Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 177
Filter
Add more filters

Publication year range
1.
J Immunol ; 199(4): 1372-1381, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28687662

ABSTRACT

Accumulating evidence shows that innate immune responses are associated with extracellular nucleotides, particularly ATP. In this article, we demonstrate extensive protection of ATP/P2X7 signaling in a host against viral infection. Interestingly, we observed a significant increase in ATP as a danger signal in vesicular stomatitis virus (VSV)-infected cell supernatant and VSV-infected mice in an exocytosis- and pannexin channel-dependent manner. Furthermore, extracellular ATP reduces the replication of VSV, Newcastle disease virus, murine leukemia virus, and HSV in vivo and in vitro through the P2X7 receptor. Meanwhile, ATP significantly increases IFN-ß expression in a concentration- and time-dependent manner. Mechanistically, ATP facilitates IFN-ß secretion through P38/JNK/ATF-2 signaling pathways, which are crucial in promoting antiviral immunity. Taken together, these results demonstrate the protective role of extracellular ATP and P2X7 in viral infection and suggest a potential therapeutic role for ATP/P2X7 in viral diseases.


Subject(s)
Adenosine Triphosphate/metabolism , Interferon-beta/biosynthesis , Receptors, Purinergic P2X7/metabolism , Vesicular Stomatitis/immunology , Vesicular stomatitis Indiana virus/physiology , Adenosine Triphosphate/pharmacology , Animals , Immunity, Innate , Interferon-beta/genetics , Interferon-beta/immunology , Leukemia Virus, Murine/drug effects , Leukemia Virus, Murine/immunology , Luminescent Measurements , Mice , Newcastle disease virus/drug effects , Newcastle disease virus/immunology , RAW 264.7 Cells , Receptors, Purinergic P2X7/immunology , Signal Transduction , Simplexvirus/drug effects , Simplexvirus/immunology , Vesicular Stomatitis/virology , Vesicular stomatitis Indiana virus/drug effects , Vesicular stomatitis Indiana virus/immunology , Virus Replication/drug effects
2.
Biotechnol Bioeng ; 114(4): 813-820, 2017 04.
Article in English | MEDLINE | ID: mdl-27800626

ABSTRACT

Inclusion of a detergent in protein biotherapeutic purification processes is a simple and very robust method for inactivating enveloped viruses. The detergent Triton X-100 has been used for many years and is part of the production process of several commercial therapeutic proteins. However, recent ecological studies have suggested that Triton X-100 and its break-down products can potentially behave as endocrine disrupters in aquatic organisms, raising concerns from an environmental impact perspective. As such, discharge of Triton X-100 into the waste water treatment plants is regulated in some jurisdictions, and alternative detergents for viral inactivation are required. In this work, we report on the identification and evaluation of more eco-friendly detergents as viable replacements for Triton X-100. Five detergent candidates with low to moderate environmental impact were initially identified and evaluated with respect to protein stability, followed by proof-of-concept virus inactivation studies using a model enveloped virus. From the set of candidates lauryldimethylamine N-oxide (LDAO) was identified as the most promising detergent due to its low ecotoxicity, robust anti-viral activity (LRV >4 at validation set-point conditions with X-MuLX), and absence of any negative impact on protein function. This detergent exhibited effective and robust virus inactivation in a broad range of protein concentrations, solution conductivities, pHs, and in several different cell culture fluid matrices. The only process parameter which correlated with reduced virus inactivation potency was LDAO concentration, and then only when the concentration was reduced to below the detergent's critical micelle concentration (CMC). Additionally, this work also demonstrated that LDAO was cleared to below detectable levels after Protein A affinity chromatography, making it suitable for use in a platform process that utilizes this chromatographic mode for protein capture. All these findings suggest that LDAO may be a practical alternative to Triton X-100 for use in protein therapeutic production processes for inactivating enveloped viruses. Biotechnol. Bioeng. 2017;114: 813-820. © 2016 Wiley Periodicals, Inc.


Subject(s)
Detergents/chemistry , Detergents/pharmacology , Dimethylamines/chemistry , Dimethylamines/pharmacology , Virus Inactivation/drug effects , Green Chemistry Technology , Herpesvirus 1, Suid/drug effects , Leukemia Virus, Murine/drug effects , Models, Molecular , Octoxynol/chemistry , Octoxynol/pharmacology
3.
Proc Natl Acad Sci U S A ; 110(30): 12379-84, 2013 Jul 23.
Article in English | MEDLINE | ID: mdl-23836649

ABSTRACT

When host cells are infected by an RNA virus, pattern-recognition receptors (PRRs) recognize the viral RNA and induce the antiviral innate immunity. Toll-like receptor 7 (TLR7) detects the genomic RNA of incoming murine leukemia virus (MLV) in endosomes and mediates the antiviral response. However, the RNA-sensing PRR that recognizes the MLV in the cytosol is not fully understood. Here, we definitively demonstrate that zinc-finger antiviral protein (ZAP) acts as a cytosolic RNA sensor, inducing the degradation of the MLV transcripts by the exosome, an RNA degradation system, on RNA granules. Although the retinoic acid inducible gene I (RIG-I)-like receptors (RLRs) RIG-I and melanoma differentiation-associated protein 5 detect various RNA viruses in the cytosol and induce the type I IFN-dependent antiviral response, RLR loss does not alter the replication efficiency of MLV. In sharp contrast, the loss of ZAP greatly enhances the replication efficiency of MLV. ZAP localizes to RNA granules, where the processing-body and stress-granule proteins assemble. ZAP induces the recruitment of the MLV transcripts and exosome components to the RNA granules. The CCCH-type zinc-finger domains of ZAP, which are RNA-binding motifs, mediate its localization to RNA granules and MLV transcripts degradation by the exosome. Although ZAP was known as a regulator of RIG-I signaling in a human cell line, ZAP deficiency does not affect the RIG-I-dependent production of type I IFN in mouse cells. Thus, ZAP is a unique member of the cytosolic RNA-sensing PRR family that targets and eliminates intracellular RNA viruses independently of TLR and RLR family members.


Subject(s)
Antiviral Agents/pharmacology , DEAD-box RNA Helicases/physiology , Leukemia Virus, Murine/drug effects , Zinc Fingers , Animals , Cells, Cultured , DEAD Box Protein 58 , Leukemia Virus, Murine/physiology , Mice , Mice, Inbred C57BL , Virus Replication/drug effects
4.
Retrovirology ; 9: 17, 2012 Feb 20.
Article in English | MEDLINE | ID: mdl-22348230

ABSTRACT

BACKGROUND: Ankyrins are cellular mediators of a number of essential protein-protein interactions. Unlike intrabodies, ankyrins are composed of highly structured repeat modules characterized by disulfide bridge-independent folding. Artificial ankyrin molecules, designed to target viral components, might act as intracellular antiviral agents and contribute to the cellular immunity against viral pathogens such as HIV-1. RESULTS: A phage-displayed library of artificial ankyrins was constructed, and screened on a polyprotein made of the fused matrix and capsid domains (MA-CA) of the HIV-1 Gag precursor. An ankyrin with three modules named Ank(GAG)1D4 (16.5 kDa) was isolated. Ank(GAG)1D4 and MA-CA formed a protein complex with a stoichiometry of 1:1 and a dissociation constant of K(d) ~ 1 µM, and the Ank(GAG)1D4 binding site was mapped to the N-terminal domain of the CA, within residues 1-110. HIV-1 production in SupT1 cells stably expressing Ank(GAG)1D4 in both N-myristoylated and non-N-myristoylated versions was significantly reduced compared to control cells. Ank(GAG)1D4 expression also reduced the production of MLV, a phylogenetically distant retrovirus. The Ank(GAG)1D4-mediated antiviral effect on HIV-1 was found to occur at post-integration steps, but did not involve the Gag precursor processing or cellular trafficking. Our data suggested that the lower HIV-1 progeny yields resulted from the negative interference of Ank(GAG)1D4-CA with the Gag assembly and budding pathway. CONCLUSIONS: The resistance of Ank(GAG)1D4-expressing cells to HIV-1 suggested that the CA-targeted ankyrin Ank(GAG)1D4 could serve as a protein platform for the design of a novel class of intracellular inhibitors of HIV-1 assembly based on ankyrin-repeat modules.


Subject(s)
Ankyrins/pharmacology , Anti-HIV Agents/pharmacology , HIV-1/drug effects , gag Gene Products, Human Immunodeficiency Virus/antagonists & inhibitors , Amino Acid Sequence , Cell Line , HIV-1/growth & development , Humans , Leukemia Virus, Murine/drug effects , Leukemia Virus, Murine/growth & development , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Interaction Mapping , Recombinant Proteins/pharmacology , Virus Assembly/drug effects , Virus Replication/drug effects
5.
Proc Natl Acad Sci U S A ; 106(49): 20865-70, 2009 Dec 08.
Article in English | MEDLINE | ID: mdl-19923437

ABSTRACT

Raltegravir is a recently, Food and Drug Administration-approved, small-molecule drug that inhibits retroviral integrase, thereby preventing HIV DNA from inserting itself into the human genome. We report here that the activity profile of raltegravir on the replication of murine leukemia virus is similar to that for HIV, and that the drug specifically affects autoimmune disease in mice, in which endogenous retroelements are suspected to play a role. While NZW and BALB/c mice, which do not succumb to autoimmune disease, are not affected by raltegravir, lupus-prone (NZBxNZW) F(1) mice die of glomerulonephritis more than a month earlier than untreated mice. Raltegravir-treated NZB mice, which share the H-2 haplotype with BALB/c mice, but which are predisposed to autoimmune hemolytic anemia, develop auto-antibodies to their red blood cells >3 months earlier than untreated mice of the same strain. Because nonautoimmune mice are not affected by raltegravir, we consider off-target effects unlikely and attribute the exacerbation of autoimmunity to the inhibition of retroviral integrase.


Subject(s)
Autoimmune Diseases/chemically induced , HIV Integrase Inhibitors/adverse effects , Pyrrolidinones/adverse effects , Amino Acid Sequence , Animals , Antibody Formation/drug effects , Autoimmune Diseases/complications , Base Sequence , DNA, Circular/genetics , DNA, Complementary/genetics , Disease Susceptibility/complications , Exodeoxyribonucleases/metabolism , Female , HIV Integrase Inhibitors/pharmacology , Kidney Diseases/chemically induced , Kidney Diseases/complications , Leukemia Virus, Murine/drug effects , Leukemia Virus, Murine/genetics , Lupus Erythematosus, Systemic/complications , Male , Mice , Molecular Sequence Data , Phosphoproteins/metabolism , Pyrrolidinones/pharmacology , Raltegravir Potassium , Sequence Deletion , Terminal Repeat Sequences/genetics , Time Factors , Viral Envelope Proteins/chemistry , Virus Integration/drug effects
6.
Trends Biochem Sci ; 32(3): 118-28, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17303427

ABSTRACT

The APOBEC family of cytidine deaminases inhibit the mobility of diverse retroviruses, retrotransposons and other viruses. Initial reports proposed that these effects were due to the DNA editing capabilities of these enzymes; however, many recent studies have provided evidence suggesting that APOBEC proteins can inhibit these elements by several mechanisms, including editing-dependent and editing-independent processes. Investigating these modes of action and the potential contribution that each one makes to the antiviral activities of various APOBEC proteins is vital if we are to understand how APOBEC proteins protect host genomes from invading nucleic acids.


Subject(s)
Antiviral Agents , Cytidine Deaminase/metabolism , Virus Replication/drug effects , APOBEC-1 Deaminase , APOBEC-3G Deaminase , Animals , Anti-HIV Agents/pharmacology , DNA, Viral/metabolism , Deltaretrovirus/drug effects , Gene Products, vif/metabolism , HIV Infections/prevention & control , Humans , Leukemia Virus, Murine/drug effects , Nucleoside Deaminases/metabolism , Proteins/metabolism , Repressor Proteins/metabolism , Retroelements/physiology , Species Specificity , Spumavirus/drug effects , vif Gene Products, Human Immunodeficiency Virus
7.
Viruses ; 13(11)2021 11 19.
Article in English | MEDLINE | ID: mdl-34835112

ABSTRACT

Anti-viral small molecules are currently lacking for treating coronavirus infection. The long development timescales for such drugs are a major problem, but could be shortened by repurposing existing drugs. We therefore screened a small library of FDA-approved compounds for potential severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) antivirals using a pseudovirus system that allows a sensitive read-out of infectivity. A group of structurally-related compounds, showing moderate inhibitory activity with IC50 values in the 2-5 µM range, were identified. Further studies demonstrated that these "kite-shaped" molecules were surprisingly specific for SARS-CoV-1 and SARS-CoV-2 and that they acted early in the entry steps of the viral infectious cycle, but did not affect virus attachment to the cells. Moreover, the compounds were able to prevent infection in both kidney- and lung-derived human cell lines. The structural homology of the hits allowed the production of a well-defined pharmacophore that was found to be highly accurate in predicting the anti-viral activity of the compounds in the screen. We discuss the prospects of repurposing these existing drugs for treating current and future coronavirus outbreaks.


Subject(s)
Antiviral Agents/pharmacology , COVID-19 Drug Treatment , COVID-19/virology , Leukemia Virus, Murine/drug effects , SARS-CoV-2 , Virus Internalization/drug effects , Animals , Cell Line , Chlorocebus aethiops , Drug Discovery/methods , Drug Repositioning , Drug Synergism , Humans , Leukemia Virus, Murine/metabolism , Mice , Molecular Docking Simulation , Spike Glycoprotein, Coronavirus/metabolism , Vero Cells , Virus Attachment/drug effects
8.
J Mol Biol ; 433(18): 167111, 2021 09 03.
Article in English | MEDLINE | ID: mdl-34153286

ABSTRACT

5-aza-cytidine (5-aza-C) has been shown to be a potent human immunodeficiency virus type 1 (HIV-1) mutagen that induces G-to-C hypermutagenesis by incorporation of the reduced form (i.e., 5-aza-dC, 5-aza-dCTP). Evidence to date suggests that this lethal mutagenesis is the primary antiretroviral mechanism for 5-aza-C. To investigate the breadth of application of 5-aza-C as an antiretroviral mutagen, we have conducted a comparative, parallel analysis of the antiviral mechanism of 5-aza-C between HIV-1 and gammaretroviruses - i.e., murine leukemia virus (MuLV) and feline leukemia virus (FeLV). Intriguingly, in contrast to the hallmark G-to-C hypermutagenesis observed with HIV-1, MuLV and FeLV did not reveal the presence of a significant increase in mutational burden, particularly that of G-to-C transversion mutations. The effect of 5-aza-dCTP on DNA synthesis revealed that while HIV-1 RT was not inhibited by 5-aza-dCTP even at 100 µM, 5-aza-dCTP was incorporated and significantly inhibited MuLV RT, generating pause sites and reducing the fully extended product. 5-aza-dCTP was found to be incorporated into DNA by MuLV RT or HIV-1 RT, but only acted as a non-obligate chain terminator for MuLV RT. This biochemical data provides an independent line of experimental evidence in support of the conclusion that HIV-1 and MuLV have distinct primary mechanisms of antiretroviral action with 5-aza-C. Taken together, our data provides striking evidence that an antiretroviral mutagen can have strong potency via distinct mechanisms of action among closely related viruses, unlinking antiviral activity from antiviral mechanism of action.


Subject(s)
Antiviral Agents/pharmacology , Azacitidine/analogs & derivatives , Cytidine Triphosphate/analogs & derivatives , HIV Infections/drug therapy , Leukemia, Experimental/drug therapy , Mutation/drug effects , Retroviridae Infections/drug therapy , Tumor Virus Infections/drug therapy , Animals , Azacitidine/pharmacology , Cats , Cytidine Triphosphate/pharmacology , HIV/drug effects , HIV Infections/virology , Humans , Leukemia Virus, Feline/drug effects , Leukemia Virus, Murine/drug effects , Leukemia, Experimental/virology , Mice , Mutagenesis , Mutagens , Retroviridae Infections/virology , Tumor Virus Infections/virology , Virus Replication
9.
J Cell Biol ; 170(2): 317-25, 2005 Jul 18.
Article in English | MEDLINE | ID: mdl-16027225

ABSTRACT

Viruses have often been observed in association with the dense microvilli of polarized epithelia as well as the filopodia of nonpolarized cells, yet whether interactions with these structures contribute to infection has remained unknown. Here we show that virus binding to filopodia induces a rapid and highly ordered lateral movement, "surfing" toward the cell body before cell entry. Virus cell surfing along filopodia is mediated by the underlying actin cytoskeleton and depends on functional myosin II. Any disruption of virus cell surfing significantly reduces viral infection. Our results reveal another example of viruses hijacking host machineries for efficient infection by using the inherent ability of filopodia to transport ligands to the cell body.


Subject(s)
Actins/physiology , Avian Leukosis Virus/physiology , Leukemia Virus, Murine/physiology , Myosins/physiology , Pseudopodia/physiology , Animals , Avian Leukosis Virus/drug effects , Avian Leukosis Virus/ultrastructure , Cell Line , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Cell Membrane/virology , Cytochalasin D/pharmacology , Heterocyclic Compounds, 4 or More Rings/pharmacology , Humans , Leukemia Virus, Murine/drug effects , Leukemia Virus, Murine/ultrastructure , Mice , Microscopy, Electron , Pseudopodia/ultrastructure , Pseudopodia/virology
10.
Biotechnol Prog ; 36(3): e2953, 2020 05.
Article in English | MEDLINE | ID: mdl-31846227

ABSTRACT

Triton X-100 has long been used either alone or in combination with solvent to inactivate enveloped viruses in biopharmaceutical manufacturing. However, European Chemicals Agency (ECHA) officially placed Triton X-100 on the Annex XIV authorization list in 2017 because 4-(1,1,3,3-tetramethylbutyl) phenol, a degradation product of Triton X-100, is of harmful endocrine disrupting activities. As a result, any use of Triton X-100 in the European Economic Area would require an ECHA issued authorization after the sunset date of January 4, 2021. In search of possible replacements for Triton X-100, we discovered that polysorbate 80 (PS80) in absence of any solvents was able to effectively inactive enveloped viruses such as xenotropic murine leukemia virus and pseudorabies virus with comparable efficacy as measured by log reduction factors. Interestingly, PS80 did not show any virucidal activities in phosphate buffered saline (PBS) while achieving robust virus inactivation in cell-free Chinese hamster ovary (CHO) bioreactor harvests. This intriguing observation led us to speculate that virus inactivation by PS80 involved components in the cell-free CHO bioreactor harvests that were absent in PBS. Specifically, we hypothesized that esterase and/or lipases in the cell-free bioreactor harvests hydrolyzed PS80 to yield oleic acid, a known potent virucidal agent, which in turn inactivated viruses. This theory was confirmed using purified recombinant lysosomal phospholipase A2 isomer (rLPLA2) in PBS. Subsequent characterization work has indicated that virus inactivation by PS80 is effective and robust within temperature and concentration ranges comparable to those of Triton X-100. Similar to Triton X-100, virus inactivation by PS80 is dually dependent on treatment time and temperature. Unlike Triton X-100, PS80 inactivation does not correlate with concentrations in a simple manner. Additionally, we have demonstrated that PS20 exhibits similar virus inactivation activities as PS80. Based on the findings described in the current work, we believe that PS80 is potentially a viable replacement for Triton X-100 and can be used in manufacturing processes for wide spectrum of biopharmaceuticals to achieve desirable virus clearance. Finally, the advantages and disadvantages of using PS80 for virus inactivation are discussed in the contexts of GMP manufacturing.


Subject(s)
Cell-Free System , Leukemia Virus, Murine/drug effects , Polysorbates/pharmacology , Virus Inactivation/drug effects , Animals , CHO Cells , Cricetinae , Cricetulus , Detergents/chemistry , Detergents/pharmacology , Hydrolysis/drug effects , Kinetics , Leukemia Virus, Murine/pathogenicity , Mice , Octoxynol , Solvents/chemistry
11.
J Virol ; 82(2): 764-74, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17977962

ABSTRACT

Integrase (IN), an essential enzyme of human immunodeficiency virus (HIV), is an attractive antiretroviral drug target. The antiviral activity and resistance profile in vitro of a novel IN inhibitor, elvitegravir (EVG) (also known as JTK-303/GS-9137), currently being developed for the treatment of HIV-1 infection are described. EVG blocked the integration of HIV-1 cDNA through the inhibition of DNA strand transfer. EVG inhibited the replication of HIV-1, including various subtypes and multiple-drug-resistant clinical isolates, and HIV-2 strains with a 50% effective concentration in the subnanomolar to nanomolar range. EVG-resistant variants were selected in two independent inductions, and a total of 8 amino acid substitutions in the catalytic core domain of IN were observed. Among the observed IN mutations, T66I and E92Q substitutions mainly contributed to EVG resistance. These two primary resistance mutations are located in the active site, and other secondary mutations identified are proximal to these primary mutations. The EVG-selected IN mutations, some of which represent novel IN inhibitor resistance mutations, conferred reduced susceptibility to other IN inhibitors, suggesting that a common mechanism is involved in resistance and potential cross-resistance. The replication capacity of EVG-resistant variants was significantly reduced relative to both wild-type virus and other IN inhibitor-resistant variants selected by L-870,810. EVG and L-870,810 both inhibited the replication of murine leukemia virus and simian immunodeficiency virus, suggesting that IN inhibitors bind to a conformationally conserved region of various retroviral IN enzymes and are an ideal drug for a range of retroviral infections.


Subject(s)
Anti-HIV Agents/pharmacology , Drug Resistance, Viral/genetics , HIV-1/drug effects , HIV-2/drug effects , Integrase Inhibitors/pharmacology , Quinolones/pharmacology , Amino Acid Substitution/genetics , Binding Sites , Humans , Leukemia Virus, Murine/drug effects , Microbial Sensitivity Tests , Simian Immunodeficiency Virus/drug effects , Virus Replication/genetics
12.
J Virol ; 82(9): 4665-70, 2008 May.
Article in English | MEDLINE | ID: mdl-18305045

ABSTRACT

We recently identified the cytoskeletal regulatory protein moesin as a novel gene that inhibits retroviral replication prior to reverse transcription by downregulation of stable microtubule formation. Here, we provide evidence that overexpression of ezrin, another closely related ezrin-radixin-moesin (ERM) family member, also blocks replication of both murine leukemia viruses and human immunodeficiency virus type 1 (HIV-1) in Rat2 fibroblasts before reverse transcription, while knockdown of endogenous ezrin increases the susceptibility of human cells to HIV-1 infection. Together, these results suggest that ERM proteins may be important determinants of retrovirus susceptibility through negative regulation of stable microtubule networks.


Subject(s)
Cytoskeletal Proteins/administration & dosage , Cytoskeletal Proteins/physiology , Microtubules/drug effects , Retroviridae Infections/drug therapy , Animals , Disease Susceptibility/therapy , HIV-1/drug effects , Humans , Leukemia Virus, Murine/drug effects , Membrane Proteins , Mice , Microfilament Proteins , RNA, Small Interfering/pharmacology , Rats , Transfection , Virus Replication/drug effects
13.
Science ; 173(3995): 441-3, 1971 Jul 30.
Article in English | MEDLINE | ID: mdl-4326720

ABSTRACT

Ethidium bromide, compared on a molar basis, was a more effective inhibitor of the DNA polymerases of the Rauscher and Moloney murine leukemia viruses than either 4-N-demethylrifampicin or 4-N-benzyldemethylrifampicin. Daunomycin inhibited the polymerases weakly, and chromomycin A(3) inhibited almost not at all. 4-N-Benzyldemethylrifampicin was a more active inhibitor than the 4-N-demethyl congener.


Subject(s)
DNA Nucleotidyltransferases/antagonists & inhibitors , Daunorubicin/pharmacology , Leukemia Virus, Murine/drug effects , Leukemia Virus, Murine/enzymology , Phenanthridines/pharmacology , Rifampin/pharmacology , Animals , Anti-Bacterial Agents/pharmacology , Detergents/pharmacology , Drug Synergism , Nucleotides/metabolism , Rats , Thymidine/metabolism
14.
Retrovirology ; 5: 59, 2008 Jul 09.
Article in English | MEDLINE | ID: mdl-18613956

ABSTRACT

BACKGROUND: TRIM5 alpha is a restriction factor that interferes with retroviral infections in a species-specific manner in primate cells. Although TRIM5 alpha is constitutively expressed, its expression has been shown to be up-regulated by type I interferon (IFN). Among primates, a particular case exists in owl monkey cells, which express a fusion protein between TRIM5 and cyclophilin A, TRIMCyp, specifically interfering with HIV-1 infection. No studies have been conducted so far concerning the possible induction of TRIMCyp by IFN. We investigated the consequences of IFN treatment on retroviral restriction in diverse primate cells and evaluated the implication of TRIM5 alpha or TRIMCyp in IFN-induced anti-retroviral activities. RESULTS: First, we show that human type I IFN can enhance TRIM5 alpha expression in human, African green monkey and macaque cells, as well as TRIMCyp expression in owl monkey cells. In TRIM5 alpha-expressing primate cell lines, type I IFN has little or no effect on HIV-1 infection, whereas it potentiates restriction activity against N-MLV in human and African green monkey cells. In contrast, type I IFN treatment of owl monkey cells induces a great enhancement of HIV-1 restriction, as well as a strain-tropism independent restriction of MLV. We were able to demonstrate that TRIM5 alpha is the main mediator of the IFN-induced activity against N-MLV in human and African green monkey cells, whereas TRIMCyp mediates the IFN-induced HIV-1 restriction enhancement in owl monkey cells. In contrast, the type I IFN-induced anti-MLV restriction in owl monkey cells is independent of TRIMCyp expression. CONCLUSION: Together, our observations indicate that both TRIM5 alpha and TRIMCyp are implicated in IFN-induced anti-retroviral response in primate cells. Furthermore, we found that type I IFN also induces a TRIMCyp-independent restriction activity specific to MLV in owl monkey cells.


Subject(s)
Antiviral Agents/pharmacology , Cyclophilin A/metabolism , HIV-1/drug effects , Interferons/pharmacology , Leukemia Virus, Murine/drug effects , Proteins/metabolism , Animals , Antiviral Restriction Factors , Aotus trivirgatus , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line , Chlorocebus aethiops , Cyclophilin A/genetics , Gene Expression Regulation , HIV-1/pathogenicity , HeLa Cells , Humans , Leukemia Virus, Murine/pathogenicity , Macaca mulatta , Proteins/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Tripartite Motif Proteins , Ubiquitin-Protein Ligases , Vero Cells
15.
Antiviral Res ; 77(2): 120-7, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18164447

ABSTRACT

Antioxidant molecules can be used both to replenish the depletion of reduced glutathione (GSH) occurring during HIV infection, and to inhibit HIV replication. The purpose of this work was to assess the efficacy of two pro-GSH molecules able to cross the cell membrane more easily than GSH. We used an experimental animal model consisting of C57BL/6 mice infected with the LP-BM5 viral complex; the treatments were based on the intramuscular administration of I-152, a pro-drug of N-acetylcysteine and S-acetyl-beta-mercaptoethylamine, and S-acetylglutathione, an acetylated GSH derivative. The results show that I-152, at a concentration of 10.7 times lower than GSH, caused a reduction in lymph node and spleen weights of about 55% when compared to infected animals and an inhibition of about 66% in spleen and lymph node virus content. S-acetylglutathione, at half the concentration of GSH, caused a reduction in lymph node weight of about 17% and in spleen and lymph node virus content of about 70% and 30%, respectively. These results show that the administration of pro-GSH molecules may favorably substitute for the use of GSH as such.


Subject(s)
Acetylcysteine/analogs & derivatives , Anti-HIV Agents/therapeutic use , Cysteamine/analogs & derivatives , Glutathione/analogs & derivatives , Murine Acquired Immunodeficiency Syndrome/drug therapy , Prodrugs/therapeutic use , Acetylcysteine/pharmacology , Acetylcysteine/therapeutic use , Animals , Anti-HIV Agents/pharmacology , Cell Proliferation/drug effects , Cysteamine/pharmacology , Cysteamine/therapeutic use , DNA, Viral/drug effects , DNA, Viral/genetics , Disease Models, Animal , Female , Glutathione/pharmacology , Glutathione/therapeutic use , Hypergammaglobulinemia/drug therapy , Immunoglobulin G/blood , Leukemia Virus, Murine/drug effects , Leukemia Virus, Murine/genetics , Leukemia Virus, Murine/isolation & purification , Lymph Nodes/drug effects , Lymph Nodes/physiopathology , Lymphocytes/drug effects , Mice , Mice, Inbred C57BL , Organ Size/drug effects , Polymerase Chain Reaction , Prodrugs/pharmacology , Spleen/drug effects , Spleen/physiopathology
16.
Retrovirology ; 4: 68, 2007 Sep 24.
Article in English | MEDLINE | ID: mdl-17892575

ABSTRACT

BACKGROUND: TRIM5alpha, which is expressed in most primates and the related TRIMCyp, which has been found in one of the New World monkey species, are antiviral proteins of the TRIM5 family that are able to intercept incoming retroviruses early after their entry into cells. The mechanism of action has been partially elucidated for TRIM5alpha, which seems to promote premature decapsidation of the restricted retroviruses. In addition, through its N-terminal RING domain, TRIM5alpha may sensitize retroviruses to proteasome-mediated degradation. TRIM5alpha-mediated restriction requires a physical interaction with the capsid protein of targeted retroviruses. It is unclear whether other cellular proteins are involved in the inhibition mediated by TRIM5alpha and TRIMCyp. A previous report suggested that the inhibition of HIV-1 by the rhesus macaque orthologue of TRIM5alpha was inefficient in the D17a canine cell line, suggesting that the cellular environment was important for the restriction mechanism. Here we investigated further the behavior of TRIM5alpha and TRIMCyp in the D17 cells. RESULTS: We found that the various TRIM5alpha orthologues studied (human, rhesus macaque, African green monkey) as well as TRIMCyp had poor antiviral activity in the D17 cells, despite seemingly normal expression levels and subcellular distribution. Restriction of both HIV-1 and the distantly related N-tropic murine leukemia virus (N-MLV) was low in D17 cells. Both TRIM5alpharh and TRIMCyp promoted early HIV-1 decapsidation in murine cells, but weak levels of restriction in D17 cells correlated with the absence of accelerated decapsidation in these cells and also correlated with normal levels of cDNA synthesis. Fv1, a murine restriction factor structurally unrelated to TRIM5alpha, was fully functional in D17 cells, showing that the loss of activity was specific to TRIM5alpha/TRIMCyp. CONCLUSION: We show that D17 cells provide a poor environment for the inhibition of retroviral replication by proteins of the TRIM5 family. Because both TRIM5alpha and TRIMCyp are poorly active in these cells, despite having quite different viral target recognition domains, we conclude that a step either upstream or downstream of target recognition is impaired. We speculate that an unknown factor required for TRIM5alpha and TRIMCyp activity is missing or inadequately expressed in D17 cells.


Subject(s)
Antiviral Agents/pharmacology , Cyclophilin A/metabolism , HIV-1/drug effects , Leukemia Virus, Murine/drug effects , Proteins/pharmacology , Animals , Anti-HIV Agents/pharmacology , Cell Line , Cyclophilin A/genetics , Dogs , Leukemia Virus, Murine/pathogenicity , Ubiquitin-Protein Ligases
17.
Microbes Infect ; 9(14-15): 1590-6, 2007.
Article in English | MEDLINE | ID: mdl-18023391

ABSTRACT

Fv-4 is a mouse gene that confers resistance against ecotropic murine leukemia virus (MLV) infection on mice. While receptor interference by the Fv-4 env gene product, Fv-4 Env, that can bind to the ecotropic MLV receptor has been shown to play an important role in the resistance, other mechanisms have also been suggested because it confers extremely efficient, complete resistance in vivo. Here, we have examined the effect of Fv-4 Env on infectious MLV production. Infectious MLV titers in supernatants obtained after transfection with a Friend MLV (FMLV) Env-expressing plasmid from MLV gag-pol producer cells harboring a retroviral vector were largely reduced by coexpression of Fv-4 Env. Syncytia formation mediated by R-peptide-deleted FMLV Env in NIH 3T3 cells was impaired by Fv-4 Env coexpression. Similarly, Fv-4 Env inhibited infectious amphotropic MLV production and syncytia formation mediated by R-peptide-deleted amphotropic MLV Env. Immunoprecipitation analysis revealed interaction of Fv-4 Env with amphotropic MLV Env as well as FMLV Env. These results indicate that Fv-4 Env inhibits infectious ecotropic and amphotropic MLV production by exerting dominant negative effect on MLV Env, suggesting contribution of this inhibitory effect to the resistance against ecotropic MLV infection in Fv-4-expressing mice.


Subject(s)
Gene Products, env/metabolism , Leukemia Virus, Murine/metabolism , Leukemia Virus, Murine/pathogenicity , Membrane Proteins/metabolism , Membrane Proteins/pharmacology , Animals , Cell Line , DNA, Viral/genetics , DNA, Viral/metabolism , Gene Products, env/genetics , Giant Cells/metabolism , Immunoprecipitation , Leukemia Virus, Murine/drug effects , Membrane Proteins/genetics , Mice , NIH 3T3 Cells , Transfection , Virion/metabolism
18.
Nucleic Acids Res ; 33(6): 1913-23, 2005.
Article in English | MEDLINE | ID: mdl-15809227

ABSTRACT

Human APOBEC3F and APOBEC3G are double-domained deaminases that can catalyze dC-->dU deamination in HIV-1 and MLV retroviral DNA replication intermediates, targeting T-C or C-C dinucleotides, respectively. HIV-1 antagonizes their action through its vif gene product, which has been shown (at least in the case of APOBEC3G) to interact with the N-terminal domain of the deaminase, triggering its degradation. Here, we compare APOBEC3F and APOBEC3G to APOBEC3C, a single-domained deaminase that can also act on both HIV-1 and MLV. We find that whereas APOBEC3C contains all the information necessary for both Vif-binding and cytidine deaminase activity in a single domain, it is the C-terminal domain of APOBEC3F and APOBEC3G that confer their target site specificity for cytidine deamination. We have exploited the fact that APOBEC3C, whilst highly homologous to the C-terminal domain of APOBEC3F, exhibits a distinct target site specificity (preferring Y-C dinucleotides) in order to identify residues in APOBEC3F that might affect its target site specificity. We find that this specificity can be altered by single amino acid substitutions at several distinct positions, suggesting that the strong dependence of APOBEC3-mediated deoxycytidine deamination on the 5'-flanking nucleotide is sensitive to relatively subtle changes in the APOBEC3 structure. The approach has allowed the isolation of APOBEC3 DNA mutators that exhibit novel target site preferences.


Subject(s)
Anti-Retroviral Agents/chemistry , Anti-Retroviral Agents/pharmacology , Cytidine Deaminase/chemistry , Cytidine Deaminase/pharmacology , DNA, Viral/drug effects , APOBEC-3G Deaminase , Amino Acid Substitution , Cytidine Deaminase/genetics , Cytosine Deaminase/chemistry , Cytosine Deaminase/genetics , Cytosine Deaminase/pharmacology , DNA Mutational Analysis , DNA, Viral/chemistry , DNA, Viral/metabolism , Humans , Leukemia Virus, Murine/drug effects , Leukemia Virus, Murine/genetics , Nucleoside Deaminases , Proteins/chemistry , Proteins/genetics , Proteins/pharmacology , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Repressor Proteins , Substrate Specificity
19.
Antiviral Res ; 72(2): 153-6, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16647765

ABSTRACT

We have isolated a substance with anti-retroviral activity from the freshly prepared crude extract of Chelidonium majus L. (greater celandine) by 9-aminoacridine precipitation method and ion exchange chromatography using Dowex-50W/H+ resin followed by the gel filtration on Sephadex-75 column. Elemental and phenol/sulfuric acid method analyses as well as the mass spectrometry of the purified substance indicated that it may represent a low-sulfated poly-glycosaminoglycan moiety with molecular weight of approximately 3800 Da. The substance prevented infection of human CD4+ T-cell lines AA2 and H9 with HIV-1 at concentration of 25 microg/mL as well as the cell-to-cell virus spread in H9 cells continuously infected with HIV-1, as determined by the measurement of reverse transcriptase activity and p24 content in cell cultures. Furthermore, we have shown in a murine AIDS model that the treatment with purified substance significantly prevented splenomegaly and the enlargement of cervical lymph nodes in C57Bl/6 mice chronically infected with the pool of murine leukemia retroviruses. The mechanism(s) of anti-retroviral activity of this substance have to be elucidated.


Subject(s)
Anti-HIV Agents/pharmacology , Anti-Retroviral Agents/pharmacology , Chelidonium/chemistry , Glycosaminoglycans/pharmacology , HIV-1/drug effects , Plant Extracts/isolation & purification , Plant Extracts/pharmacology , Animals , Anti-HIV Agents/chemistry , Anti-HIV Agents/isolation & purification , Anti-Retroviral Agents/chemistry , Anti-Retroviral Agents/isolation & purification , CD4-Positive T-Lymphocytes/virology , Cell Line , Chromatography, Gel , Chromatography, Ion Exchange , Disease Models, Animal , Fractional Precipitation , Glycosaminoglycans/chemistry , Glycosaminoglycans/isolation & purification , HIV Core Protein p24/analysis , HIV Reverse Transcriptase/analysis , HIV-1/physiology , Humans , Leukemia Virus, Murine/drug effects , Mass Spectrometry , Mice , Mice, Inbred C57BL , Molecular Weight , Plant Extracts/chemistry , Retroviridae Infections/drug therapy , Tumor Virus Infections/drug therapy
20.
Virol J ; 3: 27, 2006 Apr 07.
Article in English | MEDLINE | ID: mdl-16603064

ABSTRACT

BACKGROUND: Self-inactivating retroviral vectors (SIN) are often associated with very low titers. Promoter elements embedded within SIN designs may suppress transcription of packageable retroviral RNA which in turn results in titer reduction. We tested whether this dominant-negative effect involves histone acetylation state. We designed an MLV-derived SIN vector using the cytomegalovirus immediate early enhancer-promoter (CMVIE) as an embedded internal promoter (SINCMV) and transfected the pantropic 293GPG packaging cell line. RESULTS: The SINCMV retroviral producer had uniformly very low titers (approximately 10,000 infectious retroparticles per ml). Northern blot showed low levels of expression of retroviral mRNA in producer cells in particular that of packageable RNA transcript. Treatment of the producers with the histone deacetylase (HDAC) inhibitors sodium butyrate and trichostatin A reversed transcriptional suppression and resulted in an average 106.3 +/- 4.6 - fold (P = 0.002) and 15.5 +/- 1.3 - fold increase in titer (P = 0.008), respectively. A histone gel assay confirmed increased histone acetylation in treated producer cells. CONCLUSION: These results show that SIN retrovectors incorporating strong internal promoters such as CMVIE, are susceptible to transcriptional silencing and that treatment of the producer cells with HDAC inhibitors can overcome this blockade suggesting that histone deacetylation is implicated in the mechanism of transcriptional suppression.


Subject(s)
Genetic Vectors/genetics , Histones/metabolism , Leukemia Virus, Murine/genetics , Leukemia Virus, Murine/physiology , Transfection/methods , Acetylation/drug effects , Butyrates/pharmacology , Cell Line , Gene Expression Regulation/drug effects , Genetic Engineering , Humans , Hydroxamic Acids/pharmacology , Leukemia Virus, Murine/drug effects , RNA, Viral/metabolism , Transcription, Genetic/drug effects , Virus Replication/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL