Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
J Virol ; 93(6)2019 03 15.
Article in English | MEDLINE | ID: mdl-30602605

ABSTRACT

Pattern recognition receptors provide essential nonself immune surveillance within distinct cellular compartments. Retinoic acid-inducible gene I (RIG-I) is one of the primary cytosolic RNA sensors, with an emerging role in the nucleus. It is involved in the spatiotemporal sensing of influenza A virus (IAV) replication, leading to the induction of type I interferons (IFNs). Nonetheless, the physiological viral ligands activating RIG-I during IAV infection remain underexplored. Other than full-length viral genomes, cellular constraints that impede ongoing viral replication likely potentiate an erroneous viral polymerase generating aberrant viral RNA species with RIG-I-activating potential. Here, we investigate the origins of RIG-I-activating viral RNA under two such constraints. Using chemical inhibitors that inhibit continuous viral protein synthesis, we identify the incoming, but not de novo-synthesized, viral defective interfering (DI) genomes contributing to RIG-I activation. In comparison, deprivation of viral nucleoprotein (NP), the key RNA chain elongation factor for the viral polymerase, leads to the production of aberrant viral RNA species activating RIG-I; however, their nature is likely to be distinct from that of DI RNA. Moreover, RIG-I activation in response to NP deprivation is not adversely affected by expression of the nuclear export protein (NEP), which diminishes the generation of a major subset of aberrant viral RNA but facilitates the accumulation of small viral RNA (svRNA). Overall, our results indicate the existence of fundamentally different mechanisms of RIG-I activation under cellular constraints that impede ongoing IAV replication.IMPORTANCE The induction of an IFN response by IAV is mainly mediated by the RNA sensor RIG-I. The physiological RIG-I ligands produced during IAV infection are not fully elucidated. Cellular constraints leading to the inhibition of ongoing viral replication likely potentiate an erroneous viral polymerase producing aberrant viral RNA species activating RIG-I. Here, we demonstrate that RIG-I activation during chemical inhibition of continuous viral protein synthesis is attributable to the incoming DI genomes. Erroneous viral replication driven by NP deprivation promotes the generation of RIG-I-activating aberrant viral RNA, but their nature is likely to be distinct from that of DI RNA. Our results thus reveal distinct mechanisms of RIG-I activation by IAV under cellular constraints impeding ongoing viral replication. A better understanding of RIG-I sensing of IAV infection provides insight into the development of novel interventions to combat influenza virus infection.


Subject(s)
DEAD Box Protein 58/genetics , Influenza A virus/growth & development , Influenza, Human/virology , Virus Replication/genetics , A549 Cells , Animals , Cell Line , Cell Line, Tumor , Cells , Defective Viruses/drug effects , Defective Viruses/genetics , Dogs , Genome, Viral/drug effects , Genome, Viral/genetics , HEK293 Cells , Humans , Influenza A virus/drug effects , Influenza, Human/drug therapy , Interferon Type I/pharmacology , Madin Darby Canine Kidney Cells , Nucleoproteins/genetics , Orthomyxoviridae Infections/drug therapy , Orthomyxoviridae Infections/virology , RNA, Viral/genetics , Receptors, Immunologic , Virus Replication/drug effects
2.
J Clin Invest ; 128(9): 4074-4085, 2018 08 31.
Article in English | MEDLINE | ID: mdl-30024859

ABSTRACT

HIV posttreatment controllers (PTCs) represent a natural model of sustained HIV remission, but they are rare and little is known about their viral reservoir. We obtained 1,450 proviral sequences after near-full-length amplification for 10 PTCs and 16 posttreatment noncontrollers (NCs). Before treatment interruption, the median intact and total reservoir size in PTCs was 7-fold lower than in NCs, but the proportion of intact, defective, and total clonally expanded proviral genomes was not significantly different between the 2 groups. Quantification of total but not intact proviral genome copies predicted sustained HIV remission as 81% of NCs, but none of the PTCs had a total proviral genome greater than 4 copies per million peripheral blood mononuclear cells (PBMCs). The results highlight the restricted intact and defective HIV reservoir in PTCs and suggest that total proviral genome burden could act as the first biomarker for identifying PTCs. Total and defective but not intact proviral copy numbers correlated with levels of cell-associated HIV RNA, activated NK cell percentages, and both HIV-specific CD4+ and CD8+ responses. These results support the concept that defective HIV genomes can lead to viral antigen production and interact with both the innate and adaptive immune systems.


