Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters








Publication year range
1.
J Virol ; 90(4): 1705-17, 2016 02 15.
Article in English | MEDLINE | ID: mdl-26608311

ABSTRACT

UNLABELLED: Human respiratory syncytial virus (RSV) is a single-stranded RNA virus that causes acute, and occasionally fatal, lower respiratory illness in young infants, the elderly, and immunocompromised patients. Therapeutic interventions able to cut short viral replication and quickly return the airways to normal function are needed. An understanding of antiviral activities and their effects on host defense mechanisms is important for the design of safe and effective therapy. We targeted functionally and temporally distinct steps within the viral life cycle using small-molecule RSV inhibitors and studied their antiviral activities and their effects on innate interferon responses of airway epithelial cells in vitro. Antivirals acting upstream of RSV polymerase activity (i.e., compounds targeting the fusion protein or the nucleoprotein) reduced viral load immediately postinfection and partially attenuated interferon responses. In contrast, antivirals directed to the RSV polymerase demonstrated activity throughout the viral replication cycle and specifically modulated the RIG-I/mitochondrial antiviral signaling protein (MAVS)/TBK1/IRF3/interferon-stimulated gene (ISG) axis, causing either an upregulation or a downregulation of interferon responses, depending on the mechanism of polymerase inhibition. Notably, polymerase inhibition leading to the accumulation of abortive RNA products correlated with the amplification of interferon-stimulated genes to up to 10 times above normal infection levels. Understanding how antiviral activities and their modulation of innate immunity may affect recovery from RSV infection will help guide the development of safe and effective therapies. IMPORTANCE: RSV circulates seasonally, causing acute lower respiratory disease. Therapeutic interventions with efficacy throughout the viral replication cycle, rapid viral clearance, and prevention of potentially harmful inflammatory responses are desirable. Compounds targeting the RSV polymerase inhibited virus replication late in the viral life cycle and, depending on the functional domain targeted, either attenuated or amplified RIG-I and downstream interferon pathways in infected cells. These data will help guide the development of safe and effective therapies by providing new molecular evidence that the mechanism of inhibition by an antiviral compound can directly impact innate antiviral immune responses in the airway epithelium.


Subject(s)
Antiviral Agents/metabolism , Epithelial Cells/immunology , Epithelial Cells/virology , Interferons/biosynthesis , Respiratory Syncytial Virus, Human/drug effects , Respiratory Syncytial Virus, Human/immunology , Cell Line , Humans
2.
J Viral Hepat ; 10(1): 50-4, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12558912

ABSTRACT

Adefovir is a potent nucleotide analog inhibitor of hepatitis B virus (HBV) DNA polymerase. Its oral prodrug adefovir dipivoxil has been approved for the treatment of chronic HBV infection. In this study, adefovir was characterized for its in vitro effects on mitochondrial DNA (mtDNA) synthesis and compared with the nucleoside analogues lamivudine (3TC), fialuridine (FIAU), and zalcitabine (ddC). No substantial changes in mtDNA content were detected in human hepatoblastoma HepG2 cells and normal human skeletal muscle cells following a 9-day treatment with 0.3-30 microm adefovir, concentrations up to 500-fold higher than the peak serum levels in patients treated with adefovir dipivoxil. Similarly, mtDNA was unchanged in both cell types following treatment with 3TC. In contrast, 30-55% and > 90% reductions in mtDNA were observed following incubation with 30 microm FIAU and ddC, respectively. The effects of FIAU on mtDNA became more pronounced following prolonged 18-day treatment of skeletal muscle cells while the effects of other drugs remained unchanged.


Subject(s)
Adenine/pharmacology , Antiviral Agents/pharmacology , DNA, Mitochondrial/drug effects , Hepatitis B virus/enzymology , Mitochondria, Liver/drug effects , Muscle, Skeletal/drug effects , Nucleic Acid Synthesis Inhibitors , Organophosphonates , Adenine/analogs & derivatives , Antiviral Agents/adverse effects , Antiviral Agents/therapeutic use , DNA, Mitochondrial/biosynthesis , Hepatitis B virus/drug effects , Hepatitis B, Chronic/drug therapy , Hepatoblastoma/drug therapy , Hepatoblastoma/metabolism , Humans , Muscle, Skeletal/metabolism , Tumor Cells, Cultured
3.
Article in English | MEDLINE | ID: mdl-11563082

ABSTRACT

hOAT1 is a renal membrane protein able to efficiently transport acyclic nucleoside phosphonates (ANPs). When expressed in CHO cells, hOAT1 mediates the uptake and cytotoxicity of ANPs suggesting that it plays an active role in the nephrotoxicity associated with cidofovir CMV therapy and high-dose adefovir HIV therapy. Although efficiently transported by hOAT1, tenofovir did not show any significant cytotoxicity in isolated human proximal tubular cells, which correlates with the lack of nephrotoxicity observed in HIV-infected patients on prolonged tenofovir therapy.


Subject(s)
Adenine/analogs & derivatives , Adenine/toxicity , Antiviral Agents/toxicity , Cytosine/analogs & derivatives , Cytosine/toxicity , Kidney Diseases/chemically induced , Organic Anion Transport Protein 1/metabolism , Organophosphonates , Organophosphorus Compounds/toxicity , Adenine/pharmacokinetics , Animals , Antiviral Agents/pharmacokinetics , Biological Transport , CHO Cells , Cidofovir , Cricetinae , Cytosine/pharmacokinetics , Humans , Kidney Diseases/metabolism , Kidney Tubules, Proximal/drug effects , Organic Anion Transport Protein 1/biosynthesis , Organic Anion Transport Protein 1/genetics , Organophosphorus Compounds/pharmacokinetics
4.
Clin Infect Dis ; 33(9): e105-8, 2001 Nov 01.
Article in English | MEDLINE | ID: mdl-11577375

ABSTRACT

We describe a case of CMV ventriculoencephalitis in a severely immunocompromised bone marrow transplant recipient who was receiving combination therapy with ganciclovir and foscarnet for treatment of viremia and retinitis. Analysis of sequential viral isolates recovered from the patient's cerebrospinal fluid suggested that disease developed because of the presence of viral resistance and, possibly, low tissue penetration of antiviral agents.


Subject(s)
Antiviral Agents/therapeutic use , Bone Marrow Transplantation/adverse effects , Cytomegalovirus Retinitis/virology , Cytosine/analogs & derivatives , Cytosine/therapeutic use , Drug Resistance, Viral , Encephalitis, Viral/virology , Foscarnet/therapeutic use , Ganciclovir/therapeutic use , Immunocompromised Host , Organophosphonates , Organophosphorus Compounds/therapeutic use , Cerebral Ventricles/virology , Child , Cidofovir , Cytomegalovirus Retinitis/drug therapy , Cytomegalovirus Retinitis/physiopathology , Drug Combinations , Drug Resistance, Viral/genetics , Encephalitis, Viral/drug therapy , Encephalitis, Viral/physiopathology , Female , Humans
5.
J Virol Methods ; 98(1): 53-61, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11543884

ABSTRACT

Human parainfluenza virus types 1 (hPIV-1), 2, and 3 represent significant respiratory pathogens for which no antiviral treatment is currently available. To characterize the biochemical functions of the hPIV-1 hemagglutinin-neuraminidase (HN) glycoprotein, a potential target for antiviral therapy, we cloned and expressed a soluble portion of hPIV-1 HN (amino acid residues 137-575), lacking the N-terminal hydrophobic membrane anchorage region, in insect cells using the baculovirus secretion expression system. The expressed HN protein was purified through cation-exchange chromatography followed by metal affinity chromatography, using the 6xHis epitope introduced at the carboxyl terminus of the recombinant protein. N-terminal amino acid sequence analysis of purified HN indicated that the honeybee melittin secretion signal peptide was correctly removed during post-translational processing. Further characterization revealed that the purified HN protein was N-glycosylated and exhibited neuraminidase activity whose characteristics resembled those of the native HN protein of hPIV-1 virions. The establishment of this expression and purification system has allowed us to further explore the biochemical characteristics of paramyxovirus HN and to obtain material that could be suitable for X-ray crystallography studies.


Subject(s)
HN Protein/biosynthesis , Parainfluenza Virus 1, Human/metabolism , Amino Acid Sequence , Animals , Baculoviridae/genetics , Cell Line , Genetic Vectors , HN Protein/chemistry , HN Protein/genetics , Humans , Hydrogen-Ion Concentration , Insecta , Molecular Sequence Data , Neuraminidase/metabolism , Nickel , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Sodium Chloride
6.
J Pharmacol Exp Ther ; 295(1): 10-5, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10991954

ABSTRACT

Adefovir is a nucleotide analog with anti-human immunodeficiency virus (HIV) activity that has been extensively studied in clinical trials. While on prolonged anti-HIV therapy with adefovir, some patients may develop drug-associated nephrotoxicity manifested by changes in laboratory markers of renal tubular functions that are reversible upon drug discontinuation. It has been recently shown that adefovir is efficiently transported by the human renal organic anion transporter 1 (hOAT1), a membrane transport protein localized in the kidney, that presumably mediates the accumulation of adefovir in renal proximal tubules. In an effort to look for novel inhibitors of this transport process, we used a cell line stably expressing hOAT1 to demonstrate that nonsteroidal anti-inflammatory drugs (NSAIDs) efficiently inhibit hOAT1-specific transport of adefovir at clinically relevant concentrations. Diflunisal, ketoprofen, flurbiprofen, indomethacin, naproxen, and ibuprofen were equally or more effective (IC(50) = 0.85-8 microM) than probenecid or betamipron, two known potent inhibitors of hOAT1 (IC(50) = 8 and 6 microM, respectively) with in vivo nephroprotective effects. Importantly, NSAIDs significantly reduced the shift in adefovir cytotoxicity observed upon hOAT1 expression with ketoprofen and naproxen being 2- to 3-times more effective than probenecid. Transport experiments with [(3)H]ketoprofen and [(3)H]ibuprofen revealed that NSAIDs themselves were not efficiently transported by hOAT1. None of the NSAIDs tested showed any interference with the anti-HIV activity of adefovir. In conclusion, these observations suggest that NSAIDs may reduce or delay the emergence of adefovir nephrotoxicity.


Subject(s)
Adenine/analogs & derivatives , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antiviral Agents/toxicity , Carrier Proteins/physiology , Kidney/drug effects , Organophosphonates , Adenine/pharmacokinetics , Adenine/toxicity , Anion Transport Proteins , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Biological Transport/drug effects , Carrier Proteins/antagonists & inhibitors , Humans , Structure-Activity Relationship
7.
Anal Biochem ; 283(1): 49-55, 2000 Jul 15.
Article in English | MEDLINE | ID: mdl-10929807

ABSTRACT

Secretion of small molecules from the systemic blood circulation into urine is one of the physiologically essential functions of the kidney. The human organic anion transporter (hOAT1) is a key component in the renal tubular secretion of negatively charged molecules including a variety of important therapeutics. In some cases, compounds interacting with hOAT1 may induce pharmacokinetic drug-drug interactions or cause nephrotoxicity. We developed a fluorescence-based, 96-well format assay using CHO cells stably expressing hOAT1, which allows for the evaluation of interactions between small molecules and hOAT1. The assay is based on the inhibition of the transport of 6-carboxyfluorescein, a high-affinity hOAT1 substrate (Km = 3.9 microM), which was identified as one of several fluorescent organic anions. The relative inhibition potency of various known hOAT1 substrates determined using the 6-carboxyfluorescein-based inhibition assay correlated well with their Km values, indicating that the fluorescent assay exhibits a proper specificity. This in vitro assay can be employed to evaluate the mechanism of renal clearance of organic anions, to assess potential drug-drug interactions and/or nephrotoxic effects of various therapeutics, and to screen for novel hOAT1 inhibitors that could serve as efficient nephroprotectants.


Subject(s)
Carrier Proteins/metabolism , Spectrometry, Fluorescence/methods , Animals , Anion Transport Proteins , Anions/metabolism , Arylsulfonates/chemistry , Boron Compounds/chemistry , CHO Cells , Cricetinae , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Fluorescein/chemistry , Fluoresceins/chemistry , Fluoresceins/metabolism , Fluorescent Dyes/chemistry , Glutarates/pharmacology , Humans , Inhibitory Concentration 50 , Kinetics , Models, Chemical , Time Factors , Transfection
8.
Mol Pharmacol ; 56(3): 570-80, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10462545

ABSTRACT

Nephrotoxicity is the dose-limiting clinical adverse effect of cidofovir and adefovir, two potent antiviral therapeutics. Because renal uptake likely plays a role in the etiology of cidofovir- and adefovir-associated nephrotoxicity, we attempted to identify a renal transporter capable of interacting with these therapeutics. A cDNA clone was isolated from a human renal library and designated human organic anion transporter 1 (hOAT1). Northern analysis detected a specific 2.5-kilobase pair hOAT1 transcript only in human kidney. However, reverse transcription-polymerase chain reaction revealed hOAT1 expression in human brain and skeletal muscle, as well. Immunoblot analysis of human kidney cortex demonstrated that hOAT1 is an 80- to 90-kilodalton heterogeneous protein modified by abundant N-glycosylation. Xenopus laevis oocytes expressing hOAT1 supported probenecid-sensitive uptake of [(3)H]p-aminohippurate (K(m) = 4 microM), which was trans-stimulated in oocytes preloaded with glutarate. Importantly, both hOAT1 and rat renal organic anion transporter 1 (rROAT1) mediated saturable, probenecid-sensitive uptake of cidofovir, adefovir, and other nucleoside phosphonate antivirals. The affinity of hOAT1 toward cidofovir and adefovir (K(m) = 46 and 30 microM, respectively) was 5- to 9-fold higher compared with rROAT1 (K(m) = 238 and 270 microM, respectively). These data indicate that hOAT1 may significantly contribute to the accumulation of cidofovir and adefovir in renal proximal tubules and, thus, play an active role in the mechanism of nephrotoxicity associated with these antiviral therapeutics.


Subject(s)
Adenine/analogs & derivatives , Antiviral Agents/metabolism , Carrier Proteins/genetics , Cytosine/analogs & derivatives , Kidney/drug effects , Organophosphonates , Organophosphorus Compounds/metabolism , Adenine/metabolism , Adenine/pharmacology , Amino Acid Sequence , Animals , Anion Transport Proteins , Antiviral Agents/pharmacology , Base Sequence , Biological Transport , Carrier Proteins/metabolism , Cidofovir , Cloning, Molecular , Cytosine/metabolism , Cytosine/pharmacology , DNA, Complementary/analysis , Humans , Immunoblotting , Kidney/metabolism , Molecular Sequence Data , Nucleosides/metabolism , Organophosphorus Compounds/pharmacology , Rats
9.
Mol Pharmacol ; 54(2): 291-7, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9687570

ABSTRACT

Adefovir dipivoxil [9-(2-(bispivaloyloxymethyl)phosphonylmethoxyethyl)adenine (bis-POM PMEA)], an oral prodrug of adefovir (PMEA), is currently in phase III clinical testing for the treatment of human immunodeficiency virus-1 (HIV-1) infection. Previous in vitro experiments have shown that HIV-1 recombinant viruses expressing either a K65R or a K70E mutation in reverse transcriptase (RT) have reduced sensitivity to PMEA and that the K70E mutant also has impaired replication capacity in vitro. Genotypic analyses of samples from patients enrolled in a phase I/II clinical trial of adefovir dipivoxil demonstrated that the K70E RT mutation developed in two of 29 patients during extended therapy. To further investigate the molecular mechanisms involved in the resistance to PMEA, we cloned, expressed, and purified HIV-1 RT enzymes carrying either the K65R or K70E and, for comparison, the M184V mutation. The Km values of dNTPs for these mutant enzymes were not significantly altered from wild-type RT. The Ki values for the K65R mutant were increased from wild-type by 2-5-fold against a variety of inhibitors, whereas the Ki values for the M184V mutant were increased 12-fold specifically for 2', 3'-dideoxy-3'-thiacytidine (3TC) triphosphate. The Ki values for the K70E mutant were increased for PMEA diphosphate and 3TC triphosphate by 2-3-fold. These results are in agreement with antiviral drug susceptibility assay results. The three recombinant enzymes were also evaluated for their specific activities and processivities. All mutants were reduced in specific activity with respect to wild-type RT. In single-cycle processivity studies, the M184V mutant was, as expected, notably impaired. The K70E mutant was also slightly impaired, whereas the K65R mutant was slightly more processive than wild-type. These results with recombinant K70E RT are consistent with the reduced in vitro replication capacity of the K70E RT mutant of HIV-1 and further demonstrate that the K70E mutation confers minor PMEA and 3TC resistance to HIV-1.


Subject(s)
HIV Reverse Transcriptase/genetics , HIV Reverse Transcriptase/metabolism , Mutation , Organophosphonates , Adenine/analogs & derivatives , Adenine/pharmacology , Anti-HIV Agents/pharmacology , Cloning, Molecular , Escherichia coli/genetics , HIV Reverse Transcriptase/drug effects , Poly A/metabolism , RNA/metabolism , Recombinant Proteins/drug effects , Recombinant Proteins/metabolism , Substrate Specificity , Templates, Genetic
10.
Virology ; 248(2): 382-93, 1998 Sep 01.
Article in English | MEDLINE | ID: mdl-9721246

ABSTRACT

In cell culture, cidofovir (CDV) was used to select a human cytomegalovirus (HCMV) strain with decreased drug susceptibility. The genotypic characterization of this virus revealed a single base substitution resulting in a K513N amino acid alteration in the viral DNA polymerase (UL54). Performed in parallel, the selection of HCMV for replication in the presence of ganciclovir (GCV) selected an M460V substitution in the phosphotransferase (UL97), as well as a K513N/V812L double substitution in DNA polymerase. Neither of the two DNA polymerase mutations has been previously identified in HCMV drug-resistant strains. To precisely elucidate their role in drug resistance, corresponding recombinant mutant viruses were generated by recombination of nine overlapping viral DNA fragments. The K513N recombinant virus showed 13- and 6.5-fold decreased susceptibility to CDV and GCV in vitro, respectively, compared with the wild-type recombinant virus. Mutation V812L was associated with a moderate (2-3-fold) decrease in susceptibility to CDV, GCV, foscarnet, and adefovir. A multiplicative interaction of the K513N and V812L mutations with regard to the profile and level of drug resistance was demonstrated in recombinant virus expressing both mutations. In vitro replication kinetic experiments revealed that the K513N mutation significantly decreased HCMV replication capacity. Consistent with this finding, the K513N mutant DNA polymerase exhibited reduced specific activity in comparison with the wild-type enzyme and was severely impaired in its 3'-5' exonuclease function. Unexpectedly, the K513N mutant enzyme showed no decrease in susceptibility to CDV-diphosphate or GCV-triphosphate. However, the K513N mutation decreased the susceptibility to CDV and GCV of the oriLyt plasmid replication in the transient transfection/infection assay, suggesting that the DNA replication of the K513N mutant virus is less sensitive to the corresponding inhibitors.


Subject(s)
Antiviral Agents/pharmacology , Cytomegalovirus/drug effects , Cytosine/analogs & derivatives , DNA-Directed DNA Polymerase/genetics , Organophosphonates , Organophosphorus Compounds/pharmacology , Point Mutation , Viral Proteins , Amino Acid Sequence , Cells, Cultured , Cidofovir , Cytomegalovirus/genetics , Cytomegalovirus/growth & development , Cytomegalovirus/physiology , Cytosine/pharmacology , DNA Replication/drug effects , DNA, Viral/biosynthesis , DNA, Viral/drug effects , DNA-Directed DNA Polymerase/physiology , Drug Resistance, Microbial/genetics , Enzyme Repression , Exonucleases/metabolism , Ganciclovir/pharmacology , Molecular Sequence Data , Phenotype , Phosphodiesterase I , Phosphoric Diester Hydrolases/metabolism , Sequence Homology, Amino Acid , Virus Replication/genetics
11.
J Virol ; 72(7): 5927-36, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9621055

ABSTRACT

A number of specific point mutations in the human cytomegalovirus (HCMV) DNA polymerase (UL54) gene have been tentatively associated with decreased susceptibility to antiviral agents and consequently with clinical failure. To precisely determine the roles of UL54 mutations in HCMV drug resistance, recombinant UL54 mutant viruses were generated by using cotransfection of nine overlapping HCMV DNA fragments into permissive fibroblasts, and their drug susceptibility profiles were determined. Amino acid substitutions located in UL54 conserved region IV (N408D, F412C, and F412V), region V (A987G), and delta-region C (L501I, K513E, P522S, and L545S) conferred various levels of resistance to cidofovir and ganciclovir. Mutations in region II (T700A and V715M) and region VI (V781I) were associated with resistance to foscarnet and adefovir. The region II mutations also conferred moderate resistance to lobucavir. In contrast to mutations in other UL54 conserved regions, those residing specifically in region III (L802M, K805Q, and T821I) were associated with various drug susceptibility profiles. Mutations located outside the known UL54 conserved regions (S676G and V759M) did not confer any significant changes in HCMV drug susceptibility. Predominantly an additive effect of multiple UL54 mutations with respect to the final drug resistance phenotype was demonstrated. Finally, the influence of selected UL54 mutations on the susceptibility of viral DNA replication to antiviral drugs was characterized by using a transient-transfection-plus-infection assay. Results of this work exemplify specific roles of the UL54 conserved regions in the development of HCMV drug resistance and may help guide optimization of HCMV therapy.


Subject(s)
Antiviral Agents/pharmacology , Cytomegalovirus/drug effects , DNA, Viral/genetics , DNA-Directed DNA Polymerase/physiology , Genes, Viral , Organophosphonates , Cidofovir , Cytomegalovirus/genetics , Cytosine/analogs & derivatives , Cytosine/pharmacology , DNA Replication , DNA-Directed DNA Polymerase/genetics , Drug Resistance, Microbial , Foscarnet/pharmacology , Ganciclovir/pharmacology , Humans , Mutation , Organophosphorus Compounds/pharmacology , Recombination, Genetic , Virus Replication
12.
Antimicrob Agents Chemother ; 41(3): 641-6, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9056007

ABSTRACT

Cidofovir (HPMPC) [1-[(S)-3-hydroxy-2-(phosphonomethoxy)propyl]-cytosine] is an acyclic nucleotide analog with potent and selective activity against herpesviruses. The prodrug, cyclic HPMPC (cHPMPC) [1-[((S)-2-hydroxy-2-oxo-1,4,2-dioxaphosphorinan-5-yl) methyl]cytosine], has antiviral activity similar to that of the parent compound but exhibits reduced toxicity in animal models. cHPMPC is converted to cidofovir by a cellular cyclic CMP phosphodiesterase (EC 3.1.4.37) which hydrolyzes a variety of substrates, including adenosine 3',5'-cyclic monophosphate (cAMP) and cytidine 3',5'-cyclic monophosphate (cCMP). The K(m) and Vmax values for hydrolysis of cHPMPC by cCMP phosphodiesterase purified from human liver are 250 microM and 0.66 nmol.min-1.unit-1, respectively. These values are similar to the K(m) and Vmax values for cAMP (23 microM and 1.16 nmol.min-1.unit-1, respectively) and cCMP (75 microM and 2.32 nmol.min-1.unit of enzyme-1, respectively). The catalytic efficiency (Vmax/K(m) ratio) of this enzyme for the cHPMPC substrate is only 10- to 20-fold lower than those for the natural cyclic nucleotides, indicating that cHPMPC is a viable intracellular substrate for the human enzyme. Kinetic analysis indicates that cHPMPC, cAMP, and cCMP are competitive with respect to each other and that they are hydrolyzed by the same enzyme. cHPMPC is hydrolyzed to cidofovir in all primary human cell systems tested, including those derived from target organs that might be infected in patients with human cytomegalovirus (HCMV) disease. Importantly, hydrolysis of cHPMPC is not diminished in cells infected with HCMV.


Subject(s)
Antiviral Agents/metabolism , Cytosine/analogs & derivatives , Organophosphonates , Organophosphorus Compounds/metabolism , Phosphoric Diester Hydrolases/metabolism , Prodrugs/metabolism , 2',3'-Cyclic Nucleotide 3'-Phosphodiesterase , Animals , Caco-2 Cells , Cell Line , Chromatography, Affinity , Chromatography, Gel , Chromatography, Ion Exchange , Cidofovir , Cytosine/metabolism , Cytosol/enzymology , Fibroblasts , Humans , Kinetics , Liver/enzymology , Phosphoric Diester Hydrolases/isolation & purification , Swine
13.
Protein Expr Purif ; 11(2): 209-18, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9367818

ABSTRACT

The catalytic subunit (UL54) and accessory protein (UL44) of human cytomegalovirus (HCMV) DNA polymerase have been cloned and expressed in an in vitro-coupled transcription/translation reticulocyte lysate system. The influence of the 5'-untranslated region (5'-UTR) on the efficiency of expression from the circular plasmids has been investigated. For expression of both UL54 and UL44, a truncated form of the alfalfa mosaic virus (AMV) RNA4 5'-UTR was found to be superior to the full-length AMV 5'-UTR or the original HCMV 5'-UTRs of different lengths. Protein products with Mr approximately 140 and 55 kDa were detected by sodium dodecyl sulfate-polyacrylamide gel electrophoresis analysis in the UL54 and UL44 in vitro expression reactions, respectively. The properties of the expressed enzyme were compared with those of native HCMV DNA polymerase purified from HCMV-infected cells. DNA polymerase and 3'-5' exonuclease activities of the expressed UL54/UL44 complex were found to be dependent on salt concentration in the same manner as the activities of the native enzyme. The in vitro-expressed enzyme resembles the purified HCMV DNA polymerase in its affinity for deoxynucleoside triphosphates as well as in its sensitivity to known inhibitors (cidofovir diphosphate, ganciclovir triphosphate, and foscarnet). This straightforward method for protein expression may also be applicable to other enzymes where reproducible generation of fully functional products is desirable.


Subject(s)
Cytomegalovirus/enzymology , DNA-Binding Proteins/biosynthesis , DNA-Directed DNA Polymerase/biosynthesis , Viral Proteins/biosynthesis , Alfalfa mosaic virus/genetics , Base Sequence , Cell-Free System , Cloning, Molecular , Cytomegalovirus/genetics , DNA-Binding Proteins/genetics , DNA-Directed DNA Polymerase/genetics , Molecular Sequence Data , Nucleic Acid Synthesis Inhibitors , Plasmids , Protein Biosynthesis , Recombinant Proteins/biosynthesis , Regulatory Sequences, Nucleic Acid , Reverse Transcriptase Inhibitors/pharmacology , Transcription, Genetic , Viral Proteins/genetics
14.
Mol Pharmacol ; 50(6): 1502-10, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8967971

ABSTRACT

Cidofovir [CDV; (S)-1-(3-hydroxy-2-phosphonomethoxyethyl)cytosine] is an acyclic nucleotide analog with potent and selective in vitro and in vivo activities against a broad spectrum of herpesviruses and other DNA viruses. We studied the mechanism of enzymatic synthesis of CDV diphosphate, the putative antiviral metabolite of CDV. The phosphorylation is two-step process catalyzed by several enzymes. An enzymatic activity phosphorylating CDV to its monophosphate derivative was purified from human liver and identified as pyrimidine nucleoside monophosphate kinase (EC 2.7.4.14.). CDV (Km = 2.10 +/- 0.18 mM and Vmax = 1.10 +/- 0.05 micromol/min/mg) was found to be a substantially weaker substrate for purified enzyme than CMP, UMP, or dCMP. Pyrimidine nucleoside monophosphate kinase was used for preparative enzymatic synthesis of CDV monophosphate. Pyruvate kinase (EC 2.7.1.40), creatine kinase (EC 2.7.3.2), and nucleoside diphosphate kinase (EC 2.7.4.6) were found to catalyze CDV diphosphate synthesis from CDV monophosphate, whereas phosphoglycerate kinase (EC 2.7.2.3) and succinyl-CoA synthetase (EC 6.2.1.4) did not. Based on Vmax/Km (phosphorylation efficiency) values determined with enzymes purified from human sources, the most efficient phosphorylation of CDV monophosphate is catalyzed by pyruvate kinase. After infection of human lung fibroblasts with cytomegalovirus, the intracellular activities of pyrimidine nucleoside monophosphate kinase, pyruvate kinase, creatine kinase, and nucleoside diphosphate kinase increased 2-, 1.3-, 3-, and 5-fold, respectively. The metabolism of [3H]CDV in mock- and cytomegalovirus-infected cells was examined. The intracellular levels of CDV monophosphate and CDV diphosphate increased approximately 20- and 8-fold, respectively, in cytomegalovirus-infected cells, presumably due to the stimulation of CDV uptake and higher activities of phosphorylating enzymes.


Subject(s)
Antiviral Agents/metabolism , Cytomegalovirus Infections/metabolism , Cytosine/analogs & derivatives , Organophosphonates , Organophosphorus Compounds/metabolism , Phosphotransferases/metabolism , Catalysis , Cell Line , Cidofovir , Cytomegalovirus Infections/enzymology , Cytosine/metabolism , Humans , Kinetics , Phosphorylation , Phosphotransferases/isolation & purification
15.
Antimicrob Agents Chemother ; 39(1): 117-24, 1995 Jan.
Article in English | MEDLINE | ID: mdl-7695292

ABSTRACT

9-(2-Phosphonomethoxyethyl)adenine (PMEA) is an acyclic adenine nucleotide analog which exhibits potent and selective antiviral activity against herpesviruses and retroviruses. The study of [14C]PMEA uptake in HeLa S3 cells has shown that intracellular levels of the drug plateau after 1 h. Transport across the plasma membrane is saturable (concentration at half-maximal saturation [Kt], 0.39 microM; maximum rate of uptake [Vmax], 1.72 pmol/min.10(6) cells), and it can operate against the concentration gradient. Its significant dependence on temperature and on cellular density has been demonstrated. Following the treatment of cells with proteases, PMEA uptake strongly decreases. The transport process is considerably specific, since only a few phosphonate analogs act effectively as competitive inhibitors. Of these, 9-(2-phosphonomethoxyethyl)-2,6-diaminopurine (Ki = 0.24 microM) is the most efficient. Also, natural nucleotides competitively inhibit PMEA transport, depending on the nature of the nucleobase (thymine = adenine > guanine > cytosine < uracil) and on the position and number of phosphate groups. Nucleosides and nucleobases do not interfere with PMEA uptake. Cellular transport of adenosine and thymidine or uptake of AMP and ATP via conjugated activity of ectonucleotidases and nucleoside transporters is not affected by PMEA. By using vectorial labeling of plasma membrane proteins with Na125I combined with affinity chromatography, a 50-kDa protein which may mediate cellular transport of PMEA has been identified.


Subject(s)
Adenine/analogs & derivatives , Antiviral Agents/pharmacokinetics , Cell Membrane/metabolism , Organophosphonates , Adenine/pharmacokinetics , Biological Transport/drug effects , Cell Membrane/drug effects , Cells, Cultured , HeLa Cells/metabolism , Humans , Structure-Activity Relationship
16.
Biochem Pharmacol ; 44(10): 2067-77, 1992 Nov 17.
Article in English | MEDLINE | ID: mdl-1449524

ABSTRACT

Acyclic nucleotide analogues 9-(2-phosphonomethoxyethyl)adenine (PMEA) and 9-(S)-(3-hydroxy-2-phosphonomethoxypropyl)adenine ((S)-HPMPA) which display potent antiviral activity are transformed in the cells to their mono- and disphosphoryl derivatives. We purified from mouse L1210 cells the enzyme that in two steps phosphorylates PMEA and (S)-HPMPA to their diphosphoryl derivatives and found that it co-purifies with AMP(dAMP) kinase activity; the best substrates of this enzyme were AMP, ADP and dAMP. Other nucleoside 5'-triphosphates or creatine phosphate could not be substituted for ATP as a phosphate donor. Our results also indicated that at least one other enzyme (creatine kinase) is capable of transforming the monophosphoryl derivatives of the studied compounds to their respective diphosphates.


Subject(s)
Adenine/analogs & derivatives , Adenylate Kinase/metabolism , Leukemia L1210/enzymology , Organophosphonates , Organophosphorus Compounds/metabolism , Adenine/metabolism , Adenylate Kinase/chemistry , Adenylate Kinase/isolation & purification , Animals , Creatine Kinase/metabolism , Isoelectric Point , Mice , Molecular Weight , Substrate Specificity , Time Factors , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL