tumor-suppressive function. We therefore characterized the effects of stable HDAC5 expression in four UC cell lines (RT112, VM-Cub-1, SW1710 and UM-UC-3) with different phenotypes reflecting the heterogeneity of UC, by assessing proliferation, clonogenicity and migration ability. Further, we detailed changes in the proteome and transcriptome by immunoblotting, mass spectrometry and RNA sequencing analysis. We observed that HDAC5 overexpression in general decreased cell proliferation, but in one cell line (VM-Cub-1) induced a dramatic change from an epitheloid to a mesenchymal phenotype, i.e., epithelial-mesenchymal transition (EMT). These phenotypical changes were confirmed by comprehensive proteomics and transcriptomics analyses. In contrast to HDAC5, overexpression of HDAC4 exerted only weak effects on cell proliferation and phenotypes. We conclude that overexpression of HDAC5 may generally decrease proliferation in UC, but, intriguingly, may induce EMT on its own in certain circumstances.


Subject(s)
Carcinoma/metabolism , Cell Proliferation , Epithelial-Mesenchymal Transition , Histone Deacetylases/genetics , Urinary Bladder Neoplasms/metabolism , Urothelium/pathology , Carcinoma/genetics , Cell Line, Tumor , HEK293 Cells , Histone Deacetylases/metabolism , Humans , RNA, Messenger/genetics , RNA, Messenger/metabolism , Urinary Bladder Neoplasms/genetics , Urothelium/metabolism
10.
Front Microbiol ; 9: 2088, 2018.
Article in English | MEDLINE | ID: mdl-30233553

ABSTRACT

The most common mutational signature in urothelial carcinoma (UC), the most common type of urinary bladder cancer is assumed to be caused by the misdirected activity of APOBEC3 (A3) cytidine deaminases, especially A3A or A3B, which are known to normally restrict the propagation of exogenous viruses and endogenous retroelements such as LINE-1 (L1). The involvement of A3 proteins in urothelial carcinogenesis is unexpected because, to date, UC is thought to be caused by chemical carcinogens rather than viral activity. Therefore, we explored the relationship between A3 expression and L1 activity, which is generally upregulated in UC. We found that UC cell lines highly express A3B and in some cases A3G, but not A3A, and exhibit corresponding cytidine deamination activity in vitro. While we observed evidence suggesting that L1 expression has a weak positive effect on A3B and A3G expression and A3B promoter activity, neither efficient siRNA-mediated knockdown nor overexpression of functional L1 elements affected catalytic activity of A3 proteins consistently. However, L1 knockdown diminished proliferation of a UC cell line exhibiting robust endogenous L1 expression, but had little impact on a cell line with low L1 expression levels. Our results indicate that UC cells express A3B at levels exceeding A3A levels by far, making A3B the prime candidate for causing genomic mutations. Our data provide evidence that L1 activation constitutes only a minor and negligible factor involved in induction or upregulation of endogenous A3 expression in UC.

11.
Virology ; 523: 52-63, 2018 10.
Article in English | MEDLINE | ID: mdl-30081309

ABSTRACT

Human myxovirus resistance protein B (hMXB) is a restriction factor of HIV-1 that also inhibits a variety of retroviruses. However, hMXB is not antiviral against equine infectious anemia virus (EIAV). We show here that equine MX2 (eMX2) potently restricts EIAV in vitro. Additionally, eMX2 inhibits HIV-1 and other lentiviruses, including murine leukemia virus. Previously, it was reported that hMXB repression is reduced in hMXB Δ1-25, but not in GTP-binding mutant K131A and GTP-hydrolysis mutant T151A. In contrast to this phenomenon, our study indicates that eMX2 restriction is not diminished in eMX2 Δ1-25, but is in eMX2 K127A and T147A, which correspond to hMXB K131A and T151A, respectively. Thus, eMX2 may inhibit retroviral replication by a novel mechanism that differs from that of hMXB.


Subject(s)
HIV-1/genetics , Host-Pathogen Interactions , Infectious Anemia Virus, Equine/genetics , Myxovirus Resistance Proteins/genetics , Amino Acid Substitution , Animals , Base Sequence , Binding Sites , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Guanosine Triphosphate/metabolism , HIV-1/growth & development , HIV-1/metabolism , Horses , Humans , Infectious Anemia Virus, Equine/growth & development , Infectious Anemia Virus, Equine/metabolism , Lentivirus/genetics , Lentivirus/metabolism , Mutation , Myxovirus Resistance Proteins/metabolism , Protein Binding , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction
12.
J Virol ; 92(20)2018 10 15.
Article in English | MEDLINE | ID: mdl-30068654

ABSTRACT

The host intrinsic innate immune system drives antiviral defenses and viral restriction, which includes the production of soluble factors, such as type I and III interferon (IFN), and activation of restriction factors, including SAMHD1, a deoxynucleoside triphosphohydrolase. Interferon-stimulated gene 15 (ISG15)-specific ubiquitin-like protease 43 (USP18) abrogates IFN signaling pathways. The cyclin-dependent kinase inhibitor p21 (CIP1/WAF1), which is involved in the differentiation and maturation of monocytes, inhibits human immunodeficiency virus type 1 (HIV-1) in macrophages and dendritic cells. p21 inhibition of HIV-1 replication is thought to occur at the reverse transcription step, likely by suppressing cellular deoxynucleoside triphosphate (dNTP) biosynthesis and increasing the amount of antivirally active form of SAMHD1. SAMHD1 strongly inhibits HIV-1 replication in myeloid and resting CD4+ T cells. Here, we studied how USP18 influences HIV-1 replication in human myeloid THP-1 cells. We found that USP18 has the novel ability to inhibit the antiviral function of p21 in differentiated THP-1 cells. USP18 enhanced reverse transcription of HIV-1 by downregulating p21 expression and upregulating intracellular dNTP levels. p21 downregulation by USP18 was associated with the active form of SAMHD1, phosphorylated at T592. USP18 formed a complex with the E3 ubiquitin ligase recognition factor SKP2 (S-phase kinase associated protein 2) and SAMHD1. CRISPR-Cas9 knockout of USP18 increased p21 protein expression and blocked HIV-1 replication. Overall, we propose USP18 as a regulator of p21 antiviral function in differentiated myeloid THP-1 cells.IMPORTANCE Macrophages and dendritic cells are usually the first point of contact with pathogens, including lentiviruses. Host restriction factors, including SAMHD1, mediate the innate immune response against these viruses. However, HIV-1 has evolved to circumvent the innate immune response and establishes disseminated infection. The cyclin-dependent kinase inhibitor p21, which is involved in differentiation and maturation of monocytes, blocks HIV-1 replication at the reverse transcription step. p21 is thought to suppress key enzymes involved in dNTP biosynthesis and activates SAMHD1 antiviral function. We report here that the human USP18 protein is a novel factor potentially contributing to HIV replication by blocking the antiviral function of p21 in differentiated human myeloid cells. USP18 downregulates p21 protein expression, which correlates with upregulated intracellular dNTP levels and the antiviral inactive form of SAMHD1. Depletion of USP18 stabilizes p21 protein expression, which correlates with dephosphorylated SAMHD1 and a block to HIV-1 replication.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/metabolism , Endopeptidases/metabolism , HIV-1/immunology , Immunity, Innate , Macrophages/immunology , Macrophages/virology , Endopeptidases/genetics , Gene Knockout Techniques , Humans , THP-1 Cells , Ubiquitin Thiolesterase
13.
Clin Epigenetics ; 10(1): 100, 2018 07 31.
Article in English | MEDLINE | ID: mdl-30064501

ABSTRACT

BACKGROUND: Histone deacetylase inhibitors (HDACi) are promising anti-cancer drugs that could also be employed for urothelial carcinoma (UC) therapy. It is unclear, however, whether inhibition of all 11 zinc-dependent HDACs or of individual enzymes is more efficacious and specific. Here, we investigated the novel HDACi 19i (LMK235) with presumed preferential activity against class IIA HDAC4/5 in comparison to the pan-HDACi vorinostat (SAHA) and the HDAC4-specific HDACi TMP269 in UC cell lines with basal expression of HDAC4 and characterized two HDAC4-overexpressing UC cell lines. METHODS: Cytotoxic concentrations 50% (CC50s) for HDACi were determined by MTT assay and high-content analysis-based fluorescent live/dead assay in UC cell lines with different expression of HDAC4 and as well as in normal urothelial cell cultures, HBLAK and HEK-293 cell lines. Effects of HDACis were analyzed by flow cytometry; molecular changes were followed by qRT-PCR and Western blots. UC lines overexpressing HDAC4 were established by lentiviral transduction. Inhibitor activity profiles of HDACi were obtained by current state in vitro assays, and docking analysis was performed using an updated crystal structure of HDAC4. RESULTS: In UC cell lines, 19i CC50s ranged around 1 µM; control lines were similarly or less sensitive. Like SAHA, 19i increased the G2/M-fraction, disturbed mitosis, and elicited apoptosis or in some cells senescence. Thymidylate synthase expression was diminished, and p21CIP1 was induced; global histone acetylation and α-tubulin acetylation also increased. In most cell lines, 19i as well as SAHA induced HDAC5 and HDAC4 mRNAs while rather repressing HDAC7. UC cell lines overexpressing HDAC4 were not significantly less sensitive to 19i. Reevaluation of the in vitro HDAC isoenzyme activity inhibition profile of 19i and its docking to HDAC4 using current assays suggested rather low activity against class IIA HDACs. The specific class IIA HDAC inhibitor TMP269 impeded proliferation of UC cell lines only at concentrations > 10 µM. CONCLUSIONS: Anti-neoplastic effects of 19i on UC cells appear to be exerted by targeting class I HDACs. In fact, HDAC4 may rather impede UC growth. Our results suggest that targeting of class IIA HDACs 4/5 may not be optimal for UC therapy. Moreover, our investigation provides further evidence for cross-regulation of class IIA HDACs by class I HDACs.


Subject(s)
Carcinoma, Transitional Cell/genetics , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/genetics , Urinary Bladder Neoplasms/genetics , Vorinostat/pharmacology , Carcinoma, Transitional Cell/drug therapy , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , DNA Methylation , Drug Screening Assays, Antitumor , Gene Expression Regulation, Neoplastic/drug effects , HEK293 Cells , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylases/chemistry , Humans , Molecular Docking Simulation , Urinary Bladder Neoplasms/drug therapy , Vorinostat/chemistry
14.
Virology ; 522: 158-167, 2018 09.
Article in English | MEDLINE | ID: mdl-30032029

ABSTRACT

The MX dynamin GTPases inhibit diverse viruses at early post-entry phases. While MXA acts antiviral against influenza viruses, the anti HIV-1 activity of MXB was discovered recently. Here, we have studied the antiviral effect of MX proteins on murine cytomegalovirus (MCMV). Our data demonstrate that human MXB but not other human or murine MX proteins inhibit MCMV propagation. Evidently, the viral protein expression was delayed and the viral DNA amount in nucleus was diminished in MXB expressing cells indicating an obstruction of nuclear entry. Of note, MCMV did not deplete MX proteins. Considering the role of capsid on HIV-1 sensitivity to MXB, MXB binding to tested MCMV capsids was not detected. Moreover, MCMV restriction occurred only when MXB contained both the nuclear localization signal and a functional GTPase domain. Hence, we propose a new mode of inhibition of MCMV by MXB that is conspicuously different from that of HIV-1.


Subject(s)
Host-Pathogen Interactions , Immunologic Factors/metabolism , Muromegalovirus/immunology , Muromegalovirus/physiology , Myxovirus Resistance Proteins/metabolism , Virus Replication , HEK293 Cells , Humans , Virus Internalization
15.
Methods Mol Biol ; 1655: 97-107, 2018.
Article in English | MEDLINE | ID: mdl-28889380

ABSTRACT

Members of the APOBEC3 (A3) family of enzymes were shown to act in an oncogenic manner in several cancer types. Immunodetection of APOBEC3A (A3A), APOBEC3B (A3B), and APOBEC3G (A3G) proteins is particularly challenging due to the large sequence homology of these proteins and limited availability of antibodies. Here we combine independent immunoblotting with an in vitro activity assay technique, to detect and categorize specific A3s expressed in urothelial bladder cancer and other cancer cells.


Subject(s)
Cytosine Deaminase/metabolism , Urologic Neoplasms/metabolism , APOBEC Deaminases , Catalysis , Cell Line , Cytidine Deaminase , Cytosine Deaminase/genetics , DNA/metabolism , Enzyme Activation , Humans , Immunoblotting/methods , Multigene Family , Mutation , Urinary Bladder Neoplasms/diagnosis , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/metabolism , Urologic Neoplasms/diagnosis , Urologic Neoplasms/genetics
16.
J Mol Biol ; 429(8): 1171-1191, 2017 04 21.
Article in English | MEDLINE | ID: mdl-28315663

ABSTRACT

The retroviral restriction factors of the APOBEC3 (A3) cytidine deaminase family catalyze the deamination of cytidines in single-stranded viral DNA. APOBEC3C (A3C) is a strong antiviral factor against viral infectivity factor (vif)-deficient simian immunodeficiency virus Δvif, which is, however, a weak inhibitor against human immunodeficiency virus (HIV)-1 for reasons unknown. The precise link between the antiretroviral effect of A3C and its catalytic activity is incompletely understood. Here, we show that the S61P mutation in human A3C (A3C.S61P) boosted hypermutation in the viral genomes of simian immunodeficiency virus Δvif and murine leukemia virus but not in human immunodeficiency virus HIV-1Δvif. The enhanced antiviral activity of A3C.S61P correlated with enhanced in vitro cytidine deamination. Furthermore, the S61P mutation did not change the substrate specificity of A3C, ribonucleoprotein complex formation, self-association, Zinc coordination, or viral incorporation features. We propose that local structural changes induced by the serine-to-proline substitution are responsible for the gain of catalytic activity of A3C.S61P. Our results are a first step toward an understanding of A3C's DNA binding capacity, deamination-dependent editing, and antiviral functions at the molecular level. We conclude that the enhanced enzymatic activity of A3C is insufficient to restrict HIV-1, indicating an unknown escape mechanism of HIV-1.


Subject(s)
Cytidine Deaminase/chemistry , Cytidine Deaminase/metabolism , HIV-1/pathogenicity , Amino Acid Substitution , Animals , Cytidine Deaminase/genetics , Cytosine/metabolism , DNA, Single-Stranded/chemistry , DNA, Single-Stranded/metabolism , DNA, Viral/metabolism , HEK293 Cells/virology , HIV-1/genetics , Host-Pathogen Interactions , Humans , Leukemia Virus, Murine/metabolism , Leukemia Virus, Murine/pathogenicity , Pan troglodytes , Protein Conformation , Simian Immunodeficiency Virus/metabolism , Simian Immunodeficiency Virus/pathogenicity , Zinc/metabolism , vif Gene Products, Human Immunodeficiency Virus/genetics , vif Gene Products, Human Immunodeficiency Virus/metabolism
17.
J Virol ; 90(22): 10193-10208, 2016 Nov 15.
Article in English | MEDLINE | ID: mdl-27581978

ABSTRACT

Lentiviruses have evolved the Vif protein to counteract APOBEC3 (A3) restriction factors by targeting them for proteasomal degradation. Previous studies have identified important residues in the interface of human immunodeficiency virus type 1 (HIV-1) Vif and human APOBEC3C (hA3C) or human APOBEC3F (hA3F). However, the interaction between primate A3C proteins and HIV-1 Vif or natural HIV-1 Vif variants is still poorly understood. Here, we report that HIV-1 Vif is inactive against A3Cs of rhesus macaques (rhA3C), sooty mangabey monkeys (smmA3C), and African green monkeys (agmA3C), while HIV-2, African green monkey simian immunodeficiency virus (SIVagm), and SIVmac Vif proteins efficiently mediate the depletion of all tested A3Cs. We identified that residues N/H130 and Q133 in rhA3C and smmA3C are determinants for this HIV-1 Vif-triggered counteraction. We also found that the HIV-1 Vif interaction sites in helix 4 of hA3C and hA3F differ. Vif alleles from diverse HIV-1 subtypes were tested for degradation activities related to hA3C. The subtype F-1 Vif was identified to be inactive for degradation of hA3C and hA3F. The residues that determined F-1 Vif inactivity in the degradation of A3C/A3F were located in the C-terminal region (K167 and D182). Structural analysis of F-1 Vif revealed that impairing the internal salt bridge of E171-K167 restored reduction capacities to A3C/A3F. Furthermore, we found that D101 could also form an internal interaction with K167. Replacing D101 with glycine and R167 with lysine in NL4-3 Vif impaired its counteractivity to A3F and A3C. This finding indicates that internal interactions outside the A3 binding region in HIV-1 Vif influence the capacity to induce degradation of A3C/A3F. IMPORTANCE: The APOBEC3 restriction factors can serve as potential barriers to lentiviral cross-species transmissions. Vif proteins from lentiviruses counteract APOBEC3 by proteasomal degradation. In this study, we found that monkey-derived A3C, rhA3C and smmA3C, were resistant to HIV-1 Vif. This was determined by A3C residues N/H130 and Q133. However, HIV-2, SIVagm, and SIVmac Vif proteins were found to be able to mediate the depletion of all tested primate A3C proteins. In addition, we identified a natural HIV-1 Vif (F-1 Vif) that was inactive in the degradation of hA3C/hA3F. Here, we provide for the first time a model that explains how an internal salt bridge of E171-K167-D101 influences Vif-mediated degradation of hA3C/hA3F. This finding provides a novel way to develop HIV-1 inhibitors by targeting the internal interactions of the Vif protein.


Subject(s)
Cytidine Deaminase/metabolism , Gene Products, vif/metabolism , HIV-1/metabolism , Simian Immunodeficiency Virus/metabolism , vif Gene Products, Human Immunodeficiency Virus/metabolism , Animals , Binding Sites , Cell Line , HEK293 Cells , HIV Infections/virology , HIV-2/metabolism , Humans , Lentivirus/metabolism , Macaca mulatta , Protein Binding/physiology
18.
Retrovirology ; 13(1): 46, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27368163

ABSTRACT

BACKGROUND: Feline immunodeficiency virus (FIV) is a global pathogen of Felidae species and a model system for Human immunodeficiency virus (HIV)-induced AIDS. In felids such as the domestic cat (Felis catus), APOBEC3 (A3) genes encode for single-domain A3Z2s, A3Z3 and double-domain A3Z2Z3 anti-viral cytidine deaminases. The feline A3Z2Z3 is expressed following read-through transcription and alternative splicing, introducing a previously untranslated exon in frame, encoding a domain insertion called linker. Only A3Z3 and A3Z2Z3 inhibit Vif-deficient FIV. Feline A3s also are restriction factors for HIV and Simian immunodeficiency viruses (SIV). Surprisingly, HIV-2/SIV Vifs can counteract feline A3Z2Z3. RESULTS: To identify residues in feline A3s that Vifs need for interaction and degradation, chimeric human-feline A3s were tested. Here we describe the molecular direct interaction of feline A3s with Vif proteins from cat FIV and present the first structural A3 model locating these interaction regions. In the Z3 domain we have identified residues involved in binding of FIV Vif, and their mutation blocked Vif-induced A3Z3 degradation. We further identified additional essential residues for FIV Vif interaction in the A3Z2 domain, allowing the generation of FIV Vif resistant A3Z2Z3. Mutated feline A3s also showed resistance to the Vif of a lion-specific FIV, indicating an evolutionary conserved Vif-A3 binding. Comparative modelling of feline A3Z2Z3 suggests that the residues interacting with FIV Vif have, unlike Vif-interacting residues in human A3s, a unique location at the domain interface of Z2 and Z3 and that the linker forms a homeobox-like domain protruding of the Z2Z3 core. HIV-2/SIV Vifs efficiently degrade feline A3Z2Z3 by possible targeting the linker stretch connecting both Z-domains. CONCLUSIONS: Here we identified in feline A3s residues important for binding of FIV Vif and a unique protein domain insertion (linker). To understand Vif evolution, a structural model of the feline A3 was developed. Our results show that HIV Vif binds human A3s differently than FIV Vif feline A3s. The linker insertion is suggested to form a homeo-box domain, which is unique to A3s of cats and related species, and not found in human and mouse A3s. Together, these findings indicate a specific and different A3 evolution in cats and human.


Subject(s)
Cytidine Deaminase/chemistry , Cytidine Deaminase/metabolism , Gene Products, vif/metabolism , HIV-1/metabolism , Immunodeficiency Virus, Feline/metabolism , Animals , Cats , Cell Line , Cytidine Deaminase/genetics , Evolution, Molecular , Gene Products, vif/genetics , Genes, Homeobox , HIV-1/genetics , Humans , Immunodeficiency Virus, Feline/genetics , Models, Molecular , Recombinant Fusion Proteins/metabolism
19.
PLoS One ; 11(6): e0155422, 2016.
Article in English | MEDLINE | ID: mdl-27249646

ABSTRACT

APOBEC4 (A4) is a member of the AID/APOBEC family of cytidine deaminases. In this study we found a high mRNA expression of A4 in human testis. In contrast, there were only low levels of A4 mRNA detectable in 293T, HeLa, Jurkat or A3.01 cells. Ectopic expression of A4 in HeLa cells resulted in mostly cytoplasmic localization of the protein. To test whether A4 has antiviral activity similar to that of proteins of the APOBEC3 (A3) subfamily, A4 was co-expressed in 293T cells with wild type HIV-1 and HIV-1 luciferase reporter viruses. We found that A4 did not inhibit the replication of HIV-1 but instead enhanced the production of HIV-1 in a dose-dependent manner and seemed to act on the viral LTR. A4 did not show detectable cytidine deamination activity in vitro and weakly interacted with single-stranded DNA. The presence of A4 in virus producer cells enhanced HIV-1 replication by transiently transfected A4 or stably expressed A4 in HIV-susceptible cells. APOBEC4 was capable of similarly enhancing transcription from a broad spectrum of promoters, regardless of whether they were viral or mammalian. We hypothesize that A4 may have a natural role in modulating host promoters or endogenous LTR promoters.


Subject(s)
Cytidine Deaminase/physiology , HIV-1/physiology , Virus Replication/physiology , Cell Line , Cytidine/metabolism , Cytidine Deaminase/metabolism , Deamination , HIV Long Terminal Repeat , Humans , Male , Promoter Regions, Genetic , Testis/metabolism
20.
Virology ; 488: 51-60, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26609934

ABSTRACT

Foamy viruses (FV) are retroviruses that are widely distributed in primate and non-primate animal species. We tested here FV with capsids of simian and non-simian origin for sensitivity to interferon-ß (IFN-ß). Our data show significant inhibition of FV by IFN-ß early in infection of human HOS and THP-1 but not of HEK293T cells. The post-entry restriction of FV was not mediated by the interferon-induced MxB protein that was recently identified as a capsid-interacting restriction factor targeting Human immunodeficiency virus (HIV) before integration. Neither the ectopic expression of MxA or MxB in HEK293T cells nor the lack of MxB expression in CRISPR/CAS MxB THP-1 knockout cells impacted the infection of the tested FV. IFN-ß treated THP-1 and THP-1 KO MxB cells showed the same extend of restriction to FV. Together, the data demonstrate that IFN-ß inhibits FV early in infection and that MxB is not a restriction factor of FV.


Subject(s)
Interferon-beta/metabolism , Myxovirus Resistance Proteins/metabolism , Spumavirus/immunology , Cell Line , Humans , Myxovirus Resistance Proteins/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL