Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Nucleic Acids Res ; 41(12): 6139-48, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23620282

ABSTRACT

Apolipoprotein B mRNA-editing, enzyme-catalytic, polypeptide-like 3G (i.e., APOBEC3G or A3G) is an evolutionarily conserved cytosine deaminase that potently restricts human immunodeficiency virus type 1 (HIV-1), retrotransposons and other viruses. A3G has a nucleotide target site specificity for cytosine dinucleotides, though only certain cytosine dinucleotides are 'hotspots' for cytosine deamination, and others experience little or no editing by A3G. The factors that define these critical A3G hotspots are not fully understood. To investigate how A3G hotspots are defined, we used an in vitro fluorescence resonance energy transfer-based oligonucleotide assay to probe the site specificity of A3G. Our findings strongly suggest that the target single-stranded DNA (ssDNA) secondary structure as well as the bases directly 3' and 5' of the cytosine dinucleotide are critically important A3G recognition. For instance, A3G cannot readily deaminate a cytosine dinucleotide in ssDNA stem structures or in nucleotide base loops composed of three bases. Single-stranded nucleotide loops up to seven bases in length were poor targets for A3G activity unless cytosine residues flanked the cytosine dinucleotide. Furthermore, we observed that A3G favors adenines, cytosines and thymines flanking the cytosine dinucleotide target in unstructured regions of ssDNA. Low cytosine deaminase activity was detected when guanines flanked the cytosine dinucleotide. Taken together, our findings provide the first demonstration that A3G cytosine deamination hotspots are defined by both the sequence context of the cytosine dinucleotide target as well as the ssDNA secondary structure. This knowledge can be used to better trace the origins of mutations to A3G activity, and illuminate its impact on processes such as HIV-1 genetic variation.


Subject(s)
Cytidine Deaminase/metabolism , Cytosine/metabolism , DNA, Single-Stranded/chemistry , Base Sequence , Cell Line , Deamination , Humans , Nucleic Acid Conformation
2.
J Virol ; 87(9): 5296-9, 2013 May.
Article in English | MEDLINE | ID: mdl-23449788

ABSTRACT

The high rates of mutation, recombination, and replication drive HIV-1 diversity. In this study, we investigated how cell type affects viral mutation rate and mutation spectra. In studying four different cell types, no differences in mutation rate were observed, but intriguingly cell type differences impacted HIV-1 mutation spectra. This is the first description of significant differences in HIV-1 mutation spectra observed in different cell types in the absence of changes in the viral mutation rate.


Subject(s)
HIV Infections/virology , HIV-1/genetics , Mutation Rate , Base Sequence , Cell Line , Genetic Variation , HIV-1/classification , HIV-1/isolation & purification , Humans , Molecular Sequence Data , Species Specificity
3.
J Mol Biol ; 419(3-4): 158-70, 2012 Jun 08.
Article in English | MEDLINE | ID: mdl-22426127

ABSTRACT

RNA virus population dynamics are complex, and sophisticated approaches are needed in many cases for therapeutic intervention. One such approach, termed lethal mutagenesis, is directed at targeting the virus population structure for extinction or error catastrophe. Previous studies have demonstrated the concept of this approach with human immunodeficiency virus type 1 (HIV-1) by use of chemical mutagens [i.e., 5-azacytidine (5-AZC)] as well as by host factors with mutagenic properties (i.e., APOBEC3G). In this study, these two unrelated mutagenic agents were used concomitantly to investigate the interplay of these distinct mutagenic mechanisms. Specifically, an HIV-1 was produced from APOBEC3G (A3G)-expressing cells and used to infect permissive target cells treated with 5-AZC. Reduced viral infectivity and increased viral mutagenesis were observed with both the viral mutagen (i.e., G-to-C mutations) and the host restriction factor (i.e., G-to-A mutations); however, when combined, they had complex interactions. Intriguingly, nucleotide sequence analysis revealed that concomitant HIV-1 exposure to both 5-AZC and A3G resulted in an increase in G-to-A viral mutagenesis at the expense of G-to-C mutagenesis. A3G catalytic activity was required for the diminution in G-to-C mutagenesis. Taken together, our findings provide the first demonstration for potentiation of the mutagenic effect of a cytosine analog by A3G expression, resulting in concomitant HIV-1 lethal mutagenesis.


Subject(s)
Azacitidine/pharmacology , Cytidine Deaminase/metabolism , HIV-1/genetics , Mutagenesis , Mutagens/pharmacology , RNA, Viral/genetics , APOBEC-3G Deaminase , Cell Line , Cytidine Deaminase/genetics , HIV-1/metabolism , HIV-1/physiology , Humans , Sequence Analysis, RNA , Virus Replication/genetics , vif Gene Products, Human Immunodeficiency Virus/genetics
4.
Antimicrob Agents Chemother ; 56(4): 1942-8, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22271861

ABSTRACT

The emergence of drug resistance threatens to limit the use of current anti-HIV-1 drugs and highlights the need to expand the number of treatment options available for HIV-1-infected individuals. Our previous studies demonstrated that two clinically approved drugs, decitabine and gemcitabine, potently inhibited HIV-1 replication in cell culture through a mechanism that is distinct from the mechanisms for the drugs currently used to treat HIV-1 infection. We further demonstrated that gemcitabine inhibited replication of a related retrovirus, murine leukemia virus (MuLV), in vivo using the MuLV-based LP-BM5/murine AIDS (MAIDS) mouse model at doses that were not toxic. Since decitabine and gemcitabine inhibited MuLV and HIV-1 replication with similar potency in cell culture, the current study examined the efficacy and toxicity of the drug combination using the MAIDS model. The data demonstrate that the drug combination inhibited disease progression, as detected by histopathology, viral loads, and spleen weights, at doses lower than those that would be required if the drugs were used individually. The combination of decitabine and gemcitabine exerted antiviral activity at doses that were not toxic. These findings indicate that the combination of decitabine and gemcitabine shows potent antiretroviral activity at nontoxic doses and should be further investigated for clinical relevance.


Subject(s)
Anti-HIV Agents/therapeutic use , Azacitidine/analogs & derivatives , Deoxycytidine/analogs & derivatives , HIV-1/drug effects , Murine Acquired Immunodeficiency Syndrome/drug therapy , Animals , Anti-HIV Agents/adverse effects , Azacitidine/therapeutic use , Body Weight/drug effects , Cells, Cultured , Chemical and Drug Induced Liver Injury/pathology , Decitabine , Deoxycytidine/adverse effects , Deoxycytidine/therapeutic use , Drug Combinations , Drug Synergism , Female , Flow Cytometry , Humans , Liver/pathology , Lymph Nodes/pathology , Lymph Nodes/virology , Mice , Mice, Inbred C57BL , Murine Acquired Immunodeficiency Syndrome/pathology , Murine Acquired Immunodeficiency Syndrome/virology , Proviruses/drug effects , Spleen/pathology , Spleen/virology , T-Lymphocytes/drug effects , Transfection , Gemcitabine
5.
PLoS One ; 6(1): e15840, 2011 Jan 14.
Article in English | MEDLINE | ID: mdl-21264291

ABSTRACT

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


Subject(s)
Anti-Retroviral Agents/pharmacology , Deoxycytidine/analogs & derivatives , Leukemia Virus, Murine/drug effects , Murine Acquired Immunodeficiency Syndrome/drug therapy , Animals , Cells, Cultured , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Disease Progression , Immunoglobulin M/blood , Mice , Mice, Inbred C57BL , Ribonucleotide Reductases/antagonists & inhibitors , Spleen/pathology , Viral Load/drug effects , Gemcitabine
SELECTION OF CITATIONS
SEARCH DETAIL
...