Subject(s)
HIV Infections/virology , HIV Long-Term Survivors , HIV-1/genetics , Proviruses/genetics , Adult , Anti-HIV Agents/therapeutic use , Defective Viruses/drug effects , Defective Viruses/genetics , Defective Viruses/isolation & purification , Disease Reservoirs/virology , Female , Genome, Viral , HIV Infections/drug therapy , HIV-1/drug effects , HIV-1/isolation & purification , Humans , Male , Middle Aged , Proviruses/drug effects , Proviruses/isolation & purification , Viral Load/drug effects , Viral Load/genetics
3.
PLoS Comput Biol ; 12(5): e1004799, 2016 05.
Article in English | MEDLINE | ID: mdl-27152856

ABSTRACT

The rapid evolution of RNA-encoded viruses such as HIV presents a major barrier to infectious disease control using conventional pharmaceuticals and vaccines. Previously, it was proposed that defective interfering particles could be developed to indefinitely control the HIV/AIDS pandemic; in individual patients, these engineered molecular parasites were further predicted to be refractory to HIV's mutational escape (i.e., be 'resistance-proof'). However, an outstanding question has been whether these engineered interfering particles-termed Therapeutic Interfering Particles (TIPs)-would remain resistance-proof at the population-scale, where TIP-resistant HIV mutants may transmit more efficiently by reaching higher viral loads in the TIP-treated subpopulation. Here, we develop a multi-scale model to test whether TIPs will maintain indefinite control of HIV at the population-scale, as HIV ('unilaterally') evolves toward TIP resistance by limiting the production of viral proteins available for TIPs to parasitize. Model results capture the existence of two intrinsic evolutionary tradeoffs that collectively prevent the spread of TIP-resistant HIV mutants in a population. First, despite their increased transmission rates in TIP-treated sub-populations, unilateral TIP-resistant mutants are shown to have reduced transmission rates in TIP-untreated sub-populations. Second, these TIP-resistant mutants are shown to have reduced growth rates (i.e., replicative fitness) in both TIP-treated and TIP-untreated individuals. As a result of these tradeoffs, the model finds that TIP-susceptible HIV strains continually outcompete TIP-resistant HIV mutants at both patient and population scales when TIPs are engineered to express >3-fold more genomic RNA than HIV expresses. Thus, the results provide design constraints for engineering population-scale therapies that may be refractory to the acquisition of antiviral resistance.


Subject(s)
Anti-HIV Agents/pharmacology , Defective Viruses/drug effects , Defective Viruses/genetics , Drug Design , HIV/drug effects , HIV/genetics , Computational Biology , Drug Resistance, Viral/genetics , Evolution, Molecular , HIV/physiology , HIV Infections/drug therapy , HIV Infections/transmission , HIV Infections/virology , Humans , Models, Biological , Mutation , Selection, Genetic , Viral Load , Virus Replication/drug effects , Virus Replication/genetics
4.
J Immunol ; 196(9): 3608-17, 2016 05 01.
Article in English | MEDLINE | ID: mdl-27016602

ABSTRACT

Influenza A virus gene segment 7 encodes two proteins: the M1 protein translated from unspliced mRNA and the M2 protein produced by mRNA splicing and largely encoded by the M1 +1 reading frame. To better understand the generation of defective ribosomal products relevant to MHC class I Ag presentation, we engineered influenza A virus gene segment 7 to encode the model H-2 K(b) class I peptide ligand SIINFEKL at the M2 protein C terminus. Remarkably, after treating virus-infected cells with the RNA splicing inhibitor spliceostatin A to prevent M2 mRNA generation, K(b)-SIINFEKL complexes were still presented on the cell surface at levels ≤60% of untreated cells. Three key findings indicate that SIINFEKL is produced by cytoplasmic translation of unspliced M1 mRNA initiating at CUG codons within the +1 reading frame: 1) synonymous mutation of CUG codons in the M2-reading frame reduced K(b)-SIINFEKL generation; 2) K(b)-SIINFEKL generation was not affected by drug-mediated inhibition of AUG-initiated M1 synthesis; and 3) K(b)-SIINFEKL was generated in vitro and in vivo from mRNA synthesized in the cytoplasm by vaccinia virus, and hence cannot be spliced. These findings define a viral defective ribosomal product generated by cytoplasmic noncanonical translation and demonstrate the participation of CUG-codon-based translation initiation in pathogen immunosurveillance.


Subject(s)
Defective Viruses/genetics , Influenza A virus/genetics , Peptides/genetics , Ribosomes/metabolism , Viral Matrix Proteins/genetics , Animals , Antigen Presentation/drug effects , Cell Line , Defective Viruses/chemistry , Defective Viruses/drug effects , Defective Viruses/metabolism , Genes, MHC Class I , HeLa Cells , Humans , Influenza A virus/chemistry , Influenza A virus/drug effects , Influenza A virus/metabolism , Mice , Mice, Inbred C57BL , Peptides/chemistry , Protein Biosynthesis , Pyrans/pharmacology , RNA Splicing/drug effects , Spiro Compounds/pharmacology
5.
Bioorg Med Chem Lett ; 23(7): 2172-6, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23453839

ABSTRACT

Novel N-alkyldeoxynojirimycins (NADNJs) with two hydrophobic groups attached to a nitrogen linker on the alkyl chain were designed. A novel NADNJ containing a terminal tertiary carboxamide moiety was discovered that was a potent inhibitor against BVDV. Further optimization resulted in a structurally more stable lead compound 24 with a submicromolar EC50 against BVDV, Dengue, and Tacaribe; and low cytotoxicity.


Subject(s)
Amides/pharmacology , Antiviral Agents/pharmacology , Defective Viruses/drug effects , Dengue/drug therapy , Diarrhea Viruses, Bovine Viral/drug effects , Glucosamine/analogs & derivatives , 1-Deoxynojirimycin/chemical synthesis , 1-Deoxynojirimycin/chemistry , 1-Deoxynojirimycin/pharmacology , Amides/chemical synthesis , Amides/chemistry , Animals , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Cell Line , Cell Survival/drug effects , Cricetinae , Dose-Response Relationship, Drug , Glucosamine/chemical synthesis , Glucosamine/chemistry , Glucosamine/pharmacology , Humans , Molecular Structure , Structure-Activity Relationship
6.
Cancer Gene Ther ; 16(3): 237-45, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18949013

ABSTRACT

Combining the use of a chemotherapeutic agent with oncolytic virotherapy is a useful way to increase the efficiency of the treatment of cancer. The effect of the histone diacetylase (HDAC) inhibitor trichostatin A (TSA) on the antitumor activity of a herpes simplex virus type-1 (HSV-1) mutant was examined in oral squamous cell carcinoma (SCC) cells. Immunoblotting analysis and immunoflourescence staining revealed that a cytoplasmic nuclear factor-kappaB (NF-kappaB) component, p65, translocated into the nucleus after infection with gamma(1)34.5 gene-deficient HSV-1 R849, indicating that R849 activated NF-kappaB. TSA induced acetylation of p65 and increased the amount of p65 in the nucleus of oral SCC cells. Treatment of R849-infected cells with TSA also increased the amount of nuclear p65 and binding of NF-kappaB to its DNA-binding site and an NF-kappaB inhibitor SN50 diminished the increase in nuclear p65. In the presence of TSA, the production of virus and the expression of LacZ integrated into R849 and glycoprotein D, but not ICP0, ICP6 and thymidine kinase, were increased. The viability of cells treated with a combination of R849 and TSA was lower than that of those treated with R849 only. After treatment with TSA, expression of the cell cycle kinase inhibitor p21 was upregulated and the cell cycle was arrested at G1. These results indicate that TSA enhanced the replication of the HSV-1 mutant through the activation of NF-kappaB and induced cell cycle arrest at G1 to inhibit cell growth. TSA can be used as an enhancing agent for oncolytic virotherapy for oral SCC with gamma(1)34.5 gene-deficient HSV-1.


Subject(s)
Carcinoma, Squamous Cell/pathology , Enzyme Inhibitors/pharmacology , Herpesvirus 1, Human/drug effects , Hydroxamic Acids/pharmacology , Mouth Neoplasms/pathology , Oncolytic Virotherapy , Virus Activation/drug effects , Acetylation/drug effects , Cell Line, Tumor/enzymology , Cell Line, Tumor/virology , Defective Viruses/drug effects , Defective Viruses/genetics , Defective Viruses/physiology , Drug Screening Assays, Antitumor , Gene Expression Regulation, Viral/drug effects , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/physiology , Histone Deacetylase Inhibitors , Histones/metabolism , Humans , NF-kappa B/metabolism , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Peptides/pharmacology , Protein Processing, Post-Translational/drug effects , Protein Transport/drug effects , Transcription Factor RelA/physiology , Viral Proteins/genetics , Virus Replication/drug effects
8.
Biotechnol Prog ; 16(3): 493-6, 2000.
Article in English | MEDLINE | ID: mdl-10835254

ABSTRACT

The ability to obtain high titer replication-defective herpes simplex virus (HSV) recombinant vectors will dramatically affect their use in gene therapy clinical trials. A variety of techniques and reagents have been employed to increase the overall yield of the vector. The effects of protease inhibitors on the yield of an HSV-1-based viral vector were examined. Experiments were conducted using a commercial protease inhibitor cocktail typically used in mammalian cell culture for protein production. Contrary to our expectation for enhanced vector yield, the results showed a dramatic reduction in vector yield. Moreover, it was found that AEBSF is the only component in the protease cocktail responsible for the low vector yield. On the basis of our hypothesis regarding the mode of action of AEBSF, we suggest that it should not be included in protease inhibitor cocktails designed for use in cultures aimed at production of viral vectors derived from HSV-1 or possibly several other vectors.


Subject(s)
Genetic Vectors , Herpesvirus 1, Human/drug effects , Herpesvirus 1, Human/genetics , Protease Inhibitors/pharmacology , Culture Media , Defective Viruses/drug effects , Defective Viruses/genetics
9.
J Virol ; 73(7): 5803-13, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10364332

ABSTRACT

The nonnucleoside reverse transcriptase (RT) inhibitor (NNRTI) delavirdine (DLV) selects in vitro for the human immunodeficiency virus type 1 (HIV-1) RT mutation P236L, which confers high-level resistance to DLV but not other NNRTIs. Unexpectedly, P236L has developed infrequently in HIV-1 isolates obtained from patients receiving DLV; K103N is the predominant resistance mutation observed in that setting. We characterized the replication fitness of viruses derived from pNL4-3 containing P236L or K103N in both H9 and primary human peripheral blood mononuclear cell cultures infected in parallel with the two mutants. In the absence of DLV, p24 production by wild-type virus occurred more rapidly and to higher levels than with either mutant; P236L consistently demonstrated a two- to threefold decrease in p24 relative to K103N. At low levels of DLV, growth of wild-type virus was severely inhibited, and K103N replicated two- to threefold more efficiently than P236L. At high concentrations of DLV, P236L replication and K103N replication were both inhibited. Recombinant RTs containing K103N or P236L were analyzed for DNA polymerization on heteropolymeric RNA templates and RNase H degradation of RNA-DNA hybrids. Neither mutant demonstrated defects in polymerization. K103N demonstrated normal RNA 5'-end-directed RNase H cleavage and slowed DNA 3'-end-directed RNase H cleavage compared to wild-type RT. P236L demonstrated slowing of both DNA 3'-end- and RNA 5'-end-directed RNase H cleavage, consistent with its reduced replication efficiency relative to K103N. These data suggest that NNRTI resistance mutations can lead to reductions in the efficiency of RNase H cleavage, which may contribute to a reduction in the replication fitness of HIV-1.


Subject(s)
Anti-HIV Agents/pharmacology , DNA, Viral/metabolism , Defective Viruses/physiology , Delavirdine/pharmacology , HIV-1/physiology , RNA, Viral/metabolism , Reverse Transcriptase Inhibitors/pharmacology , Ribonuclease H/metabolism , Virus Replication/drug effects , 5' Untranslated Regions , Cell Line , Defective Viruses/drug effects , Drug Resistance, Microbial , HIV Reverse Transcriptase/genetics , HIV-1/drug effects , HeLa Cells , Humans , Kinetics , Mutagenesis
10.
J Virol ; 72(7): 5811-9, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9621041

ABSTRACT

Inhibitors of the phosphatidylinositol 3-kinase (PI3 kinase)-FKBP-rapamycin-associated protein (FRAP) pathway, such as rapamycin and wortmannin, induce dephosphorylation and activation of the suppressor of cap-dependent translation, 4E-BP1. Encephalomyocarditis virus (EMCV) infection leads to activation of 4E-BP1 at the time of host translation shutoff. Consistent with these data, rapamycin mildly enhances the synthesis of viral proteins and the shutoff of host cell protein synthesis after EMCV infection. In this study, two defective EMCV strains were generated by deleting portions of the 2A coding region of an infectious cDNA clone. These deletions dramatically decreased the efficiency of viral protein synthesis and abolished the virus-induced shutoff of host translation after infection of BHK-21 cells. Both translation and processing of the P1-2A capsid precursor polypeptide are impaired by the deletions in 2A. The translation and yield of mutant viruses were increased significantly by the presence of rapamycin and wortmannin during infection. Thus, inhibition of the PI3 kinase-FRAP signaling pathway partly complements mutations in 2A protein and reverses a slow-virus phenotype.


Subject(s)
Androstadienes/pharmacology , Defective Viruses/drug effects , Encephalomyocarditis virus/drug effects , Enzyme Inhibitors/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Polyenes/pharmacology , Virus Replication/drug effects , Animals , Cells, Cultured , Cricetinae , Encephalomyocarditis virus/physiology , Protein Biosynthesis , Rabbits , Sirolimus , Viral Proteins/biosynthesis , Wortmannin
11.
Biochem Biophys Res Commun ; 237(3): 504-11, 1997 Aug 28.
Article in English | MEDLINE | ID: mdl-9299393

ABSTRACT

The role of the N-myristoylation of the human immunodeficiency virus type 1 (HIV-1) gag protein in ACH-2 cells was studied. The infectivity of HIV-1 from the cells stimulated with phorbol 12-myristate 13-acetate (PMA) was suppressed by pretreatment with N-myristoyl glycinal diethylacetal (N-Myr-GOA), a potent N-myristoylation inhibitor, and the blockage of myristoylation resulted in accumulation of immature gag precursors. The viral particles which budded from the non-N-Myr-GOA-treated ACH-2 cells stimulated with PMA exhibited a typical viral phenotype, whereas those which budded from the N-Myr-GOA-treated ACH-2 cells stimulated with PMA were twisted, as observed electron microscopically. In electron microscopic analyses with gold-labeled monoclonal antibodies to gag and env, gag and env were detected adjacent to each other in the PMA-stimulated ACH-2, but no env was detected in the cells treated with N-Myr-GOA. Taken together, the results suggest that the myristoylation of HIV-1 gag seems to be responsible for both maturation of gag and acquisition of HIV-1 infectivity.


Subject(s)
Acetaldehyde/pharmacology , Acetals/pharmacology , Defective Viruses/physiology , Gene Products, gag/metabolism , HIV-1/physiology , Myristic Acids/metabolism , Protein Processing, Post-Translational , Cell Line , Colonic Neoplasms , Defective Viruses/drug effects , Defective Viruses/ultrastructure , HIV Core Protein p24/biosynthesis , HIV Reverse Transcriptase/metabolism , HIV-1/drug effects , HIV-1/ultrastructure , Humans , Myristic Acid , Protein Precursors/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Tumor Cells, Cultured
12.
J Virol ; 70(2): 1271-6, 1996 Feb.
Article in English | MEDLINE | ID: mdl-8551594

ABSTRACT

Herpes simplex virus (HSV) mutants defective for thymidine kinase expression (TK-) have been reported to establish latent infection of sensory ganglia of mice, in that HSV latency-associated transcript is expressed, but to be defective for reactivation. In the present study, the mechanism of defective reactivation by TK- HSV was investigated. Latent infection established by each of three reactivation-defective HSV type 1 mutants was studied. Reactivation in explant culture was markedly enhanced by the addition of thymidine (dTdR) to the explant culture medium. Without added dTdR, reactivation occurred in 0 of 32 ganglia, while when dTdR (200 microM) was present, reactivation occurred in 32 of 37 ganglia (86%). Reactivation was minimal or did not occur after treatment with other nucleosides; specificity for dTdR would suggest the importance of dTdR nucleotide levels rather than more general nucleotide pool imbalance. Enhanced reactivation by dTdR was dose dependent and was blocked by acyclovir. While some degree of inhibition of TK- HSV by acyclovir may be expected, the complete block of dTdR-enhanced reactivation was unexpected. This result may suggest that HSV is particularly vulnerable during initial reactivation events. The mechanism of dTdR-enhanced reactivation of TK- HSV was further evaluated during in vivo infection by TK- HSV. For mice infected with TK- HSV, virus was undetectable in ganglia 3 days later. However, for mice infected with TK- HSV and treated with dTdR, virus was readily detected (2.8 x 10(3) PFU per ganglion). This result suggested that in vivo treatment with dTdR enhanced replication of TK- HSV in ganglion neurons. In turn, this suggests that in latently infected ganglia, dTdR-enhanced reactivation of TK- HSV occurred as a result of viral replication in neurons following initial reactivation events.


Subject(s)
Herpes Simplex/virology , Simplexvirus/enzymology , Thymidine Kinase/metabolism , Thymidine/pharmacology , Virus Activation/drug effects , Acyclovir/pharmacology , Animals , Antiviral Agents/pharmacology , Chlorocebus aethiops , Defective Viruses/drug effects , Defective Viruses/enzymology , Gene Deletion , Mice , Simplexvirus/drug effects , Thymidine Kinase/genetics , Vero Cells
13.
Nat Med ; 1(9): 938-43, 1995 Sep.
Article in English | MEDLINE | ID: mdl-7585221

ABSTRACT

We have created a double mutant of the herpes simplex virus (HSV) type 1 (termed G207) with favourable properties for treating human malignant brain tumours: replication-competence in glioblastoma cells (and other dividing cells), attenuated neurovirulence, temperature sensitivity, ganciclovir hypersensitivity, and the presence of an easily detectable histochemical marker. G207 has deletions at both gamma 34.5 (RL1) loci and a lacZ gene insertion inactivating the ICP6 gene (UL39). G207 kills human glioma cells in monolayer cultures. In nude mice harbouring subcutaneous or intracerebral U-87MG gliomas, intraneoplastic inoculation with G207 causes decreased tumour growth and/or prolonged survival. G207 is avirulent upon intracerebral inoculation of mice and HSV-sensitive non-human primates. These results suggest that G207 should be considered for clinical evaluation in the treatment of glioblastomas.


Subject(s)
Brain Neoplasms/therapy , Defective Viruses/physiology , Genetic Therapy , Glioblastoma/therapy , Simplexvirus/physiology , Animals , Aotidae , Chlorocebus aethiops , Cytopathogenic Effect, Viral , Defective Viruses/drug effects , Defective Viruses/genetics , Defective Viruses/pathogenicity , Ganciclovir/pharmacology , Genes, Reporter , Humans , Injections, Intralesional , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Rats , Safety , Simplexvirus/drug effects , Simplexvirus/genetics , Simplexvirus/pathogenicity , Temperature , Vero Cells , Viral Proteins/genetics , Virulence/genetics , Virus Latency , Virus Replication , beta-Galactosidase/genetics
14.
J Photochem Photobiol B ; 26(3): 241-7, 1994 Dec.
Article in English | MEDLINE | ID: mdl-7853115

ABSTRACT

The replication defective retrovirus, pXM5(N2), was used for an easy, safe and reproducible test for the screening of furocoumarins with antiretroviral activity. High titer viral supernatants have been photomodified by UVA light (20 kJ m-2) in the presence of different concentrations of two psolarens (8-methoxypsoralen, 8-MOP and 4,5',8-trimethylpsoralen, TMP) and one angelicin (4,6,4'-trimethylangelicin, TMA). At low concentrations (100-250 ng ml-1) 8-MOP and TMA did not show any significant antiviral activity, while TMP demonstrated a reduction of virus infectivity by one log at 250 ng ml-1. At the highest concentration (5 micrograms ml-1), TMA and TMP reduced the virus titer by one and more than two logs, respectively, being, therefore, two and four times more active than 8-MOP. The most active compound, TMP, was further tested on HIV-1 viral supernatants. Total inactivation of the HIV-1 (200 SFU) was obtained in the presence of 1 microgram ml-1 of TMP and 20 kJ m-2 of UVA light. Our results support the validity of the N2 system to detect the antiretroviral activity of furocoumarins and suggest the potential of TMP in combination with UVA light against HIV-1.


Subject(s)
Antiviral Agents/pharmacology , Coumarins/pharmacology , Defective Viruses/drug effects , HIV-1/drug effects , Photosensitizing Agents/pharmacology , Retroviridae/drug effects , Ultraviolet Rays , 3T3 Cells , Animals , Cell Line , Defective Viruses/physiology , Defective Viruses/radiation effects , Dose-Response Relationship, Drug , Furocoumarins/pharmacology , HIV-1/physiology , HIV-1/radiation effects , Humans , Methoxsalen/pharmacology , Mice , Retroviridae/physiology , Retroviridae/radiation effects , Trioxsalen/pharmacology , Virus Replication
15.
J Virol ; 68(3): 1324-33, 1994 Mar.
Article in English | MEDLINE | ID: mdl-8107197

ABSTRACT

Tat-independent transcription of human immunodeficiency virus type 1 (HIV-1) plays an important role in virus life cycle before biologically significant levels of Tat protein have been accumulated. Using a latently infected T-cell line containing an integrated Tat-defective HIV-1 provirus, we examined whether factors known to up-regulate the HIV-1 expression in vitro can replace the requirement for a functional Tat protein and induce the expression of the Tat-defective HIV-1 provirus. Both tumor necrosis factor alpha (TNF-alpha) and herpes simplex virus type 1 (HSV-1) infection stimulated transcription of the Tat-defective HIV-1 provirus to comparable levels, but in HSV-1-infected cells, the cytoplasmic HIV-1 transcripts were not efficiently translated in the absence of Tat protein and were excluded from the large polysomes. However, HSV-1 infection did not affect the distribution of cellular gamma-actin RNA or 28S RNA in the polysomal fractions. The translational block of HIV-1 RNA was not mediated by the virion-associated host cell shutoff protein (vhs); dissociation of HIV-1 transcripts from the polysomes and inefficient translation was also observed in cells infected with the vhs-defective mutant of HSV-1 (vhs-1). Overexpression of Rev protein did not rescue the synthesis of HIV-1 proteins in these cells; however, the observed inhibition of HIV-1 RNA translation was efficiently overcome in the presence of Tat protein or TNF-alpha. These findings suggest that, in contrast to TNF-alpha, HSV-1 infection is not able to induce a full cycle of HIV-1 replication and that cytokines and Tat have a critical role in the activation of HIV-1 provirus by HSV-1.


Subject(s)
Defective Viruses/growth & development , Gene Products, tat/pharmacology , HIV-1/growth & development , Herpesvirus 1, Human , T-Lymphocytes/microbiology , Tumor Necrosis Factor-alpha/pharmacology , Virus Activation/drug effects , Cell Compartmentation , Cell Line , Defective Viruses/drug effects , Genes, rev , Genome, Viral , HIV-1/drug effects , Polyribosomes/chemistry , Protein Biosynthesis , RNA, Messenger/analysis , Transfection , Viral Proteins/biosynthesis , tat Gene Products, Human Immunodeficiency Virus
16.
Proc Natl Acad Sci U S A ; 90(24): 11835-9, 1993 Dec 15.
Article in English | MEDLINE | ID: mdl-8265634

ABSTRACT

Infection with herpes simplex viruses (HSVs) resistant to treatment with acyclovir (9-[(2-hydroxyethoxy)-methyl]guanine, Zovirax) is a growing clinical problem in patients with AIDS and other immunosuppressed states. Most virus isolates resistant to acyclovir are deficient or defective in virally coded thymidine kinase (TK), which converts acyclovir to acyclovir monophosphate in virus-infected cells. To restore acyclovir efficacy, we synthesized acyclovir diphosphate dimyristoylglycerol, an analog of a naturally occurring phospholipid, CDP-diacylglycerol. Its biological activity was tested in WI38 human lung fibroblasts infected with the acyclovir-resistant DM21 strain of HSV, which is TK negative due to an 816-base-pair deletion in the TK coding region. Acyclovir diphosphate dimyristoylglycerol has substantial activity in DM21-infected cells (IC50 = 0.25 microM), whereas acyclovir and acyclovir monophosphate were ineffective (IC50 > 100 microM). Similar results were obtained in TK-altered and TK-deficient strains of HSV-1 and in acyclovir-resistant isolates of HSV-2 obtained from two AIDS patients. The phospholipid prodrug is active by means of TK-independent metabolic pathways that liberate acyclovir monophosphate inside the host cell. Acyclovir phosphates were 56 times greater in WI38 human lung fibroblasts incubated for 24 hr with [8-3H]acyclovir diphosphate dimyristoylglycerol relative to acyclovir. Acyclovir monophosphate added to the culture medium (outside the cell) did not circumvent the acyclovir resistance of the TK-negative DM21 mutant, presumably due to its conversion to acyclovir by phosphatases. Acyclovir diphosphate diacylglycerol prodrugs may be useful in treating TK-deficient mutant and wild-type strains of HSV.


Subject(s)
Acyclovir/analogs & derivatives , Acyclovir/toxicity , Antiviral Agents/toxicity , Drug Resistance, Microbial , Herpesvirus 1, Human/drug effects , Herpesvirus 2, Human/drug effects , Phosphatidylglycerols/toxicity , Prodrugs/toxicity , Acquired Immunodeficiency Syndrome/microbiology , Acyclovir/chemistry , Acyclovir/metabolism , Adult , Antiviral Agents/chemical synthesis , Cell Line , Defective Viruses/drug effects , Defective Viruses/isolation & purification , Defective Viruses/physiology , Herpesvirus 1, Human/isolation & purification , Herpesvirus 1, Human/physiology , Herpesvirus 2, Human/isolation & purification , Herpesvirus 2, Human/physiology , Humans , Lung , Male , Microbial Sensitivity Tests , Molecular Structure , Phosphatidylglycerols/chemistry , Phosphatidylglycerols/metabolism , Thymidine Kinase/genetics , Thymidine Kinase/metabolism , Virus Replication/drug effects
17.
Vaccine ; 11(9): 957-60, 1993.
Article in English | MEDLINE | ID: mdl-8212843

ABSTRACT

Direct DNA inoculations have been used to demonstrate that in vivo transfections can be used to elicit protective immune responses. The direct inoculation of an H7 haemagglutinin-expressing DNA protected chickens against lethal challenge with an H7N7 influenza virus. Three-week-old chickens were vaccinated by inoculating 100 micrograms of plasma DNA by each of three routes (intravenous, intraperitoneal and subcutaneous). One month later, chickens were boosted with 100 micrograms of DNA by each of the three routes. At 1-2 weeks postboost, chickens were challenged via the nares with 100 lethal doses of an H7N7 virus. Low to undetectable levels of H7-specific antibodies were present postvaccination and boost. High titres of H7-specific antibodies appeared within 1 week of challenge. In a series of four experiments, 50% (28/56) of the DNA-vaccinated and < 2% (1/67) of the control chickens survived the challenge. This exceptionally simple method of immunization holds high promise for the development of subunit vaccines.


Subject(s)
Avian Leukosis Virus/genetics , DNA, Viral/genetics , Defective Viruses/immunology , Genetic Vectors , Hemagglutinins, Viral/immunology , Influenza A virus/immunology , Influenza Vaccines/immunology , Orthomyxoviridae Infections/prevention & control , Plasmids , Recombinant Fusion Proteins/immunology , Amantadine/pharmacology , Animals , Chickens/immunology , Defective Viruses/drug effects , Defective Viruses/genetics , Hemagglutinin Glycoproteins, Influenza Virus , Hemagglutinins, Viral/biosynthesis , Hemagglutinins, Viral/genetics , Immunity, Active , Immunization , Influenza A virus/drug effects , Influenza A virus/genetics , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Specific Pathogen-Free Organisms , Transfection
18.
AIDS Res Hum Retroviruses ; 6(4): 491-501, 1990 Apr.
Article in English | MEDLINE | ID: mdl-1692729

ABSTRACT

A new class of membrane-active ether lipid (EL) analogs of platelet-activating factor were studied for in vitro anti-HIV-1 activity. Human T-cell (CEM-ss) monolayers or suspension cultures were used to determine effects of structural modifications of Type A phosphorus-containing and Type B nonphosphorus EL analogs on (a) the inhibitory concentration50 (IC50) for HIV-1 syncytial plaque formation and cell growth, and, (b) virus budding at the cell plasma membrane. Results indicate that representative Type A and Type B EL inhibit HIV-1 but not herpes simplex virus type 2 plaque formation when added before or up to 2 days after viral infection. Anti-HIV-1 activity does not involve direct inactivation of virus infectivity. Type A EL (IC50 range = 0.2-1.4 microM) with alkyoxy, alkylthio, or alkyamido substitution at glycerol position 1 and ethoxy or methoxy substitution at position 2, and Type B compounds (IC50 range = 0.33-0.63 microM) with an inverse choline or nitrogen heterocyclic substitution at position 3 have selective activity against HIV-1-infected T-cells. EL treatment of HIV-1-infected cells is associated with subsequent release of reverse transcriptase activity, but infectious virus production is inhibited with time after infection. Electron microscopic examination of HIV-1-infected and EL-treated cells revealed absence of detectable budding virus at the plasma membrane but presence of intracytoplasmic vacuolar virus particles. In summary, these data suggest that EL analogs are a novel class of agents that induce defective intracytoplasmic vacuolar HIV-1 formation in T-cells. Being membrane interactive, EL are ideally suited for combination chemotherapy with DNA-interactive anti-HIV nucleoside analogs.


Subject(s)
Defective Viruses/drug effects , HIV-1/drug effects , Lipids/pharmacology , Phospholipid Ethers/pharmacology , Virus Replication/drug effects , Cell Membrane/microbiology , Dose-Response Relationship, Drug , HIV-1/growth & development , RNA-Directed DNA Polymerase/analysis
20.
Virus Res ; 13(4): 295-302, 1989 Aug.
Article in English | MEDLINE | ID: mdl-2554610

ABSTRACT

We here report the results of an investigation of the effect of interferon on the establishment of new infections by a retrovirus. For this study, we used an infectious but replication-incompetent retrovirus carrying a drug-resistance gene and assayed for infectivity by measuring drug-resistant colony formation. Mouse interferon-beta inhibited retroviral infection of mouse CG1 cells in a dose-dependent manner. However, a higher dose of interferon was needed for eliciting the antiretroviral effects than for action against vesicular stomatitis virus. The degree of antiretroviral effect was comparable over at least a 100-fold range of multiplicity of infection and the effect was most pronounced when the cells were continuously treated with interferon before infections and during infection and drug-selection.


Subject(s)
Interferon Type I/pharmacology , Retroviridae Infections/prevention & control , Animals , Cell Line , Defective Viruses/drug effects , Defective Viruses/genetics , Defective Viruses/physiology , Drug Resistance, Microbial/genetics , Lysogeny/drug effects , Retroviridae/drug effects , Retroviridae/genetics , Retroviridae/physiology , Retroviridae Infections/microbiology , Vesicular stomatitis Indiana virus/drug effects , Vesicular stomatitis Indiana virus/physiology , Virus Replication/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...