Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 177
1.
Viruses ; 13(11)2021 11 19.
Article En | MEDLINE | ID: mdl-34835112

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.


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
2.
J Mol Biol ; 433(18): 167111, 2021 09 03.
Article En | MEDLINE | ID: mdl-34153286

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.


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
3.
Biotechnol Prog ; 36(3): e2953, 2020 05.
Article En | MEDLINE | ID: mdl-31846227

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.


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
4.
J Immunol ; 199(4): 1372-1381, 2017 08 15.
Article En | MEDLINE | ID: mdl-28687662

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.


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
5.
Biotechnol Bioeng ; 114(4): 813-820, 2017 04.
Article En | MEDLINE | ID: mdl-27800626

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.


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
6.
Oncotarget ; 7(44): 71255-71273, 2016 Nov 01.
Article En | MEDLINE | ID: mdl-27655726

The mechanism by which type II interferon (IFN) inhibits virus replications remains to be identified. Murine leukemia virus (MLV) replication was significantly restricted by γ-IFN, but not human immunodeficiency virus type 1 (HIV-1) replication. Because MLV enters host cells via endosomes, we speculated that certain cellular factors among γ-IFN-induced, endosome-localized proteins inhibit MLV replication. We found that γ-IFN-inducible lysosomal thiolreductase (GILT) significantly restricts HIV-1 replication as well as MLV replication by its thiolreductase activity. GILT silencing enhanced replication-defective HIV-1 vector infection and virion production in γ-IFN-treated cells, although γ-IFN did not inhibit HIV-1 replication. This result showed that GILT is required for the anti-viral activity of γ-IFN. Interestingly, GILT protein level was increased by γ-IFN in uninfected cells and env-deleted HIV-1-infected cells, but not in full-length HIV-1-infected cells. γ-IFN-induced transcription from the γ-IFN-activation sequence was attenuated by the HIV-1 Env protein. These results suggested that the γ-IFN cannot restrict HIV-1 replication due to the inhibition of γ-IFN signaling by HIV-1 Env. Finally, we found that 4,4'-dithiodipyridine (4-PDS), which inhibits S-S bond formation at acidic pH, significantly suppresses HIV-1 vector infection and virion production, like GILT. In conclusion, this study showed that GILT functions as a host restriction factor against the retroviruses, and a GILT mimic, 4-PDS, is the leading compound for the development of novel concept of anti-viral agents.


Anti-Retroviral Agents/pharmacology , HIV-1/physiology , Oxidoreductases Acting on Sulfur Group Donors/physiology , Animals , COS Cells , Chlorocebus aethiops , Dithionitrobenzoic Acid/pharmacology , Gene Products, env/physiology , HIV-1/drug effects , Humans , Interferon-gamma/pharmacology , Leukemia Virus, Murine/drug effects , Leukemia Virus, Murine/physiology , Mice , Tetraspanin 30/physiology , Virion/physiology , Virus Replication/drug effects
8.
Proc Natl Acad Sci U S A ; 110(30): 12379-84, 2013 Jul 23.
Article En | MEDLINE | ID: mdl-23836649

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.


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
9.
Int J Biol Macromol ; 53: 122-6, 2013 Feb.
Article En | MEDLINE | ID: mdl-23153762

This study is to investigate the synergistic effect of Anglica polysaccharide sulfate (APS-1) and Combivir, an anti-AIDS drug, on murine leukemia virus in vivo. As the results shown, the virus replication was significantly decreased by the combination of APS-1 and Combivir, which tended to be further decreased (58% inhibition) when compared with that of Combivir alone (51% inhibition). Furthermore, both the percentage of CD4(+) cells and CD4(+)/CD8(+) ratio in peripheral blood cells were significantly enhanced by this combined administration, while the CD4(+) cells was only slightly increased and CD4(+)/CD8(+) ratio was not affected by Combivir alone. Additionally, combination of APS-1 and Combivir also alleviated the toxicity of Combivir. APS-1 not only increased the survival rate of mice administered with LD(50) dose of Combivir, but also reduced the hematologic toxicity induced by Combivir, RBC, HGB and PLT were restored to normal level. These results suggest that APS-1 had synergistic effect with Combivir, which provided new insight into the potential clinical use of polysaccharide sulfate in anti-AIDS field.


Anti-HIV Agents/pharmacology , Lamivudine/pharmacology , Leukemia Virus, Murine/physiology , Polysaccharides/pharmacology , Virus Replication/drug effects , Zidovudine/pharmacology , Animals , Anti-HIV Agents/toxicity , Bone Marrow Cells/drug effects , CD4-CD8 Ratio , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/virology , Cell Line, Tumor , Drug Combinations , Drug Evaluation, Preclinical , Drug Synergism , Drug Therapy, Combination , Humans , Lamivudine/toxicity , Lethal Dose 50 , Leukemia Virus, Murine/drug effects , Mice , Mice, Inbred BALB C , Organ Size/drug effects , Polysaccharides/toxicity , Thymus Gland/drug effects , Thymus Gland/pathology , Viral Load , Zidovudine/toxicity
10.
Retrovirology ; 9: 17, 2012 Feb 20.
Article En | MEDLINE | ID: mdl-22348230

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.


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
11.
Int J Biol Macromol ; 50(3): 768-72, 2012 Apr 01.
Article En | MEDLINE | ID: mdl-22155400

This study is to synthesize sulfated Angelica polysaccharides (APSs) and investigate the activity of one of the sulfated derivatives APS-1 on murine leukemia virus in vivo. Six sulfated derivatives with degree of sulfation ranging from 0.68 to 1.91 were obtained. And the virus replication was inhibited by APS-1 at the dose of 10 and 30 mg/kg (26% and 30% inhibition respectively). Furthermore, both the percentage of CD4(+) cells and CD4(+)/CD8(+) ratio in peripheral blood cells were significantly enhanced by APS-1 at 3-30 mg/kg. In addition, the reduced thymus/body weight index by murine leukemia virus infection was increased by ASP-1 in a dose dependent manner. These results suggest that APS-1 could not only inhibit virus replication, but also improve the immune function. APS-1 may be a potential new and better antiviral drug.


Angelica sinensis/chemistry , Polysaccharides/chemistry , Polysaccharides/pharmacology , Sulfates/chemistry , Animals , Antiviral Agents/chemistry , Antiviral Agents/isolation & purification , Antiviral Agents/pharmacology , Immunologic Factors/chemistry , Immunologic Factors/isolation & purification , Immunologic Factors/pharmacology , Leukemia Virus, Murine/drug effects , Lymphocytes/drug effects , Lymphocytes/immunology , Lymphocytes/metabolism , Mice , Polysaccharides/isolation & purification
12.
PLoS One ; 6(1): e15840, 2011 Jan 14.
Article En | MEDLINE | ID: mdl-21264291

Replication of retroviral and host genomes requires ribonucleotide reductase to convert rNTPs to dNTPs, which are then used as substrates for DNA synthesis. Inhibition of ribonucleotide reductase by hydroxyurea (HU) has been previously used to treat cancers as well as HIV. However, the use of HU as an antiretroviral is limited by its associated toxicities such as myelosuppression and hepatotoxicity. In this study, we examined the ribonucleotide reductase inhibitor, gemcitabine, both in cell culture and in C57Bl/6 mice infected with LP-BM5 murine leukemia virus (LP-BM5 MuLV, a murine AIDS model). Gemcitabine decreased infectivity of MuLV in cell culture with an EC50 in the low nanomolar range with no detectable cytotoxicity. Similarly, gemcitabine significantly decreased disease progression in mice infected with LP-BM5. Specifically, gemcitabine treatment decreased spleen size, plasma IgM, and provirus levels compared to LP-BM5 MuLV infected, untreated mice. Gemcitabine efficacy was observed at doses as low as 1 mg/kg/day in the absence of toxicity. Higher doses of gemcitabine (3 mg/kg/day and higher) were associated with toxicity as determined by a loss in body mass. In summary, our findings demonstrate that gemcitabine has antiretroviral activity ex vivo and in vivo in the LP-BM5 MuLV model. These observations together with a recent ex vivo study with HIV-1, suggest that gemcitabine has broad antiretroviral activity and could be particularly useful in vivo when used in combination drug therapy.


Anti-Retroviral Agents/pharmacology , Deoxycytidine/analogs & derivatives , Leukemia Virus, Murine/drug effects , Murine Acquired Immunodeficiency Syndrome/drug therapy , Animals , Cells, Cultured , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Disease Progression , Immunoglobulin M/blood , Mice , Mice, Inbred C57BL , Ribonucleotide Reductases/antagonists & inhibitors , Spleen/pathology , Viral Load/drug effects , Gemcitabine
13.
Virology ; 410(1): 234-9, 2011 Feb 05.
Article En | MEDLINE | ID: mdl-21131013

Xenotropic murine leukemia virus-related virus (XMRV) is a novel retrovirus, related to murine leukemia virus (MLV), that has been implicated in human disease. If XMRV is indeed able to replicate in humans, then one might predict that XMRV would have developed resistance to human innate antiviral resistance factors such as APOBEC3G (hA3G). In fact, we observed that XMRV and MLV are both highly sensitive to inhibition by hA3G and equally resistant to inhibition by murine APOBEC3. While several human prostate cancer cell lines were found to lack hA3G, stable expression of physiological levels of hA3G rendered these cells refractory to XMRV replication. Few human tissues fail to express hA3G, and we therefore hypothesize that XMRV replicates in one or more hA3G-negative reservoir tissues and/or that human XMRV infections are likely to be rare and potentially of zoonotic origin.


Cytosine Deaminase , Xenotropic murine leukemia virus-related virus/physiology , APOBEC Deaminases , Animals , Cell Line, Tumor , Cytidine Deaminase , Cytosine Deaminase/classification , Cytosine Deaminase/genetics , Gene Expression Regulation , Humans , Leukemia Virus, Murine/drug effects , Leukemia Virus, Murine/genetics , Leukemia Virus, Murine/physiology , Mutation , Xenotropic murine leukemia virus-related virus/genetics
14.
Nat Rev Urol ; 7(7): 392-402, 2010 Jul.
Article En | MEDLINE | ID: mdl-20517289

Xenotropic murine leukemia virus-related virus (XMRV) is an authentic, newly recognized human retrovirus first identified in prostate cancer tissues from men with a deficiency in the innate immunity gene RNASEL. At present, studies have detected XMRV at widely different rates in prostate cancer cases (0-27%) and in patients with chronic fatigue syndrome (CFS; 0-67%). Indirect or direct modes of carcinogenesis by XMRV have been suggested depending on whether the virus was found in stroma or malignant epithelium. Viral replication in the prostate might be affected by androgens, which stimulate XMRV through a transcriptional enhancer site in viral DNA. By contrast, host restriction factors, such as APOBEC3 and tetherin, inhibit virus replication. Immune dysfunction mediated by XMRV has been suggested as a possible factor in CFS. Recent studies show that some existing antiretroviral drugs suppress XMRV infections and diagnostic assays are under development. Although other retroviruses of the same genus as XMRV (gammaretroviruses) cause cancer and neurological disease in animals, whether XMRV is a cause of either prostate cancer or CFS remains unknown. Emerging science surrounding XMRV is contributing to our knowledge of retroviral infections while focusing intense interest on two major human diseases.


Fatigue Syndrome, Chronic/virology , Prostatic Neoplasms/virology , Retroviridae Infections/virology , Tumor Virus Infections/virology , Anti-Retroviral Agents/pharmacology , Anti-Retroviral Agents/therapeutic use , Fatigue Syndrome, Chronic/immunology , Fatigue Syndrome, Chronic/therapy , Female , Gammaretrovirus/drug effects , Gammaretrovirus/immunology , Gammaretrovirus/pathogenicity , Humans , Leukemia Virus, Murine/drug effects , Leukemia Virus, Murine/immunology , Leukemia Virus, Murine/pathogenicity , Male , Prostatic Neoplasms/immunology , Prostatic Neoplasms/therapy , Retroviridae/drug effects , Retroviridae/immunology , Retroviridae/pathogenicity , Retroviridae Infections/immunology , Retroviridae Infections/therapy , Tumor Virus Infections/immunology , Tumor Virus Infections/therapy
15.
PLoS One ; 5(4): e9948, 2010 Apr 01.
Article En | MEDLINE | ID: mdl-20376347

BACKGROUND: Xenotropic murine leukemia-related retrovirus (XMRV) is a recently discovered retrovirus that has been linked to human prostate cancer and chronic fatigue syndrome (CFS). Both diseases affect a large fraction of the world population, with prostate cancer affecting one in six men, and CFS affecting an estimated 0.4 to 1% of the population. PRINCIPAL FINDINGS: Forty-five compounds, including twenty-eight drugs approved for use in humans, were evaluated against XMRV replication in vitro. We found that the retroviral integrase inhibitor, raltegravir, was potent and selective against XMRV at submicromolar concentrations, in MCF-7 and LNCaP cells, a breast cancer and prostate cancer cell line, respectively. Another integrase inhibitor, L-000870812, and two nucleoside reverse transcriptase inhibitors, zidovudine (ZDV), and tenofovir disoproxil fumarate (TDF) also inhibited XMRV replication. When combined, these drugs displayed mostly synergistic effects against this virus, suggesting that combination therapy may delay or prevent the selection of resistant viruses. CONCLUSIONS: If XMRV proves to be a causal factor in prostate cancer or CFS, these discoveries may allow for rational design of clinical trials.


Drug Evaluation, Preclinical/methods , Fatigue Syndrome, Chronic/virology , Gammaretrovirus/drug effects , Prostatic Neoplasms/virology , Pyrrolidinones/pharmacology , Retroviridae Infections/drug therapy , Tumor Virus Infections/drug therapy , Antiviral Agents , Cell Line, Tumor , Female , Humans , Leukemia Virus, Murine/drug effects , Male , Microbial Sensitivity Tests , Pyrrolidinones/therapeutic use , Raltegravir Potassium , Virus Replication/drug effects
16.
Comb Chem High Throughput Screen ; 13(4): 352-7, 2010 May.
Article En | MEDLINE | ID: mdl-20156147

Cell-based assays for the inhibition of viral infections most commonly couple a positive signal (e.g., an increase in fluorescence) to the infection itself and not to its inhibition. When performing drug screens, compounds decreasing the signal are therefore considered as putative inhibitors. However, this approach can cause the selection of many false positives, since, for example, both killing of the host cell and inhibiting viral cell-entry results in the same signal. Using a model system based on murine leukemia virus (MLV) particles pseudotyped with the G-protein of vesicular stomatitis virus (VSV-G), we have developed generic assays coupling a positive readout to the inhibition of viral transduction. Consequently, the system favors drug candidates (and concentrations thereof) that do not harm human cells and significantly decreases the probability for selecting false positives. The assay allows Z-factors of approximately 0.9, takes cytotoxic side effects into account and could in theory be adapted for high-throughput screening of inhibitors against further viral species.


Antiviral Agents/pharmacology , Leukemia Virus, Murine/drug effects , Vesiculovirus/drug effects , Base Sequence , Cell Line , Fluorescence , Humans , Oligonucleotides
17.
Virology ; 396(2): 272-9, 2010 Jan 20.
Article En | MEDLINE | ID: mdl-19913868

Retroviral DNA integration leaves behind a single-strand DNA discontinuity at each virus:host DNA junction. It has long been proposed that cellular proteins detect and repair the integrated DNA and that failure to do so might lead to apoptotic cell death, but their identity remains unknown. PIKK family members ATM, DNA-PKcs and ATR have all been proposed to be important for HIV-1 replication, but these findings turned out to be very controversial. In order to clarify their role in retroviral replication, we analyzed the effect of pharmacological inhibitors and of a dominant-negative version of ATR on the replication of retroviruses in cell lines relevant to HIV-1 infection. Our data show that ATR and probably other PIKKs as well are involved in retroviral replication in some but not all cell lines and that ATR increases the frequency of retroviral transduction by a mechanism other than the enhancement of infected cell survival.


Cell Cycle Proteins/physiology , Protein Serine-Threonine Kinases/physiology , Retroviridae Infections/virology , Retroviridae/physiology , Virus Replication/physiology , Androstadienes/pharmacology , Animals , Ataxia Telangiectasia Mutated Proteins , Caffeine/pharmacology , Cell Line , Cell Transformation, Viral/drug effects , HIV Infections/virology , HIV-1/drug effects , HIV-1/physiology , HIV-2/drug effects , HIV-2/physiology , Leukemia Virus, Murine/drug effects , Leukemia Virus, Murine/physiology , Leukemia, Experimental/virology , Protein Kinase Inhibitors/pharmacology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/drug effects , Simian Immunodeficiency Virus/physiology , Transduction, Genetic , Tumor Virus Infections/virology , Virus Replication/drug effects , Wortmannin
18.
Proc Natl Acad Sci U S A ; 106(49): 20865-70, 2009 Dec 08.
Article En | MEDLINE | ID: mdl-19923437

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.


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
19.
Virology ; 394(2): 227-34, 2009 Nov 25.
Article En | MEDLINE | ID: mdl-19781728

Recently it has been reported that a cathepsin B inhibitor, CA-074Me, attenuates ecotropic murine leukemia virus (Eco-MLV) infection in NIH3T3 cells, suggesting that cathepsin B is required for the Eco-MLV infection. However, cathepsin B activity was negative or extremely low in NIH3T3 cells. How did CA-074Me attenuate the Eco-MLV infection? The CA-074Me treatment of NIH3T3 cells inhibited cathepsin L activity, and a cathepsin L specific inhibitor, CLIK148, attenuated the Eco-MLV vector infection. These results indicate that the suppression of cathepsin L activity by CA-074Me induces the inhibition of Eco-MLV infection, suggesting that cathepsin L is required for the Eco-MLV infection in NIH3T3 cells. The CA-074Me treatment inhibited the Eco-MLV infection in human cells expressing the exogenous mouse ecotropic receptor and endogenous cathepsins B and L, but the CLIK148 treatment did not, showing that only the cathepsin L suppression by CLIK148 is not enough to prevent the Eco-MLV infection in cells expressing both of cathepsins B and L, and CA-074Me inhibits the Eco-MLV infection by suppressing both of cathepsins B and L. These results suggest that either cathepsin B or L is sufficient for the Eco-MLV infection.


Cathepsin L/physiology , Leukemia Virus, Murine/enzymology , Animals , Base Sequence , Cathepsin B/antagonists & inhibitors , Cathepsin B/genetics , Cathepsin B/physiology , Cathepsin L/antagonists & inhibitors , Cell Line , Cysteine Proteinase Inhibitors/pharmacology , Dipeptides/pharmacology , Epoxy Compounds/pharmacology , Host-Pathogen Interactions/drug effects , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/physiology , Humans , Leukemia Virus, Murine/drug effects , Leukemia Virus, Murine/pathogenicity , Leukemia, Experimental/etiology , Leukemia, Experimental/prevention & control , Membrane Glycoproteins/genetics , Membrane Glycoproteins/physiology , Mice , NIH 3T3 Cells , Pyridines/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Receptors, Virus/genetics , Receptors, Virus/physiology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Retroviridae Infections/etiology , Retroviridae Infections/prevention & control , Tumor Virus Infections/etiology , Tumor Virus Infections/prevention & control
20.
Retrovirology ; 5: 59, 2008 Jul 09.
Article En | MEDLINE | ID: mdl-18613956

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.


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
...