44 results on '"Prichard MN"'
Search Results
2. Filociclovir Is a Potent In Vitro and In Vivo Inhibitor of Human Adenoviruses.
- Author
-
Toth K, Hussein ITM, Tollefson AE, Ying B, Spencer JF, Eagar J, James SH, Prichard MN, Wold WSM, and Bowlin TL
- Subjects
- Animals, Antiviral Agents pharmacology, Antiviral Agents therapeutic use, Cricetinae, Humans, Virus Replication, Adenovirus Infections, Human drug therapy, Adenoviruses, Human, Cytomegalovirus Infections drug therapy
- Abstract
Human adenovirus (HAdV) infection is common in the general population and can cause a range of clinical manifestations, among which pneumonia and keratoconjunctivitis are the most common. Although HAdV infections are mostly self-limiting, infections in immunocompromised individuals can be severe. No antiviral drug has been approved for treating adenoviruses. Filociclovir (FCV) is a nucleoside analogue which has successfully completed phase I human clinical safety studies and is now being developed for treatment of human cytomegalovirus (HCMV)-related disease in immunocompromised patients. In this report, we show that FCV is a potent broad-spectrum inhibitor of HAdV types 4 to 8, with 50% effective concentrations (EC
50 s) ranging between 1.24 and 3.6 μM and a 50% cytotoxic concentration (CC50 ) of 100 to 150 μM in human foreskin fibroblasts (HFFs). We also show that the prophylactic oral administration of FCV (10 mg/kg of body weight) 1 day prior to virus challenge and then daily for 14 days to immunosuppressed Syrian hamsters infected intravenously with HAdV6 was sufficient to prevent morbidity and mortality. FCV also mitigated tissue damage and inhibited virus replication in the liver. The 10-mg/kg dose had similar effects even when the treatment was started on day 4 after virus challenge. Furthermore, FCV administered at the same dose after intranasal challenge with HAdV6 partially mitigated body weight loss but significantly reduced pathology and virus replication in the lung. These findings suggest that FCV could potentially be developed as a pan-adenoviral inhibitor., (Copyright © 2020 American Society for Microbiology.)- Published
- 2020
- Full Text
- View/download PDF
3. Hospital-acquired viral respiratory infections in neonates hospitalized since birth in a tertiary neonatal intensive care unit.
- Author
-
Poole CL, Camins BC, Prichard MN, Faye-Petersen O, and Hutto C
- Subjects
- Alabama, Bronchopulmonary Dysplasia diagnosis, Cross Infection diagnosis, Female, Gestational Age, Hospitalization, Humans, Infant, Low Birth Weight, Infant, Newborn, Infant, Premature, Male, Polymerase Chain Reaction, Prospective Studies, Respiratory Tract Infections diagnosis, Cross Infection virology, Intensive Care Units, Neonatal, Respiratory Tract Infections virology, Virus Diseases diagnosis
- Abstract
Objective: To determine frequency of hospital-acquired viral respiratory infections (HA-VRI) and associated outcomes in a NICU., Study Design: Prospective cohort study conducted from 4 October 2016 to 21 March 2017. Infants hospitalized from birth in the NICU had a weekly nasal swab collected for testing using a multiplex PCR assay capable of detecting 16 different respiratory viruses., Results: Seventy-four infants enrolled, with 5 (6.8%) testing positive for a virus (incidence rate of 1.3/1000 patient days). VRI positive infants had a younger gestational age (median 27 w vs. 32 w, p = 0.048); were hospitalized longer (97 d vs 43 d, p = 0.013); required more antibiotics (8 d vs. 4 d, p = 0.037) and were more likely to be diagnosed with bronchopulmonary dysplasia (p = 0.008) compared to VRI negative infants., Conclusion: Respiratory viruses are a frequent cause of HAI in the NICU and are associated with negative outcomes.
- Published
- 2019
- Full Text
- View/download PDF
4. Genotypic and Phenotypic Diversity of Herpes Simplex Virus 2 within the Infected Neonatal Population.
- Author
-
Akhtar LN, Bowen CD, Renner DW, Pandey U, Della Fera AN, Kimberlin DW, Prichard MN, Whitley RJ, Weitzman MD, and Szpara ML
- Subjects
- Cell Line, Encephalitis, Viral virology, Female, Genome, Viral, Genomics, Genotype, Gestational Age, Herpes Simplex complications, Herpesvirus 1, Human genetics, Herpesvirus 2, Human isolation & purification, Herpesvirus 2, Human pathogenicity, Humans, Infant, Newborn, Male, Phenotype, Pregnancy, Viral Proteins genetics, Genetic Variation, Herpes Simplex virology, Herpesvirus 2, Human genetics, Pregnancy Complications, Infectious virology
- Abstract
More than 14,000 neonates are infected with herpes simplex virus (HSV) annually. Approximately half display manifestations limited to the skin, eyes, or mouth (SEM disease). The rest develop invasive infections that spread to the central nervous system (CNS disease or encephalitis) or throughout the infected neonate (disseminated disease). Invasive HSV disease is associated with significant morbidity and mortality, but the viral and host factors that predispose neonates to these forms are unknown. To define viral diversity within the infected neonatal population, we evaluated 10 HSV-2 isolates from newborns with a range of clinical presentations. To assess viral fitness independently of host immune factors, we measured viral growth characteristics in cultured cells and found diverse in vitro phenotypes. Isolates from neonates with CNS disease were associated with larger plaque size and enhanced spread, with the isolates from cerebrospinal fluid (CSF) exhibiting the most robust growth. We sequenced complete viral genomes of all 10 neonatal viruses, providing new insights into HSV-2 genomic diversity in this clinical setting. We found extensive interhost and intrahost genomic diversity throughout the viral genome, including amino acid differences in more than 90% of the viral proteome. The genes encoding glycoprotein G (gG; US4), glycoprotein I (gI; US7), and glycoprotein K (gK; UL53) and viral proteins UL8, UL20, UL24, and US2 contained variants that were found in association with CNS isolates. Many of these viral proteins are known to contribute to cell spread and neurovirulence in mouse models of CNS disease. This report represents the first application of comparative pathogen genomics to neonatal HSV disease. IMPORTANCE Herpes simplex virus (HSV) causes invasive disease in half of infected neonates, resulting in significant mortality and permanent cognitive morbidity. The factors that contribute to invasive disease are not understood. This study revealed diversity among HSV isolates from infected neonates and detected the first associations between viral genetic variations and clinical disease manifestations. We found that viruses isolated from newborns with encephalitis showed enhanced spread in culture. These viruses contained protein-coding variations not found in viruses causing noninvasive disease. Many of these variations were found in proteins known to impact neurovirulence and viral spread between cells. This work advances our understanding of HSV diversity in the neonatal population and how it may impact disease outcome., (Copyright © 2019 Akhtar et al.)
- Published
- 2019
- Full Text
- View/download PDF
5. Differential properties of cytomegalovirus pUL97 kinase isoforms affect viral replication and maribavir susceptibility.
- Author
-
Webel R, Hakki M, Prichard MN, Rawlinson WD, Marschall M, and Chou S
- Subjects
- Cytomegalovirus drug effects, Humans, Microbial Sensitivity Tests, Protein Isoforms genetics, Protein Isoforms metabolism, Antiviral Agents pharmacology, Benzimidazoles pharmacology, Cytomegalovirus enzymology, Cytomegalovirus physiology, Phosphotransferases (Alcohol Group Acceptor) genetics, Phosphotransferases (Alcohol Group Acceptor) metabolism, Ribonucleosides pharmacology, Virus Replication drug effects
- Abstract
Unlabelled: The human cytomegalovirus (HCMV)-encoded kinase pUL97 is required for efficient viral replication. Previous studies described two isoforms of pUL97, the full-length isoform (M1) and a smaller isoform likely resulting from translation initiation at codon 74 (M74). Here, we report the detection of a third pUL97 isoform during viral infection resulting from translation initiation at codon 157 (isoform M157). The consistent expression of isoform M157 as a minor component of pUL97 during infection with clinical and laboratory-adapted HCMV strains was suppressed when codon 157 was mutagenized. Viral mutants expressing specific isoforms were generated to compare their growth and drug susceptibility phenotypes, as well as pUL97 intracellular localization patterns and kinase activities. The exclusive expression of isoform M157 resulted in substantially reduced viral growth and resistance to the pUL97 inhibitor maribavir while retaining susceptibility to ganciclovir. Confocal imaging demonstrated reduced nuclear import of amino-terminal deletion isoforms compared to isoform M1. Isoform M157 showed reduced efficiency of various substrate protein interactions and autophosphorylation, whereas Rb phosphorylation was preserved. These results reveal differential properties of pUL97 isoforms that affect viral replication, with implications for the antiviral efficacy of maribavir., Importance: The HCMV UL97 kinase performs important functions in viral replication that are targeted by the antiviral drug maribavir. Here, we describe a naturally occurring short isoform of the kinase that when expressed by itself in a recombinant virus results in altered intracellular localization, impaired growth, and high-level resistance to maribavir compared to those of the predominant full-length counterpart. This is another factor to consider in explaining why maribavir appears to have variable antiviral activity in cell culture and in vivo.
- Published
- 2014
- Full Text
- View/download PDF
6. New small molecule entry inhibitors targeting hemagglutinin-mediated influenza a virus fusion.
- Author
-
Basu A, Antanasijevic A, Wang M, Li B, Mills DM, Ames JA, Nash PJ, Williams JD, Peet NP, Moir DT, Prichard MN, Keith KA, Barnard DL, Caffrey M, Rong L, and Bowlin TL
- Subjects
- Animals, Antiviral Agents chemistry, Cell Line, Chickens, Hemagglutinins, Viral genetics, Humans, Influenza A Virus, H1N1 Subtype drug effects, Influenza A Virus, H1N1 Subtype genetics, Influenza A Virus, H1N1 Subtype physiology, Influenza A Virus, H3N2 Subtype drug effects, Influenza A Virus, H3N2 Subtype genetics, Influenza A Virus, H3N2 Subtype physiology, Influenza A Virus, H5N1 Subtype drug effects, Influenza A Virus, H5N1 Subtype genetics, Influenza A Virus, H5N1 Subtype physiology, Influenza A virus genetics, Influenza A virus physiology, Small Molecule Libraries chemistry, Antiviral Agents pharmacology, Hemagglutinins, Viral metabolism, Influenza A virus drug effects, Influenza in Birds virology, Influenza, Human virology, Poultry Diseases virology, Small Molecule Libraries pharmacology, Virus Internalization drug effects
- Abstract
Influenza viruses are a major public health threat worldwide, and options for antiviral therapy are limited by the emergence of drug-resistant virus strains. The influenza virus glycoprotein hemagglutinin (HA) plays critical roles in the early stage of virus infection, including receptor binding and membrane fusion, making it a potential target for the development of anti-influenza drugs. Using pseudotype virus-based high-throughput screens, we have identified several new small molecules capable of inhibiting influenza virus entry. We prioritized two novel inhibitors, MBX2329 and MBX2546, with aminoalkyl phenol ether and sulfonamide scaffolds, respectively, that specifically inhibit HA-mediated viral entry. The two compounds (i) are potent (50% inhibitory concentration [IC50] of 0.3 to 5.9 μM); (ii) are selective (50% cytotoxicity concentration [CC(50)] of >100 μM), with selectivity index (SI) values of >20 to 200 for different influenza virus strains; (iii) inhibit a wide spectrum of influenza A viruses, which includes the 2009 pandemic influenza virus A/H1N1/2009, highly pathogenic avian influenza (HPAI) virus A/H5N1, and oseltamivir-resistant A/H1N1 strains; (iv) exhibit large volumes of synergy with oseltamivir (36 and 331 μM(2) % at 95% confidence); and (v) have chemically tractable structures. Mechanism-of-action studies suggest that both MBX2329 and MBX2546 bind to HA in a nonoverlapping manner. Additional results from HA-mediated hemolysis of chicken red blood cells (cRBCs), competition assays with monoclonal antibody (MAb) C179, and mutational analysis suggest that the compounds bind in the stem region of the HA trimer and inhibit HA-mediated fusion. Therefore, MBX2329 and MBX2546 represent new starting points for chemical optimization and have the potential to provide valuable future therapeutic options and research tools to study the HA-mediated entry process.
- Published
- 2014
- Full Text
- View/download PDF
7. Human cytomegalovirus UL97 kinase is involved in the mechanism of action of methylenecyclopropane analogs with 6-ether and -thioether substitutions.
- Author
-
Komazin-Meredith G, Chou S, Prichard MN, Hartline CB, Cardinale SC, Comeau K, Williams JD, Khan AR, Peet NP, and Bowlin TL
- Subjects
- Cell Line, Humans, Antiviral Agents chemistry, Antiviral Agents pharmacology, Cyclopropanes chemistry, Cyclopropanes pharmacology, Cytomegalovirus drug effects, Cytomegalovirus enzymology, Ether chemistry, Sulfides chemistry
- Abstract
Methylenecyclopropane nucleoside (MCPN) analogs are being investigated for treatment of human cytomegalovirus (HCMV) infection because of favorable preclinical data and limited ganciclovir cross-resistance. Monohydroxymethyl MCPNs bearing ether and thioether functionalities at the purine 6 position have antiviral activity against herpes simplex virus (HSV) and varicella-zoster virus (VZV) in addition to HCMV. The role of the HCMV UL97 kinase in the mechanism of action of these derivatives was examined. When tested against a kinase-inactive UL97 K355M virus, a moderate 5- to 7-fold increase in 50% effective concentration (EC50) was observed, in comparison to a 13- to 25-fold increase for either cyclopropavir or ganciclovir. Serial propagation of HCMV under two of these compounds selected for three novel UL97 mutations encoding amino acid substitutions D456N, C480R,and Y617del. When transferred to baseline laboratory HCMV strains, these mutations individually conferred resistance to all of the tested MCPNs, ganciclovir, and maribavir. However, the engineered strains also demonstrated severe growth defects and abnormal cytopathic effects similar to the kinase-inactive mutant. Expressed and purified UL97 kinase showed in vitro phosphorylation of the newly tested MCPNs. Thus, HCMV UL97 kinase is involved in the antiviral action of these MCPNs, but the in vitro selection of UL97-defective viruses suggests that their activity against more typical ganciclovir-resistant growth-competent UL97 mutants may be relatively preserved.
- Published
- 2014
- Full Text
- View/download PDF
8. Human herpesvirus 6 U69 kinase phosphorylates the methylenecyclopropane nucleosides cyclopropavir, MBX 2168, and MBX 1616 to their monophosphates.
- Author
-
Komazin-Meredith G, Cardinale SC, Williams JD, Peet NP, Prichard MN, and Bowlin TL
- Subjects
- Baculoviridae genetics, Enzyme Assays, Guanine chemistry, Herpesvirus 6, Human chemistry, Humans, Kinetics, Phosphorylation, Protein Kinases genetics, Protein Kinases isolation & purification, Recombinant Fusion Proteins chemistry, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins isolation & purification, Substrate Specificity, Viral Proteins genetics, Viral Proteins isolation & purification, Antiviral Agents chemistry, Cyclopropanes chemistry, Guanine analogs & derivatives, Herpesvirus 6, Human enzymology, Nucleosides chemistry, Protein Kinases chemistry, Viral Proteins chemistry
- Abstract
Dihydroxymethyl and monohydroxymethyl methylenecyclopropane nucleosides are effective inhibitors of both variants of human herpesvirus 6 (HHV-6). We investigated involvement of HHV-6 U69 protein kinase in their mechanism of action. Phosphorylation of the dihydroxymethyl analogue cyclopropavir and monohydroxymethyl nucleosides with either a 6-ether moiety (MBX 2168) or a 6-thioether moiety (MBX 1616) with purified U69 was examined. All three compounds were substrates of this viral kinase and had similar Michaelis-Menten kinetic parameters.
- Published
- 2013
- Full Text
- View/download PDF
9. Synthesis and antiviral activities of methylenecyclopropane analogs with 6-alkoxy and 6-alkylthio substitutions that exhibit broad-spectrum antiviral activity against human herpesviruses.
- Author
-
Prichard MN, Williams JD, Komazin-Meredith G, Khan AR, Price NB, Jefferson GM, Harden EA, Hartline CB, Peet NP, and Bowlin TL
- Subjects
- Antiviral Agents pharmacology, Cell Proliferation drug effects, Cells, Cultured, Cyclopropanes chemistry, Cytomegalovirus enzymology, DNA, Viral analysis, Drug Evaluation, Preclinical, Guanine analogs & derivatives, Guanine pharmacology, Herpesviridae physiology, Herpesvirus 4, Human drug effects, Herpesvirus 4, Human physiology, Herpesvirus 6, Human drug effects, Herpesvirus 6, Human physiology, Herpesvirus 8, Human drug effects, Herpesvirus 8, Human physiology, Humans, Phosphotransferases (Alcohol Group Acceptor) chemistry, Phosphotransferases (Alcohol Group Acceptor) genetics, Purine Nucleosides chemical synthesis, Purine Nucleosides pharmacology, Viral Plaque Assay, Virus Replication drug effects, Antiviral Agents chemical synthesis, Cyclopropanes pharmacology, Cytomegalovirus drug effects, Herpesviridae drug effects
- Abstract
Methylenecyclopropane nucleosides have been reported to be active against many of the human herpesviruses. The most active compound of this class is cyclopropavir (CPV), which exhibits good antiviral activity against human cytomegalovirus (HCMV), Epstein-Barr virus, both variants of human herpesvirus 6, and human herpesvirus 8. CPV has two hydroxymethyl groups on the methylenecyclopropane ring, but analogs with a single hydroxymethyl group, such as the prototypical (S)-synguanol, are also active and exhibit a broader spectrum of antiviral activity that also includes hepatitis B virus and human immunodeficiency virus. Here, a large set of monohydroxymethyl compounds with ether and thioether substituents at the 6 position of the purine was synthesized and evaluated for antiviral activity against a range of human herpesviruses. Some of these analogs had a broader spectrum of antiviral activity than CPV, in that they also inhibited the replication of herpes simplex viruses 1 and 2 and varicella-zoster virus. Interestingly, the antiviral activity of these compounds appeared to be dependent on the activity of the HCMV UL97 kinase but was relatively unaffected by the absence of thymidine kinase activity in HSV. These data taken together indicate that the mechanism of action of these analogs is distinct from that of CPV. They also suggest that they might be useful as broad-spectrum antiherpesvirus agents and may be effective in the treatment of resistant virus infections.
- Published
- 2013
- Full Text
- View/download PDF
10. Selection and recombinant phenotyping of a novel CMX001 and cidofovir resistance mutation in human cytomegalovirus.
- Author
-
James SH, Price NB, Hartline CB, Lanier ER, and Prichard MN
- Subjects
- Amino Acid Sequence, Base Sequence, Cells, Cultured, Chromosomes, Artificial, Bacterial, Cidofovir, Cytomegalovirus genetics, Cytomegalovirus Infections drug therapy, Cytomegalovirus Infections virology, Cytosine pharmacology, DNA-Directed DNA Polymerase chemistry, DNA-Directed DNA Polymerase genetics, Foscarnet pharmacology, Ganciclovir pharmacology, Humans, Phenotype, Sequence Analysis, DNA, Antiviral Agents pharmacology, Cytomegalovirus drug effects, Cytosine analogs & derivatives, Drug Resistance, Viral genetics, Mutation, Organophosphonates pharmacology
- Abstract
CMX001 is an orally available lipid acyclic nucleotide phosphonate that delivers high intracellular levels of cidofovir (CDV)-diphosphate and exhibits enhanced in vitro antiviral activity against a wide range of double-stranded DNA viruses, including cytomegalovirus (CMV). Mutations in the DNA polymerase of CMV that impart resistance to CDV also render the virus resistant to CMX001. Here, we report a novel resistance mutation that arose under the selective pressure of CMX001. The wild-type CMV strain AD169 was propagated in human foreskin fibroblasts under increasing concentrations of CMX001 over 10 months, and the resulting strain (named CMX001(R)) was less susceptible to CDV and CMX001 in a plaque reduction assay. Genotypic analysis of virus strain CMX001(R) via conventional sequencing of the genes encoding the CMV DNA polymerase (UL54) and UL97 kinase (UL97) demonstrated one mutation that changed the wild-type aspartate to glutamate at position 542 in UL54. A recombinant virus with this novel D542E mutation was generated via bacterial artificial chromosome-mediated marker transfer experiments. Subsequent phenotypic resistance analysis of the D542E mutant demonstrated reductions in susceptibility of greater than 10-fold to CMX001 and CDV, but no resistance to foscarnet (FOS) or ganciclovir (GCV). Analysis of replicative fitness showed that both strain CMX001(R) and the D542E mutant viruses demonstrated a smaller plaque phenotype and slower replication kinetics than their respective parent viruses. These data describe the first resistance mutation generated under the selective pressure of CMX001 and suggest that CMX001 may have a unique resistance profile associated with reduced viral replication and maintenance of sensitivity to FOS and GCV.
- Published
- 2013
- Full Text
- View/download PDF
11. Human cytomegalovirus UL97 kinase alters the accumulation of CDK1.
- Author
-
Gill RB, James SH, and Prichard MN
- Subjects
- CDC2 Protein Kinase genetics, Cell Line, Cytomegalovirus genetics, Humans, Phosphotransferases (Alcohol Group Acceptor) genetics, Protein Transport, Reverse Transcriptase Polymerase Chain Reaction, Virus Replication, CDC2 Protein Kinase metabolism, Cytomegalovirus metabolism, Gene Expression Regulation physiology, Gene Expression Regulation, Enzymologic physiology, Phosphotransferases (Alcohol Group Acceptor) metabolism
- Abstract
The UL97 protein kinase is a serine/threonine kinase expressed by human cytomegalovirus (CMV) that phosphorylates ganciclovir. An investigation of the subcellular localization of pUL97 in infected cells indicated that, early in infection, pUL97 localized to focal sites in the nucleus that transitioned to subnuclear compartments and eventually throughout the entire nucleus. When UL97 kinase activity was eliminated with a K355M mutation or pharmacologically inhibited with maribavir, the expansion and redistribution of pUL97 foci within the nucleus was delayed, nuclear reorganization did not occur and assembly complexes in the cytoplasm failed to form normally. As UL97 kinase and its homologues appear to be functionally related to CDK1, a known regulator of nuclear structural organization, the effects of the UL97 kinase on CDK1 were investigated. Expression of CDK1 in infected cells appeared to be induced by UL97 kinase activity at the level of transcription and was not tied to other virus life-cycle events, such as viral DNA replication or virion assembly. These results suggest that, in addition to phosphorylating CDK1 targets, the UL97 kinase modifies G₂/M cell-cycle checkpoint regulators, specifically CDK1, to promote virus replication.
- Published
- 2012
- Full Text
- View/download PDF
12. The natural antimicrobial peptide subtilosin acts synergistically with glycerol monolaurate, lauric arginate, and ε-poly-L-lysine against bacterial vaginosis-associated pathogens but not human lactobacilli.
- Author
-
Noll KS, Prichard MN, Khaykin A, Sinko PJ, and Chikindas ML
- Subjects
- Arginine pharmacology, Drug Synergism, Female, Gardnerella vaginalis drug effects, Humans, Kinetics, Microbial Sensitivity Tests, Polylysine pharmacology, Anti-Bacterial Agents pharmacology, Arginine analogs & derivatives, Bacteria drug effects, Bacteriocins pharmacology, Lactobacillus drug effects, Laurates pharmacology, Monoglycerides pharmacology, Peptides, Cyclic pharmacology, Vaginosis, Bacterial microbiology
- Abstract
Subtilosin is a cyclical antimicrobial peptide produced by Bacillus amyloliquefaciens that has antimicrobial activity against the bacterial vaginosis-associated human pathogen Gardnerella vaginalis. The ability of subtilosin to inhibit G. vaginalis alone and in combination with the natural antimicrobial agents glycerol monolaurate (Lauricidin), lauric arginate, and ε-poly-L-lysine was tested using a checkerboard approach. Subtilosin was found to act synergistically with all of the chosen antimicrobials. These promising results indicate that lower concentrations of subtilosin in combination with other compounds could effectively be used to inhibit growth of the pathogen, thereby decreasing the risk of developed antimicrobial resistance. This is the first report on the effects of subtilosin combined with other natural antimicrobials against G. vaginalis.
- Published
- 2012
- Full Text
- View/download PDF
13. Identification of protein-protein interaction inhibitors targeting vaccinia virus processivity factor for development of antiviral agents.
- Author
-
Schormann N, Sommers CI, Prichard MN, Keith KA, Noah JW, Nuth M, Ricciardi RP, and Chattopadhyay D
- Subjects
- Cell Line, DNA Glycosylases metabolism, Humans, Protein Binding, Antiviral Agents pharmacology, Vaccinia virus drug effects, Viral Proteins metabolism
- Abstract
Poxvirus uracil DNA glycosylase D4 in association with A20 and the catalytic subunit of DNA polymerase forms the processive polymerase complex. The binding of D4 and A20 is essential for processive polymerase activity. Using an AlphaScreen assay, we identified compounds that inhibit protein-protein interactions between D4 and A20. Effective interaction inhibitors exhibited both antiviral activity and binding to D4. These results suggest that novel antiviral agents that target the protein-protein interactions between D4 and A20 can be developed for the treatment of infections with poxviruses, including smallpox.
- Published
- 2011
- Full Text
- View/download PDF
14. CMX001 potentiates the efficacy of acyclovir in herpes simplex virus infections.
- Author
-
Prichard MN, Kern ER, Hartline CB, Lanier ER, and Quenelle DC
- Subjects
- Acyclovir therapeutic use, Acyclovir toxicity, Animals, Antiviral Agents therapeutic use, Antiviral Agents toxicity, Cells, Cultured, Cytosine pharmacology, Cytosine therapeutic use, Cytosine toxicity, Drug Resistance, Viral, Drug Synergism, Drug Therapy, Combination, Female, Herpes Simplex virology, Humans, Mice, Mice, Inbred BALB C, Organophosphonates therapeutic use, Organophosphonates toxicity, Acyclovir pharmacology, Antiviral Agents pharmacology, Cytosine analogs & derivatives, Herpes Simplex drug therapy, Herpesvirus 1, Human drug effects, Herpesvirus 2, Human drug effects, Organophosphonates pharmacology
- Abstract
Although acyclovir (ACV) has proven to be of value in the therapy of certain herpes simplex virus (HSV) infections, there is a need for more effective therapies, particularly for serious infections in neonates and immunocompromised individuals, where resistance to this drug can be problematic. CMX001 is an orally bioavailable lipid conjugate of cidofovir that is substantially less nephrotoxic than the parent drug and has excellent antiviral activity against all the human herpesviruses. This compound retains full antiviral activity against ACV-resistant laboratory and clinical isolates. The combined efficacy of CMX001 and ACV was evaluated in a new real-time PCR combination assay, which demonstrated that the combination synergistically inhibited the replication of HSV in cell culture. This was also confirmed in murine models of HSV infection, where the combined therapy with these two drugs synergistically reduced mortality. These results suggest that CMX001 may be effective in the treatment of ACV-resistant HSV infections and as an adjunct therapy in individuals with suboptimal responses to ACV.
- Published
- 2011
- Full Text
- View/download PDF
15. Cyclopropavir inhibits the normal function of the human cytomegalovirus UL97 kinase.
- Author
-
James SH, Hartline CB, Harden EA, Driebe EM, Schupp JM, Engelthaler DM, Keim PS, Bowlin TL, Kern ER, and Prichard MN
- Subjects
- Animals, Base Sequence, Benzimidazoles pharmacology, COS Cells, Cell Line, Chlorocebus aethiops, Cytomegalovirus genetics, Cytomegalovirus isolation & purification, Drug Resistance, Viral genetics, Frameshift Mutation, Ganciclovir pharmacology, Guanine pharmacology, Herpesvirus 6, Human drug effects, Herpesvirus 8, Human drug effects, High-Throughput Nucleotide Sequencing, Humans, Phosphotransferases (Alcohol Group Acceptor) metabolism, Ribonucleosides pharmacology, Sequence Analysis, DNA, Antiviral Agents pharmacology, Cyclopropanes pharmacology, Cytomegalovirus drug effects, Cytomegalovirus enzymology, DNA, Viral biosynthesis, Guanine analogs & derivatives, Phosphotransferases (Alcohol Group Acceptor) antagonists & inhibitors
- Abstract
Cyclopropavir (CPV) is active against human cytomegalovirus (CMV), as well as both variants of human herpesvirus 6 and human herpesvirus 8. The mechanism of action of CPV against CMV is similar to that of ganciclovir (GCV) in that it is phosphorylated initially by the CMV UL97 kinase, resulting in inhibition of viral DNA synthesis. Resistance to CPV maps to the UL97 kinase but is associated primarily with H520Q mutations and thus retains good antiviral activity against most GCV-resistant isolates. An examination of CMV-infected cultures treated with CPV revealed unusual cell morphology typically associated with the absence of UL97 kinase activity. A surrogate assay for UL97 kinase activity confirmed that CPV inhibited the activity of this enzyme and that its action was similar to the inhibition seen with maribavir (MBV) in this assay. Combination studies using real-time PCR indicated that, like MBV, CPV also antagonized the efficacy of GCV and were consistent with the observed inhibition of the UL97 kinase. Deep sequencing of CPV-resistant laboratory isolates identified a frameshift mutation in UL27, presumably to compensate for a loss of UL97 enzymatic activity. We conclude that the mechanism of action of CPV against CMV is complex and involves both the inhibition of DNA synthesis and the inhibition of the normal activity of the UL97 kinase.
- Published
- 2011
- Full Text
- View/download PDF
16. Benzimidazole analogs inhibit human herpesvirus 6.
- Author
-
Prichard MN, Frederick SL, Daily S, Borysko KZ, Townsend LB, Drach JC, and Kern ER
- Subjects
- Antiviral Agents chemistry, Benzimidazoles chemistry, Cytomegalovirus drug effects, Humans, Virus Replication drug effects, Antiviral Agents pharmacology, Benzimidazoles pharmacology, Herpesvirus 6, Human drug effects
- Abstract
Several benzimidazole nucleoside analogs, including 1H-β-D-ribofuranosyl-2-bromo-5,6-dichlorobenzimidazole (BDCRB) and 1H-β-L-ribofuranosyl-2-isopropylamino-5,6-dichlorobenzimidazole (maribavir [MBV]), inhibit the replication of human cytomegalovirus. Neither analog inhibited the related betaherpesvirus human herpesvirus 6 (HHV-6). Additional analogs of these compounds were evaluated against both variants of HHV-6, and two L-analogs of BDCRB had good antiviral activity against HHV-6A, as well as more modest inhibition of HHV-6B replication.
- Published
- 2011
- Full Text
- View/download PDF
17. Antiviral Activity of 4'-thioIDU and Thymidine Analogs against Orthopoxviruses.
- Author
-
Prichard MN and Kern ER
- Abstract
The search for effective therapies for orthopoxvirus infections has identified diverse classes of molecules with antiviral activity. Pyrimidine analogs, such as 5-iodo-2'-deoxyuridine (idoxuridine, IDU) were among the first compounds identified with antiviral activity against a number of orthopoxviruses and have been reported to be active both in vitro and in animal models of infection. More recently, additional analogs have been reported to have improved antiviral activity against orthopoxviruses including several derivatives of deoxyuridine with large substituents in the 5 position, as well as analogs with modifications in the deoxyribose moiety including (north)-methanocarbathymidine, and 5-iodo-4'-thio-2'-deoxyuridine (4'-thioIDU). The latter molecule has proven to have good antiviral activity against the orthopoxviruses both in vitro and in vivo and has the potential to be an effective therapy in humans.
- Published
- 2010
- Full Text
- View/download PDF
18. Triple combination of amantadine, ribavirin, and oseltamivir is highly active and synergistic against drug resistant influenza virus strains in vitro.
- Author
-
Nguyen JT, Hoopes JD, Le MH, Smee DF, Patick AK, Faix DJ, Blair PJ, de Jong MD, Prichard MN, and Went GT
- Subjects
- Animals, Antiviral Agents pharmacology, Cell Line, Drug Synergism, Influenza A Virus, H1N1 Subtype drug effects, Influenza A Virus, H3N2 Subtype drug effects, Inhibitory Concentration 50, Amantadine pharmacology, Drug Resistance, Viral drug effects, Orthomyxoviridae genetics, Oseltamivir pharmacology, Ribavirin pharmacology
- Abstract
The rapid emergence and subsequent spread of the novel 2009 Influenza A/H1N1 virus (2009 H1N1) has prompted the World Health Organization to declare the first pandemic of the 21st century, highlighting the threat of influenza to public health and healthcare systems. Widespread resistance to both classes of influenza antivirals (adamantanes and neuraminidase inhibitors) occurs in both pandemic and seasonal viruses, rendering these drugs to be of marginal utility in the treatment modality. Worldwide, virtually all 2009 H1N1 and seasonal H3N2 strains are resistant to the adamantanes (rimantadine and amantadine), and the majority of seasonal H1N1 strains are resistant to oseltamivir, the most widely prescribed neuraminidase inhibitor (NAI). To address the need for more effective therapy, we evaluated the in vitro activity of a triple combination antiviral drug (TCAD) regimen composed of drugs with different mechanisms of action against drug-resistant seasonal and 2009 H1N1 influenza viruses. Amantadine, ribavirin, and oseltamivir, alone and in combination, were tested against amantadine- and oseltamivir-resistant influenza A viruses using an in vitro infection model in MDCK cells. Our data show that the triple combination was highly synergistic against drug-resistant viruses, and the synergy of the triple combination was significantly greater than the synergy of any double combination tested (P<0.05), including the combination of two NAIs. Surprisingly, amantadine and oseltamivir contributed to the antiviral activity of the TCAD regimen against amantadine- and oseltamivir-resistant viruses, respectively, at concentrations where they had no activity as single agents, and at concentrations that were clinically achievable. Our data demonstrate that the TCAD regimen composed of amantadine, ribavirin, and oseltamivir is highly synergistic against resistant viruses, including 2009 H1N1. The TCAD regimen overcomes baseline drug resistance to both classes of approved influenza antivirals, and thus may represent a highly active antiviral therapy for seasonal and pandemic influenza.
- Published
- 2010
- Full Text
- View/download PDF
19. Inhibition of herpesvirus replication by 5-substituted 4'-thiopyrimidine nucleosides.
- Author
-
Prichard MN, Quenelle DC, Hartline CB, Harden EA, Jefferson G, Frederick SL, Daily SL, Whitley RJ, Tiwari KN, Maddry JA, Secrist JA 3rd, and Kern ER
- Subjects
- Animals, Antiviral Agents adverse effects, Antiviral Agents chemistry, Cell Line, Cell Proliferation drug effects, Cells, Cultured, DNA-Directed DNA Polymerase genetics, DNA-Directed DNA Polymerase physiology, Drug Resistance, Viral drug effects, Drug Resistance, Viral genetics, Fluorescent Antibody Technique, Indirect, Herpesviridae genetics, Herpesviridae Infections virology, Herpesvirus 1, Human drug effects, Herpesvirus 1, Human genetics, Herpesvirus 2, Human drug effects, Herpesvirus 2, Human genetics, Herpesvirus 6, Human drug effects, Herpesvirus 6, Human genetics, Herpesvirus 8, Human drug effects, Herpesvirus 8, Human genetics, Humans, Mice, Mice, Inbred BALB C, Microscopy, Fluorescence, Molecular Structure, Pyrimidine Nucleosides chemical synthesis, Pyrimidine Nucleosides chemistry, Viral Proteins genetics, Viral Proteins physiology, Antiviral Agents pharmacology, Antiviral Agents therapeutic use, Herpesviridae drug effects, Herpesviridae Infections drug therapy, Pyrimidine Nucleosides pharmacology, Pyrimidine Nucleosides therapeutic use, Virus Replication drug effects
- Abstract
A series of 4'-thionucleosides were synthesized and evaluated for activities against orthopoxviruses and herpesviruses. We reported previously that one analog, 5-iodo-4'-thio-2'-deoxyuridine (4'-thioIDU), exhibits good activity both in vitro and in vivo against two orthopoxviruses. This compound also has good activity in cell culture against many of the herpesviruses. It inhibited the replication of herpes simplex virus type 1 (HSV-1), HSV-2, and varicella-zoster virus with 50% effective concentrations (EC(50)s) of 0.1, 0.5, and 2 microM, respectively. It also inhibited the replication of human cytomegalovirus (HCMV) with an EC(50) of 5.9 microM but did not selectively inhibit Epstein-Barr virus, human herpesvirus 6, or human herpesvirus 8. While acyclovir-resistant strains of HSV-1 and HSV-2 were comparatively resistant to 4'-thioIDU, it retained modest activity (EC(50)s of 4 to 12 microM) against these strains. Some ganciclovir-resistant strains of HCMV also exhibited reduced susceptibilities to the compound, which appeared to be related to the specific mutations in the DNA polymerase, consistent with the observed incorporation of the compound into viral DNA. The activity of 4'-thioIDU was also evaluated using mice infected intranasally with the MS strain of HSV-2. Although there was no decrease in final mortality rates, the mean length of survival after inoculation increased significantly (P < 0.05) for all animals receiving 4'-thioIDU. The findings from the studies presented here suggest that 4'-thioIDU is a good inhibitor of some herpesviruses, as well as orthopoxviruses, and this class of compounds warrants further study as a therapy for infections with these viruses.
- Published
- 2009
- Full Text
- View/download PDF
20. Triple combination of oseltamivir, amantadine, and ribavirin displays synergistic activity against multiple influenza virus strains in vitro.
- Author
-
Nguyen JT, Hoopes JD, Smee DF, Prichard MN, Driebe EM, Engelthaler DM, Le MH, Keim PS, Spence RP, and Went GT
- Subjects
- Animals, Cell Line, Dogs, Drug Synergism, Influenza A Virus, H1N1 Subtype drug effects, Influenza A Virus, H3N2 Subtype drug effects, Influenza A Virus, H5N1 Subtype drug effects, Polymerase Chain Reaction, Amantadine pharmacology, Antiviral Agents pharmacology, Orthomyxoviridae drug effects, Oseltamivir pharmacology, Ribavirin pharmacology
- Abstract
The recurring emergence of influenza virus strains that are resistant to available antiviral medications has become a global health concern, especially in light of the potential for a new influenza virus pandemic. Currently, virtually all circulating strains of influenza A virus in the United States are resistant to either of the two major classes of anti-influenza drugs (adamantanes and neuraminidase inhibitors). Thus, new therapeutic approaches that can be rapidly deployed and that will address the issue of recurring resistance should be developed. We have tested double and triple combinations of the approved anti-influenza drugs oseltamivir and amantadine together with ribavirin against three influenza virus strains using cytopathic effect inhibition assays in MDCK cells. We selected A/New Caledonia/20/99 (H1N1) and A/Sydney/05/97 (H3N2) as representatives of the wild-type versions of the predominant circulating seasonal influenza virus strains and A/Duck/MN/1525/81 (H5N1) as a representative of avian influenza virus strains. Dose-response curves were generated for all drug combinations, and the degree of drug interaction was quantified using a model that calculates the synergy (or antagonism) between the drugs in double and triple combinations. This report demonstrates that a triple combination of antivirals was highly synergistic against influenza A virus. Importantly, the synergy of the triple combination was 2- to 13-fold greater than the synergy of any double combination depending on the influenza virus subtype. These data support the investigation of a novel combination of oseltamivir, amantadine, and ribavirin as an effective treatment for both seasonal and pandemic influenza virus, allowing the efficient use of the existing drug supplies.
- Published
- 2009
- Full Text
- View/download PDF
21. Activities of certain 5-substituted 4'-thiopyrimidine nucleosides against orthopoxvirus infections.
- Author
-
Kern ER, Prichard MN, Quenelle DC, Keith KA, Tiwari KN, Maddry JA, and Secrist JA 3rd
- Subjects
- Animals, Cell Line, Cell Proliferation drug effects, Cell Survival drug effects, Cowpox drug therapy, Cytopathogenic Effect, Viral drug effects, DNA, Viral biosynthesis, DNA, Viral genetics, Drug Resistance, Viral, Humans, Mice, Mice, Inbred BALB C, Poxviridae Infections virology, Structure-Activity Relationship, Vaccinia drug therapy, beta-Galactosidase metabolism, Antiviral Agents therapeutic use, Nucleosides therapeutic use, Orthopoxvirus, Poxviridae Infections drug therapy
- Abstract
As part of a program to identify new compounds that have activity against orthopoxviruses, a number of 4'-thionucleosides were synthesized and evaluated for their efficacies against vaccinia and cowpox viruses. Seven compounds that were active at about 1 microM against both viruses in human cells but that did not have significant toxicity were identified. The 5-iodo analog, 1-(2-deoxy-4-thio-beta-d-ribofuranosyl)-5-iodouracil (4'-thioIDU), was selected as a representative molecule; and this compound also inhibited viral DNA synthesis at less than 1 microM but only partially inhibited the replication of a recombinant vaccinia virus that lacked a thymidine kinase. This compound retained complete activity against cidofovir- and ST-246-resistant mutants. To determine if this analog had activity in an animal model, mice were infected intranasally with vaccinia or cowpox virus and treatment with 4'-thioIDU was given intraperitoneally or orally twice daily at 50, 15, 5, or 1.5 mg/kg of body weight beginning at 24 to 120 h postinfection and was continued for 5 days. Almost complete protection (87%) was observed when treatment with 1.5 mg/kg was begun at 72 h postinfection, and significant protection (73%) was still obtained when treatment with 5 mg/kg was initiated at 96 h. Virus titers in the liver, spleen, and kidney were reduced by about 4 log(10) units and about 2 log(10) units in mice infected with vaccinia virus and cowpox virus, respectively. These results indicate that 4'-thioIDU is a potent, nontoxic inhibitor of orthopoxvirus replication in cell culture and experimental animal infections and suggest that it may have potential for use in the treatment of orthopoxvirus infections in animals and humans.
- Published
- 2009
- Full Text
- View/download PDF
22. Conserved retinoblastoma protein-binding motif in human cytomegalovirus UL97 kinase minimally impacts viral replication but affects susceptibility to maribavir.
- Author
-
Gill RB, Frederick SL, Hartline CB, Chou S, and Prichard MN
- Subjects
- Amino Acid Motifs, Cell Line, Cells, Cultured, Cytomegalovirus genetics, Cytomegalovirus physiology, Humans, Mutation, Phosphotransferases (Alcohol Group Acceptor) genetics, Phosphotransferases (Alcohol Group Acceptor) metabolism, Retinoblastoma Protein genetics, Retinoblastoma Protein metabolism, Benzimidazoles pharmacology, Cytomegalovirus drug effects, Cytomegalovirus enzymology, Enzyme Inhibitors pharmacology, Phosphotransferases (Alcohol Group Acceptor) chemistry, Ribonucleosides pharmacology, Virus Replication
- Abstract
The UL97 kinase has been shown to phosphorylate and inactivate the retinoblastoma protein (Rb) and has three consensus Rb-binding motifs that might contribute to this activity. Recombinant viruses containing mutations in the Rb-binding motifs generally replicated well in human foreskin fibroblasts with only a slight delay in replication kinetics. Their susceptibility to the specific UL97 kinase inhibitor, maribavir, was also examined. Mutation of the amino terminal motif, which is involved in the inactivation of Rb, also renders the virus hypersensitive to the drug and suggests that the motif may play a role in its mechanism of action.
- Published
- 2009
- Full Text
- View/download PDF
23. Inhibition of herpesvirus replication by hexadecyloxypropyl esters of purine- and pyrimidine-based phosphonomethoxyethyl nucleoside phosphonates.
- Author
-
Prichard MN, Hartline CB, Harden EA, Daily SL, Beadle JR, Valiaeva N, Kern ER, and Hostetler KY
- Subjects
- Antiviral Agents chemistry, Cell Line, Esters chemistry, Herpesviridae classification, Herpesviridae physiology, Humans, Organophosphonates chemistry, Purine Nucleosides chemistry, Pyrimidine Nucleosides chemistry, Viral Plaque Assay, Antiviral Agents pharmacology, Esters pharmacology, Herpesviridae drug effects, Organophosphonates pharmacology, Purine Nucleosides pharmacology, Pyrimidine Nucleosides pharmacology, Virus Replication drug effects
- Abstract
Patients infected with human immunodeficiency virus (HIV) often suffer from herpesvirus infections as a result of immunosuppression. These infections can occur while patients are receiving antiretroviral therapy, and additional drugs required to treat their infection can adversely affect compliance. It would be useful to have antivirals with a broader spectrum of activity that included both HIV and the herpesviruses. We reported previously that alkoxyalkyl ester prodrugs of cidofovir are up to 3 orders of magnitude more active against herpesvirus replication and may be less toxic than the unmodified drug. To determine if this strategy would be effective for certain phosphonomethoxyethyl nucleoside phosphonates which are also active against HIV infections, the hexadecyloxypropyl (HDP) esters of 1-(phosphonomethoxyethyl)-cytosine, 1-(phosphonomethoxyethyl)-5-bromo-cytosine (PME-5BrC), 1-(phosphonomethoxyethyl)-5-fluoro-cytosine, 9-(phosphonomethoxyethyl)-2,6-diaminopurine (PME-DAP), and 9-(phosphonomethoxyethyl)-2-amino-6-cyclopropylaminopurine (PME-cPrDAP) were evaluated for activity against herpesvirus replication. The HDP esters were substantially more active than the unmodified acyclic nucleoside phosphonates, indicating that esterification with alkoxyalkyl groups increases the antiviral activity of many acyclic nucleoside phosphonates. The most interesting compounds included HDP-PME-cPrDAP and HDP-PME-DAP, which were 12- to 43-fold more active than the parent nucleoside phosphonates against herpes simplex virus and cytomegalovirus, and HDP-PME-cPrDAP and HDP-PME-5BrC which were especially active against Epstein-Barr virus. The results presented here indicate that HDP-esterified acyclic nucleoside phosphonates with antiviral activity against HIV also inhibit the replication of some herpesviruses and can extend the spectrum of activity for these compounds.
- Published
- 2008
- Full Text
- View/download PDF
24. Isolation and characterization of cidofovir resistant vaccinia viruses.
- Author
-
Becker MN, Obraztsova M, Kern ER, Quenelle DC, Keith KA, Prichard MN, Luo M, and Moyer RW
- Subjects
- Animals, Antiviral Agents pharmacology, Cell Line, Chlorocebus aethiops, Cidofovir, Cytosine pharmacology, DNA-Directed DNA Polymerase chemistry, DNA-Directed DNA Polymerase genetics, DNA-Directed DNA Polymerase metabolism, Female, Humans, Mice, Models, Molecular, Mutation, Vaccinia mortality, Vaccinia virology, Vaccinia virus pathogenicity, Vaccinia virus physiology, Vero Cells, Viral Plaque Assay, Virulence, Cytosine analogs & derivatives, Drug Resistance, Viral, Organophosphonates pharmacology, Vaccinia virus drug effects, Vaccinia virus isolation & purification
- Abstract
Background: The emergence of drug resistant viruses, together with the possibility of increased virulence, is an important concern in the development of new antiviral compounds. Cidofovir (CDV) is a phosphonate nucleotide that is approved for use against cytomegalovirus retinitis and for the emergency treatment of smallpox or complications following vaccination. One mode of action for CDV has been demonstrated to be the inhibition of the viral DNA polymerase., Results: We have isolated several CDV resistant (CDVR) vaccinia viruses through a one step process, two of which have unique single mutations within the DNA polymerase. An additional resistant virus isolate provides evidence of a second site mutation within the genome involved in CDV resistance. The CDVR viruses were 3-7 fold more resistant to the drug than the parental viruses. The virulence of the CDVR viruses was tested in mice inoculated intranasally and all were found to be attenuated., Conclusion: Resistance to CDV in vaccinia virus can be conferred individually by at least two different mutations within the DNA polymerase gene. Additional genes may be involved. This one step approach for isolating resistant viruses without serial passage and in the presence of low doses of drug minimizes unintended secondary mutations and is applicable to other potential antiviral agents.
- Published
- 2008
- Full Text
- View/download PDF
25. Human cytomegalovirus UL97 kinase activity is required for the hyperphosphorylation of retinoblastoma protein and inhibits the formation of nuclear aggresomes.
- Author
-
Prichard MN, Sztul E, Daily SL, Perry AL, Frederick SL, Gill RB, Hartline CB, Streblow DN, Varnum SM, Smith RD, and Kern ER
- Subjects
- Amino Acid Sequence, Animals, Binding Sites, Cell Line, Cell Nucleus chemistry, Chlorocebus aethiops, Chromatography, Liquid, Conserved Sequence, Cytomegalovirus genetics, Cytoplasm chemistry, Humans, Mass Spectrometry, Microscopy, Fluorescence, Molecular Sequence Data, Phosphorylation, Phosphotransferases (Alcohol Group Acceptor) genetics, Protein Binding, Proteins isolation & purification, Sequence Alignment, Cytomegalovirus physiology, Phosphotransferases (Alcohol Group Acceptor) metabolism, Retinoblastoma Protein metabolism
- Abstract
Cells infected with human cytomegalovirus in the absence of UL97 kinase activity produce large nuclear aggregates that sequester considerable quantities of viral proteins. A transient expression assay suggested that pp71 and IE1 were also involved in this process, and this suggestion was significant, since both proteins have been reported to interact with components of promyelocytic leukemia (PML) bodies (ND10) and also interact functionally with retinoblastoma pocket proteins (RB). PML bodies have been linked to the formation of nuclear aggresomes, and colocalization studies suggested that viral proteins were recruited to these structures and that UL97 kinase activity inhibited their formation. Proteins associated with PML bodies were examined by Western blot analysis, and pUL97 appeared to specifically affect the phosphorylation of RB in a kinase-dependent manner. Three consensus RB binding motifs were identified in the UL97 kinase, and recombinant viruses were constructed in which each was mutated to assess a potential role in the phosphorylation of RB and the inhibition of nuclear aggresome formation. The mutation of either the conserved LxCxE RB binding motif or the lysine required for kinase activity impaired the ability of the virus to stabilize and phosphorylate RB. We concluded from these studies that both UL97 kinase activity and the LxCxE RB binding motif are required for the phosphorylation and stabilization of RB in infected cells and that this effect can be antagonized by the antiviral drug maribavir. These data also suggest a potential link between RB function and the formation of aggresomes.
- Published
- 2008
- Full Text
- View/download PDF
26. Vaccinia virus lacking the deoxyuridine triphosphatase gene (F2L) replicates well in vitro and in vivo, but is hypersensitive to the antiviral drug (N)-methanocarbathymidine.
- Author
-
Prichard MN, Kern ER, Quenelle DC, Keith KA, Moyer RW, and Turner PC
- Subjects
- Animals, Female, Green Fluorescent Proteins metabolism, Humans, Mice, Mice, Inbred BALB C, Pyrimidines metabolism, Pyrophosphatases genetics, Recombinant Fusion Proteins metabolism, Sequence Deletion, Thymidine pharmacology, Vaccinia virus drug effects, Vaccinia virus enzymology, Vaccinia virus pathogenicity, Viral Plaque Assay, Virulence, Antiviral Agents pharmacology, Pyrophosphatases metabolism, Thymidine analogs & derivatives, Vaccinia virology, Vaccinia virus physiology, Virus Replication drug effects
- Abstract
Background: The vaccinia virus (VV) F2L gene encodes a functional deoxyuridine triphosphatase (dUTPase) that catalyzes the conversion of dUTP to dUMP and is thought to minimize the incorporation of deoxyuridine residues into the viral genome. Previous studies with with a complex, multigene deletion in this virus suggested that the gene was not required for viral replication, but the impact of deleting this gene alone has not been determined in vitro or in vivo. Although the crystal structure for this enzyme has been determined, its potential as a target for antiviral therapy is unclear., Results: The F2L gene was replaced with GFP in the WR strain of VV to assess its effect on viral replication. The resulting virus replicated well in cell culture and its replication kinetics were almost indistinguishable from those of the wt virus and attained similar titers. The virus also appeared to be as pathogenic as the WR strain suggesting that it also replicated well in mice. Cells infected with the dUTPase mutant would be predicted to affect pyrimidine deoxynucleotide pools and might be expected to exhibit altered susceptibility to pyrimidine analogs. The antiviral activity of cidofovir and four thymidine analogs were evaluated both in the mutant and the parent strain of this virus. The dUTPase knockout remained fully susceptible to cidofovir and idoxuridine, but was hypersensitive to the drug (N)-methanocarbathymidine, suggesting that pyrimidine metabolism was altered in cells infected with the mutant virus. The absence of dUTPase should reduce cellular dUMP pools and may result in a reduced conversion to dTMP by thymidylate synthetase or an increased reliance on the salvage of thymidine by the viral thymidine kinase., Conclusion: We confirmed that F2L was not required for replication in cell culture and determined that it does not play a significant role on virulence of the virus in intranasally infected mice. The recombinant virus is hypersensitive to (N)-methanocarbathymidine and may reflect metabolic differences in the mutant virus.
- Published
- 2008
- Full Text
- View/download PDF
27. Synergistic efficacy of the combination of ST-246 with CMX001 against orthopoxviruses.
- Author
-
Quenelle DC, Prichard MN, Keith KA, Hruby DE, Jordan R, Painter GR, Robertson A, and Kern ER
- Subjects
- Administration, Oral, Animals, Antiviral Agents pharmacology, Antiviral Agents therapeutic use, Benzamides administration & dosage, Benzamides therapeutic use, Cells, Cultured, Chlorocebus aethiops, Cowpox virus drug effects, Cytosine pharmacology, Cytosine therapeutic use, Dose-Response Relationship, Drug, Drug Synergism, Drug Therapy, Combination, Female, Humans, Isoindoles administration & dosage, Isoindoles therapeutic use, Male, Mice, Mice, Inbred BALB C, Organophosphonates therapeutic use, Poxviridae Infections drug therapy, Poxviridae Infections virology, Treatment Outcome, Vaccinia virus drug effects, Vero Cells, Virus Replication drug effects, Benzamides pharmacology, Cytosine analogs & derivatives, Isoindoles pharmacology, Organophosphonates pharmacology, Orthopoxvirus drug effects
- Abstract
The combination of ST-246 and hexadecyloxypropyl-cidofovir or CMX001 was evaluated for synergistic activity in vitro against vaccinia virus and cowpox virus (CV) and in vivo against CV. In cell culture the combination was highly synergistic against both viruses, and the results suggested that combined treatment with these agents might offer superior efficacy in vivo. For animal models, ST-246 was administered orally with or without CMX001 to mice lethally infected with CV. Treatments began 1, 3, or 6 days postinfection using lower dosages than previously used for single-drug treatment. ST-246 was given at 10, 3, or 1 mg/kg of body weight with or without CMX001 at 3, 1, or 0.3 mg/kg to evaluate potential synergistic interactions. Treatment beginning 6 days post-viral inoculation with ST-246 alone only increased the mean day to death at 10 or 3 mg/kg but had no effect on survival. CMX001 alone also had no effect on survival. When the combination of the two drugs was begun 6 days after viral infection using various dosages of the two, a synergistic reduction in mortality was observed. No evidence of increased toxicity was noted with the combination either in vitro or in vivo. These results indicate that combinations of ST-246 and CMX001 are synergistic both in vitro and in vivo and suggest that combination therapy using ST-246 and CMX001 for treatment of orthopoxvirus disease in humans or animals may provide an additional benefit over the use of the two drugs by themselves.
- Published
- 2007
- Full Text
- View/download PDF
28. Selective phosphorylation of antiviral drugs by vaccinia virus thymidine kinase.
- Author
-
Prichard MN, Keith KA, Johnson MP, Harden EA, McBrayer A, Luo M, Qiu S, Chattopadhyay D, Fan X, Torrence PF, and Kern ER
- Subjects
- Amino Acid Sequence, Herpesviridae drug effects, Humans, Kinetics, Molecular Sequence Data, Orthopoxvirus drug effects, Phosphorylation, Thymidine Kinase chemistry, Antiviral Agents metabolism, Antiviral Agents pharmacology, Thymidine Kinase metabolism, Vaccinia virus enzymology
- Abstract
The antiviral activity of a new series of thymidine analogs was determined against vaccinia virus (VV), cowpox virus (CV), herpes simplex virus, and varicella-zoster virus. Several compounds were identified that had good activity against each of the viruses, including a set of novel 5-substituted deoxyuridine analogs. To investigate the possibility that these drugs might be phosphorylated preferentially by the viral thymidine kinase (TK) homologs, the antiviral activities of these compounds were also assessed using TK-deficient strains of some of these viruses. Some of these compounds were shown to be much less effective in the absence of a functional TK gene in CV, which was unexpected given the high degree of amino acid identity between this enzyme and its cellular homolog. This unanticipated result suggested that the CV TK was important in the mechanism of action of these compounds and also that it might phosphorylate a wider variety of substrates than other type II enzymes. To confirm these data, we expressed the VV TK and human TK1 in bacteria and isolated the purified enzymes. Enzymatic assays demonstrated that the viral TK could efficiently phosphorylate many of these compounds, whereas most of the compounds were very poor substrates for the cellular kinase, TK1. Thus, the specific phosphorylation of these compounds by the viral kinase may be sufficient to explain the TK dependence. This unexpected result suggests that selective phosphorylation by the viral kinase may be a promising new approach in the discovery of highly selective inhibitors of orthopoxvirus replication.
- Published
- 2007
- Full Text
- View/download PDF
29. Antiviral activities of novel 5-phosphono-pent-2-en-1-yl nucleosides and their alkoxyalkyl phosphonoesters.
- Author
-
Choo H, Beadle JR, Kern ER, Prichard MN, Keith KA, Hartline CB, Trahan J, Aldern KA, Korba BE, and Hostetler KY
- Subjects
- Antiviral Agents chemical synthesis, Antiviral Agents chemistry, Cytomegalovirus drug effects, HIV-1 drug effects, Hepatitis B virus drug effects, Humans, Nucleosides chemical synthesis, Nucleosides chemistry, Organophosphorus Compounds chemical synthesis, Organophosphorus Compounds chemistry, Antiviral Agents pharmacology, Nucleosides pharmacology, Organophosphorus Compounds pharmacology
- Abstract
Three acyclic nucleoside phosphonates are currently approved for clinical use against infections caused by cytomegalovirus (Vistide), hepatitis B virus (Hepsera), and human immunodeficiency virus type 1 (Viread). This important antiviral class inhibits viral polymerases after cellular uptake and conversion to their diphosphates, bypassing the first phosphorylation, which is required for conventional nucleoside antivirals. Small chemical alterations in the acyclic side chain lead to marked differences in antiviral activity and the spectrum of activity of acyclic nucleoside phosphonates against various classes of viral agents. We synthesized a new class of acyclic nucleoside phosphonates based on a 5-phosphono-pent-2-en-1-yl base motif in which the oxygen heteroatom usually present in acyclic nucleoside phosphonates has been replaced with a double bond. Since the intrinsic phosphonate moiety leads to low oral bioavailability and impaired cellular penetration, we also prepared the hexadecyloxypropyl esters of the 5-phosphono-pent-2-en-1-yl nucleosides. Our earlier work showed that this markedly increases antiviral activity and oral bioavailability. Although the 5-phosphono-pent-2-en-1-yl nucleosides themselves were not active, the hexadecyloxypropyl esters were active against DNA viruses and hepatitis B virus, in vitro. Notably, the hexadecyloxypropyl ester of 9-(5-phosphono-pent-2-en-1-yl)-adenine was active against hepatitis B virus mutants resistant to lamivudine, emtricitabine, and adefovir.
- Published
- 2007
- Full Text
- View/download PDF
30. Activity and mechanism of action of N-methanocarbathymidine against herpesvirus and orthopoxvirus infections.
- Author
-
Prichard MN, Keith KA, Quenelle DC, and Kern ER
- Subjects
- Animals, Cells, Cultured, DNA, Viral biosynthesis, Female, Humans, Mice, Mice, Inbred BALB C, Phosphorylation, Thymidine pharmacology, Thymidine therapeutic use, Thymidine Kinase metabolism, Antiviral Agents therapeutic use, Herpesviridae Infections drug therapy, Poxviridae Infections drug therapy, Thymidine analogs & derivatives
- Abstract
N-Methanocarbathymidine [(N)-MCT] is a conformationally locked nucleoside analog that is active against some herpesviruses and orthopoxviruses in vitro. The antiviral activity of this molecule is dependent on the type I thymidine kinase (TK) in herpes simplex virus and also appears to be dependent on the type II TK expressed by cowpox and vaccinia viruses, suggesting that it is a substrate for both of these divergent forms of the enzyme. The drug is also a good inhibitor of viral DNA synthesis in both viruses and is consistent with inhibition of the viral DNA polymerase once it is activated by the viral TK homologs. This mechanism of action explains the rather unusual spectrum of activity, which is limited to orthopoxviruses, alphaherpesviruses, and Epstein-Barr virus, since these viruses express molecules with TK activity that can phosphorylate and thus activate the drug. The compound is also effective in vivo and reduces the mortality of mice infected with orthopoxviruses, as well as those infected with herpes simplex virus type 1 when treatment is initiated 24 h after infection. These results indicate that (N)-MCT is active in vitro and in vivo, and its mechanism of action suggests that the molecule may be an effective therapeutic for orthopoxvirus and herpesvirus infections, thus warranting further development.
- Published
- 2006
- Full Text
- View/download PDF
31. Human cytomegalovirus UL27 is not required for viral replication in human tissue implanted in SCID mice.
- Author
-
Prichard MN, Quenelle DC, Bidanset DJ, Komazin G, Chou S, Drach JC, and Kern ER
- Subjects
- Animals, Cytomegalovirus genetics, Gene Deletion, Humans, Liver virology, Liver Transplantation, Mice, Mice, SCID, Retina transplantation, Retina virology, Thymus Gland transplantation, Thymus Gland virology, Viral Proteins genetics, Cytomegalovirus metabolism, Viral Proteins metabolism, Virus Replication physiology
- Abstract
Inhibition of the human cytomegalovirus UL97 kinase by maribavir is thought to be responsible for the antiviral activity of this compound. Some mutations that confer resistance to maribavir map to UL97, however additional mutations that also confer resistance to the drug were mapped to UL27. These open reading frames share a low level of homology, yet the function of pUL27 remains unknown. A recombinant virus with a deletion in the UL27 open reading frame was reported previously to exhibit a slight replication deficit, but a more important function in vivo was hypothesized given its homology to the UL97 kinase. The potential for an important function in vivo was investigated by determining if these knockout viruses could replicate in human tissue implanted in SCID mice. None of the AD169 derived viruses replicated well in the implanted thymus/liver tissue, and is consistent with previous observations, although all of the viruses replicated to some degree in retinal tissue implants. Replication of the parent viruses was observed at 7 days post inoculation, whereas no replication was detected with any of the recombinant viruses with deletions in UL27. By day 14, replication was detected in two of the three knockout viruses and in all of the viruses by day 42. These data are consistent with minimal defects observed in cell culture, but are not consistent with an important role for UL27 in vivo. We conclude that UL27 is not required for viral replication in vivo.
- Published
- 2006
- Full Text
- View/download PDF
32. Human cytomegalovirus UL97 Kinase is required for the normal intranuclear distribution of pp65 and virion morphogenesis.
- Author
-
Prichard MN, Britt WJ, Daily SL, Hartline CB, and Kern ER
- Subjects
- Animals, COS Cells, Cell Nucleus virology, Cells, Cultured, Chlorocebus aethiops, Cytomegalovirus enzymology, Fibroblasts virology, Gene Expression Regulation, Viral physiology, Humans, Skin, Cell Nucleus metabolism, Cytomegalovirus physiology, Morphogenesis physiology, Phosphoproteins metabolism, Phosphotransferases (Alcohol Group Acceptor) physiology, Viral Matrix Proteins metabolism, Virion physiology
- Abstract
Recombinant human cytomegaloviruses that do not express UL97 kinase activity exhibit a distinctive plaque morphology characterized by the formation of highly refractile bodies late in infection. These structures were also observed in infected cells treated with the UL97 kinase inhibitor maribavir. Nuclear inclusions were purified to near homogeneity, and the constituent proteins were identified by matrix-assisted laser desorption ionization-time-of-flight mass spectrometry. This analysis demonstrated that the aggregates were formed principally of the tegument proteins pp65 and ppUL25 but also contained additional virion structural proteins including the major capsid protein. Immunoblotting experiments confirmed these results and identified a number of additional viral proteins present in the purified tegument aggregates. Interestingly, the formation of these structures appeared to be dependent on pp65, since it was not induced in cells infected with a recombinant virus with this open reading frame deleted. Morphologically similar aggregates could be reproduced in nuclei of uninfected cells by overexpressing pp65, and their formation was prevented by coexpressing the UL97 kinase. Inhibition of UL97 kinase activity with maribavir or mutation of an essential amino acid in the kinase abolished its ability to prevent aggregate formation. These data taken together suggest that the UL97 kinase impacts the aggregation of pp65 in the nuclei of infected cells. We propose that the kinase plays an important role in the acquisition of tegument during virion morphogenesis in the nucleus and that this activity represents an important step in the production of mature virus particles.
- Published
- 2005
- Full Text
- View/download PDF
33. Comparative activities of lipid esters of cidofovir and cyclic cidofovir against replication of herpesviruses in vitro.
- Author
-
Williams-Aziz SL, Hartline CB, Harden EA, Daily SL, Prichard MN, Kushner NL, Beadle JR, Wan WB, Hostetler KY, and Kern ER
- Subjects
- Antiviral Agents chemistry, Cell Adhesion drug effects, Cell Proliferation drug effects, Cell Survival drug effects, Cells, Cultured, Cidofovir, Cyclization, Cytomegalovirus drug effects, Cytosine chemistry, Cytosine pharmacology, Enzyme-Linked Immunosorbent Assay, Esters pharmacology, Fibroblasts virology, Flow Cytometry, Humans, In Situ Hybridization, Organophosphonates chemistry, Structure-Activity Relationship, Tetrazolium Salts, Thiazoles, Viral Plaque Assay, Antiviral Agents pharmacology, Cytosine analogs & derivatives, Herpesviridae drug effects, Organophosphonates pharmacology, Virus Replication drug effects
- Abstract
Cidofovir (CDV) is an effective therapy for certain human cytomegalovirus (HCMV) infections in immunocompromised patients that are resistant to other antiviral drugs, but the compound is not active orally. To improve oral bioavailability, a series of lipid analogs of CDV and cyclic CDV (cCDV), including hexadecyloxypropyl-CDV and -cCDV and octadecyloxyethyl-CDV and -cCDV, were synthesized and found to have multiple-log-unit enhanced activity against HCMV in vitro. On the basis of the activity observed with these analogs, additional lipid esters were synthesized and evaluated for their activity against herpes simplex virus (HSV) types 1 and 2, human cytomegalovirus, murine cytomegalovirus, varicella-zoster virus (VZV), Epstein-Barr virus (EBV), human herpesvirus 6 (HHV-6), and HHV-8. Using several different in vitro assays, concentrations of drug as low as 0.001 microM reduced herpesvirus replication by 50% (EC50) with the CDV analogs, whereas the cCDV compounds were generally less active. In most of the assays performed, the EC50 values of the lipid esters were at least 100-fold lower than the EC50 values for unmodified CDV or cCDV. The lipid analogs were also active against isolates that were resistant to CDV, ganciclovir, or foscarnet. These results indicate that the lipid ester analogs are considerably more active than CDV itself against HSV, VZV, CMV, EBV, HHV-6, and HHV-8 in vitro, suggesting that they may have potential for the treatment of infections caused by a variety of herpesviruses.
- Published
- 2005
- Full Text
- View/download PDF
34. Human cytomegalovirus uracil DNA glycosylase associates with ppUL44 and accelerates the accumulation of viral DNA.
- Author
-
Prichard MN, Lawlor H, Duke GM, Mo C, Wang Z, Dixon M, Kemble G, and Kern ER
- Subjects
- Cells, Cultured, Cytomegalovirus genetics, DNA, Single-Stranded biosynthesis, DNA, Viral biosynthesis, DNA-Binding Proteins genetics, Humans, Uracil-DNA Glycosidase genetics, Viral Proteins genetics, Virus Replication, Cytomegalovirus metabolism, DNA-Binding Proteins metabolism, Uracil-DNA Glycosidase metabolism, Viral Proteins metabolism
- Abstract
Background: Human cytomegalovirus UL114 encodes a uracil-DNA glycosylase homolog that is highly conserved in all characterized herpesviruses that infect mammals. Previous studies demonstrated that the deletion of this nonessential gene delays significantly the onset of viral DNA synthesis and results in a prolonged replication cycle. The gene product, pUL114, also appears to be important in late phase DNA synthesis presumably by introducing single stranded breaks., Results: A series of experiments was performed to formally assign the observed phenotype to pUL114 and to characterize the function of the protein in viral replication. A cell line expressing pUL114 complemented the observed phenotype of a UL114 deletion virus in trans, confirming that the observed defects were the result of a deficiency in this gene product. Stocks of recombinant viruses without elevated levels of uracil were produced in the complementing cells; however they retained the phenotype of poor growth in normal fibroblasts suggesting that poor replication was unrelated to uracil content of input genomes. Recombinant viruses expressing epitope tagged versions of this gene demonstrated that pUL114 was expressed at early times and that it localized to viral replication compartments. This protein also coprecipitated with the DNA polymerase processivity factor, ppUL44 suggesting that these proteins associate in infected cells. This apparent interaction did not appear to require other viral proteins since ppUL44 could recruit pUL114 to the nucleus in uninfected cells. An analysis of DNA replication kinetics revealed that the initial rate of DNA synthesis and the accumulation of progeny viral genomes were significantly reduced compared to the parent virus., Conclusion: These data suggest that pUL114 associates with ppUL44 and that it functions as part of the viral DNA replication complex to increase the efficiency of both early and late phase viral DNA synthesis.
- Published
- 2005
- Full Text
- View/download PDF
35. In vitro activity and mechanism of action of methylenecyclopropane analogs of nucleosides against herpesvirus replication.
- Author
-
Kern ER, Kushner NL, Hartline CB, Williams-Aziz SL, Harden EA, Zhou S, Zemlicka J, and Prichard MN
- Subjects
- Ganciclovir pharmacology, Herpesviridae physiology, Antiviral Agents pharmacology, Herpesviridae drug effects, Nucleosides pharmacology, Virus Replication drug effects
- Abstract
We have reported previously that methylenecyclopropane analogs of nucleosides have excellent activity against certain members of the herpesvirus family. A second generation, the 2,2-bis-hydroxymethyl derivatives, were synthesized, and 18 compounds were tested for activity in vitro against herpes simplex virus types 1 and 2 (HSV-1 and HSV-2), human and murine cytomegalovirus (HCMV and MCMV), varicella-zoster virus (VZV), and Epstein-Barr virus (EBV). Selected analogs were also evaluated against human herpesvirus type 6 (HHV-6) and HHV-8. None of the 18 compounds had activity against HSV-1 or HSV-2, but four were active against VZV by plaque reduction (PR) assay at 50% effective concentration (EC(50)) levels of < or =50 microM. Six of the 18 compounds were active against HCMV by cytopathic effect or PR assays with EC(50)s of 0.5 to 44 microM, and all were active against MCMV by PR (0.3 to 54 microM). Four of the compounds were active against EBV by enzyme-linked immunosorbent assay (<0.3 to 4.4 microM). Four compounds with CMV activity were also active against HHV-6A and HHV-6B (0.7 to 28 microM), and three compounds were active against HHV-8 (5.5 to 16 microM). One of these, ZSM-I-62, had particularly good activity against CMV, HHV-6, and HHV-8, with EC(50)s of 0.7 to 8 microM. Toxicity was evaluated in adherent and nonadherent cells, and minimal cytotoxicity was observed. Mechanism of action studies with HCMV suggested that these compounds are phosphorylated by the ppUL97 phosphotransferase and are potent inhibitors of viral DNA synthesis. These results indicate that at least one of these compounds may have potential for use in treating CMV and other herpesvirus infections in humans.
- Published
- 2005
- Full Text
- View/download PDF
36. Human cytomegalovirus plasmid-based amplicon vector system for gene therapy.
- Author
-
Mahmood K, Prichard MN, Duke GM, Kemble GW, and Spaete RR
- Abstract
We have constructed and evaluated the utility of a helper-dependent virus vector system that is derived from Human Cytomegalovirus (HCMV). This vector is based on the herpes simplex virus (HSV) amplicon system and contains the HCMV orthologs of the two cis-acting functions required for replication and packaging of HSV genomes, the complex HCMV viral DNA replication origin (oriLyt), and the cleavage packaging signal (the a sequence). The HCMV amplicon vector replicated independently and was packaged into infectious virions in the presence of helper virus. This vector is capable of delivering and expressing foreign genes in infected cells including progenitor cells such as human CD34+ cells. Packaged defective viral genomes were passaged serially in fibroblasts and could be detected at passage 3; however, the copy number appeared to diminish upon serial passage. The HCMV amplicon offers an alternative vector strategy useful for gene(s) delivery to cells of the hematopoietic lineage.
- Published
- 2005
- Full Text
- View/download PDF
37. Requirement for uracil-DNA glycosylase during the transition to late-phase cytomegalovirus DNA replication.
- Author
-
Courcelle CT, Courcelle J, Prichard MN, and Mocarski ES
- Subjects
- Amino Acid Sequence, Cell Line, DNA Replication, DNA, Viral, Humans, Molecular Sequence Data, Sequence Alignment, Uracil-DNA Glycosidase, Cytomegalovirus physiology, DNA Glycosylases, N-Glycosyl Hydrolases physiology, Virus Replication physiology
- Abstract
Cytomegalovirus gene UL114, a homolog of mammalian uracil-DNA glycosylase (UNG), is required for efficient viral DNA replication. In quiescent fibroblasts, UNG mutant virus replication is delayed for 48 h and follows the virus-induced expression of cellular UNG. In contrast, mutant virus replication proceeds without delay in actively growing fibroblasts that express host cell UNG. In the absence of viral or host cell UNG expression, mutant virus fails to proceed to late-phase DNA replication, characterized by rapid DNA amplification. The data suggest that uracil incorporated early during wild-type viral DNA replication must be removed by virus or host UNG prior to late-phase amplification and encapsidation into progeny virions. The process of uracil incorporation and excision may introduce strand breaks to facilitate the transition from early-phase replication to late-phase amplification.
- Published
- 2001
- Full Text
- View/download PDF
38. Distinct and separate roles for herpesvirus-conserved UL97 kinase in cytomegalovirus DNA synthesis and encapsidation.
- Author
-
Wolf DG, Courcelle CT, Prichard MN, and Mocarski ES
- Subjects
- Capsid biosynthesis, Cells, Cultured, Cytomegalovirus genetics, Humans, Viral Proteins metabolism, Cytomegalovirus physiology, DNA Replication physiology, DNA, Viral biosynthesis, Phosphotransferases (Alcohol Group Acceptor) physiology, Virus Replication physiology
- Abstract
The human cytomegalovirus UL97 kinase, an important target of antiviral therapy, has an impact on at least two distinct phases of viral replication. Compared with wild-type virus, the UL97 deletion mutant exhibits an early replication defect that reduces DNA accumulation by 4- to 6-fold, as well as a late capsid maturation defect responsible for most of the observed 100- to 1000-fold reduction in replication. Block-release experiments with the antiviral 2-bromo-5,6-dichloro-1-(beta-D-ribofuranosyl)-benzimidazole revealed an important role for UL97 kinase in capsid assembly. Although cleavage of concatemeric DNA intermediates to unit-length genomes remained unaffected, progeny mutant virus maturation was delayed, with accumulation of progeny at significantly reduced levels compared with wild type after release of this block. Transmission electron microscopy confirmed the aberrant accumulation of empty A-like capsids containing neither viral DNA nor an internal scaffold structure, consistent with a failure to stably package DNA in mutant virus-infected cells. The function of UL97 in DNA synthesis as well as capsid assembly suggests that protein phosphorylation mediated by this herpesvirus-conserved kinase increases the efficiency of these two distinct phases of virus replication.
- Published
- 2001
- Full Text
- View/download PDF
39. A recombinant human cytomegalovirus with a large deletion in UL97 has a severe replication deficiency.
- Author
-
Prichard MN, Gao N, Jairath S, Mulamba G, Krosky P, Coen DM, Parker BO, and Pari GS
- Subjects
- Animals, Cell Line, Cells, Cultured, Cytomegalovirus genetics, DNA-Binding Proteins genetics, Gene Expression, Genetic Complementation Test, Humans, Mice, Open Reading Frames, Phosphotransferases (Alcohol Group Acceptor) genetics, Recombination, Genetic, Viral Proteins genetics, Cytomegalovirus physiology, Phosphotransferases (Alcohol Group Acceptor) physiology, Virus Replication
- Abstract
Human cytomegalovirus encodes a protein kinase (UL97) that confers sensitivity to ganciclovir by phosphorylating it to the monophosphate. The function of this unusual kinase in viral replication is unknown. We constructed two independent isolates of a recombinant virus, RCDelta97, that contain large deletions in this gene and carry a 4.8-kb insertion containing a selectable genetic marker. These mutant viruses were isolated by using a population of primary cells (HEL97) that express this gene from integrated copies of a defective retroviral vector. The recombinant viruses were severely impaired in their ability to replicate in primary fibroblasts, attaining virus titers that were 2 to 3 orders of magnitude lower than those produced by the parent virus. Despite the severe replication deficit, both of these viruses retained the ability to form small, slowly growing plaques in primary fibroblasts, demonstrating that UL97 is not absolutely essential for replication in cell culture. The replication deficit was relieved when UL97 was provided in trans in the complementing cell line, showing that the phenotype was due to a deficiency in UL97. Thus, the UL97 gene product plays a very important role in viral replication in tissue culture and may be a good target for antiviral chemotherapy.
- Published
- 1999
- Full Text
- View/download PDF
40. Identification of persistent RNA-DNA hybrid structures within the origin of replication of human cytomegalovirus.
- Author
-
Prichard MN, Jairath S, Penfold ME, St Jeor S, Bohlman MC, and Pari GS
- Subjects
- Binding Sites, Cell Line, Chromosome Mapping, Humans, Nucleic Acid Conformation, Restriction Mapping, Sodium Hydroxide, Cytomegalovirus genetics, DNA, Viral, RNA, Viral, Recombination, Genetic, Replication Origin
- Abstract
Human cytomegalovirus (HCMV) lytic-phase DNA replication initiates at the cis-acting origin of replication, oriLyt. oriLyt is a structurally complex region containing repeat elements and transcription factor binding sites. We identified two site-specific alkali-labile regions within oriLyt which flank an alkali-resistant DNA segment. These alkali-sensitive regions were the result of the degradation of two RNA species embedded within oriLyt and covalently linked to viral DNA. The virus-associated RNA, vRNA, was identified by DNase I treatment of HCMV DNA obtained from sucrose gradient purified virus. This heterogeneous population of vRNA was end labeled and used as a hybridization probe to map the exact location of vRNAs within oriLyt. vRNA-1 is localized between restriction endonuclease sites XhoI at nucleotide (nt) 93799 and SacI at nt 94631 and is approximately 500 bases long. The second vRNA, vRNA-2, lies within a region which exhibits a heterogeneous restriction pattern located between the SphI (nt 92636) and BamHI (nt 93513) and is approximately 300 bases long. This region was previously shown to be required for oriLyt replication (D. G. Anders, M. A. Kacica, G. S. Pari, and S. M. Punturieri, J. Virol. 66:3373-3384, 1992). RNase H analysis determined that vRNA-2 forms a persistent RNA-DNA hybrid structure in the context of the viral genome and in an oriLyt-containing plasmid used in the transient-replication assay.
- Published
- 1998
- Full Text
- View/download PDF
41. Reassessing the organization of the UL42-UL43 region of the human cytomegalovirus strain AD169 genome.
- Author
-
Mocarski ES, Prichard MN, Tan CS, and Brown JM
- Subjects
- Amino Acid Sequence, Humans, Molecular Sequence Data, Sequence Analysis, Cytomegalovirus genetics, Genome, Human, Polymorphism, Genetic
- Abstract
A polymorphism in the UL42-UL43 region of the human cytomegalovirus genome has been characterized by nucleotide sequence analysis, revealing a 929-bp insertion following nt 54,612 relative to the published strain AD169-UK genome sequence (M.S. Chee et al., 1990, Curr. Top. Microbiol Immunol. 154, 125-170). Although AD169-UK exhibited polymorphism in this genomic region, other CMV strains (Towne, Toledo, and AD169-ATCC) carried only the newly characterized longer form. The additional sequence altered the assignment of UL42 and UL43 open reading frames. UL42 decreased in size from 157 to 125 codons, retaining 76 of the previously reported carboxyl terminal codons, and UL43 increased in size from 187 to 423 codons, retaining 185 of the previously reported amino terminal codons. This additional sequence makes UL43 a more conserved betaherpesvirus US22 family member. Only AD169-UK exhibited restriction fragment length polymorphism in this region, suggesting that a deletion occurred during the propagation of this strain in cell culture. The additional sequence should be considered a bona fide part of the cytomegalovirus genome and the AD169 genome size should be corrected to 230,283 bp.
- Published
- 1997
- Full Text
- View/download PDF
42. Human cytomegalovirus uracil DNA glycosylase is required for the normal temporal regulation of both DNA synthesis and viral replication.
- Author
-
Prichard MN, Duke GM, and Mocarski ES
- Subjects
- Base Sequence, Bromodeoxyuridine toxicity, Cell Division, Cells, Cultured, Conserved Sequence, Cytomegalovirus enzymology, Cytomegalovirus genetics, Escherichia coli, Fibroblasts, Gene Expression Regulation, Viral, Genes, Viral, Herpesviridae genetics, Humans, Kinetics, Male, N-Glycosyl Hydrolases genetics, Open Reading Frames, Phenotype, Sequence Deletion, Skin, Time Factors, Uracil-DNA Glycosidase, Cytomegalovirus physiology, DNA Glycosylases, DNA Repair, DNA Replication drug effects, DNA, Viral biosynthesis, N-Glycosyl Hydrolases metabolism, Virus Replication drug effects
- Abstract
Human cytomegalovirus (CMV) encodes a gene, UL114, whose product is homologous to the uracil DNA glycosylase and is highly conserved in all herpesviruses. This DNA repair enzyme excises uracil residues in DNA that result from the misincorporation of dUTP or spontaneous deamination of cytosine. We constructed a recombinant virus, RC2620, that contains a large deletion in the UL114 open reading frame and carries a 1.2-kb insert containing the Escherichia coli gpt gene. RC2620 retains the capacity to replicate in primary human fibroblasts and reaches titers that are similar to those produced by the parent virus but exhibits a significantly longer replication cycle. Although the rate of expression of alpha and beta gene products appears to be unaffected by the mutation, DNA synthesis fails to proceed normally. Once initiated, DNA synthesis in mutant virus-infected cells proceeds at the same rate as with wild-type virus, but initiation is delayed by 48 h. The mutant virus also exhibits two predicted phenotypes: (i) hypersensitivity to the nucleoside analog 5-bromodeoxyuridine and (ii) retention of more uracil residues in genomic DNA than the parental virus. Together, these data suggest UL114 is required for the proper excision of uracil residues from viral DNA but in addition plays some role in establishing the correct temporal progression of DNA synthesis and viral replication. Although such involvement has not been previously observed in herpesviruses, a requirement for uracil DNA glycosylase in DNA replication has been observed in poxviruses.
- Published
- 1996
- Full Text
- View/download PDF
43. Strategic design and three-dimensional analysis of antiviral drug combinations.
- Author
-
Prichard MN, Prichard LE, and Shipman C Jr
- Subjects
- Acyclovir pharmacology, Animals, Cell Line, Chlorocebus aethiops, Drug Combinations, Drug Synergism, Floxuridine pharmacology, Kidney, Thiosemicarbazones pharmacology, Antiviral Agents pharmacology, Drug Design, Simplexvirus drug effects
- Abstract
The development of new drugs effective against human viral diseases has proven to be both difficult and time-consuming. Indeed, there are but 10 drugs licensed for such applications in the United States today. An attractive solution to this problem may be to optimize the efficacy and selectivity of existing antiviral drugs by combining them with agents that strategically block carefully selected metabolic pathways. This approach was used in the rational design of a three-drug combination to increase the apparent potency of acyclovir against herpes simplex virus. Recent advances in analytical techniques have made the evaluation of this complex drug strategy both possible and practical. A modified version of a previously described analytical method was used to identify optimal drug concentrations and to quantitate statistically significant synergy. Concentrations of 0.25 microM 5-fluorodeoxyuridine, 3.6 microM 2-acetylpyridine thiosemicarbazone, and 0.3 microM acyclovir were determined to be optimal in terms of antiviral activity. The volume of synergy produced was nearly 2,000 microM3% at a 95% level of confidence (corresponding to a 186-fold decrease in the apparent 50% inhibitory concentration of acyclovir with the addition of 0.25 microM 5-fluorodeoxyuridine and 3.6 microM 2-acetylpyridine thiosemicarbazone). We anticipate that this strategic approach and the supporting three-dimensional analytical method will prove valuable in designing and understanding multidrug therapies.
- Published
- 1993
- Full Text
- View/download PDF
44. Three-dimensional analysis of the synergistic cytotoxicity of ganciclovir and zidovudine.
- Author
-
Prichard MN, Prichard LE, Baguley WA, Nassiri MR, and Shipman C Jr
- Subjects
- Cell Division drug effects, Drug Evaluation, Preclinical, Drug Synergism, Humans, KB Cells drug effects, Tumor Cells, Cultured drug effects, Cell Survival drug effects, Ganciclovir pharmacology, Zidovudine pharmacology
- Abstract
The combined cytotoxicity of zidovudine and ganciclovir in three cell lines of human origin was examined. The data were generated by a new rapid cell proliferation assay and a more sensitive plating efficiency assay. A three-dimensional analytical approach was used to evaluate the drug-drug interactions, and the results were compared with those obtained by two conventional methods of analysis. Synergistic cytotoxicity was observed in all cell lines examined and by both assays. Moreover, this synergistic cytotoxicity was statistically significant at physiologically relevant concentrations. It is not known whether these drug-drug interactions manifest themselves in vivo as granulocytopenia or other untoward side effects. These results, however, indicate that further investigation is warranted and that the coadministration of zidovudine and ganciclovir may be contraindicated.
- Published
- 1991
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.