Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Cell ; 81(21): 4467-4480.e7, 2021 11 04.
Article in English | MEDLINE | ID: mdl-34687604

ABSTRACT

Viral RNA-dependent RNA polymerases (RdRps) are a target for broad-spectrum antiviral therapeutic agents. Recently, we demonstrated that incorporation of the T-1106 triphosphate, a pyrazine-carboxamide ribonucleotide, into nascent RNA increases pausing and backtracking by the poliovirus RdRp. Here, by monitoring enterovirus A-71 RdRp dynamics during RNA synthesis using magnetic tweezers, we identify the "backtracked" state as an intermediate used by the RdRp for copy-back RNA synthesis and homologous recombination. Cell-based assays and RNA sequencing (RNA-seq) experiments further demonstrate that the pyrazine-carboxamide ribonucleotide stimulates these processes during infection. These results suggest that pyrazine-carboxamide ribonucleotides do not induce lethal mutagenesis or chain termination but function by promoting template switching and formation of defective viral genomes. We conclude that RdRp-catalyzed intra- and intermolecular template switching can be induced by pyrazine-carboxamide ribonucleotides, defining an additional mechanistic class of antiviral ribonucleotides with potential for broad-spectrum activity.


Subject(s)
Pyrazines/chemistry , RNA Viruses/genetics , RNA, Viral/genetics , RNA-Dependent RNA Polymerase/genetics , Recombination, Genetic , Ribonucleotides/chemistry , Animals , Antiviral Agents , Catalysis , Cells, Cultured , Genetic Techniques , Genome , Genome, Viral , Homologous Recombination , Humans , Kinetics , Mice , Mice, Transgenic , Molecular Dynamics Simulation , Mutagenesis , Nucleotides/genetics , Protein Conformation , RNA/chemistry , RNA-Dependent RNA Polymerase/metabolism , RNA-Seq , Transgenes , Virulence
2.
Cell Rep ; 21(4): 1063-1076, 2017 Oct 24.
Article in English | MEDLINE | ID: mdl-29069588

ABSTRACT

RNA viruses pose a threat to public health that is exacerbated by the dearth of antiviral therapeutics. The RNA-dependent RNA polymerase (RdRp) holds promise as a broad-spectrum, therapeutic target because of the conserved nature of the nucleotide-substrate-binding and catalytic sites. Conventional, quantitative, kinetic analysis of antiviral ribonucleotides monitors one or a few incorporation events. Here, we use a high-throughput magnetic tweezers platform to monitor the elongation dynamics of a prototypical RdRp over thousands of nucleotide-addition cycles in the absence and presence of a suite of nucleotide analog inhibitors. We observe multiple RdRp-RNA elongation complexes; only a subset of which are competent for analog utilization. Incorporation of a pyrazine-carboxamide nucleotide analog, T-1106, leads to RdRp backtracking. This analysis reveals a mechanism of action for this antiviral ribonucleotide that is corroborated by cellular studies. We propose that induced backtracking represents a distinct mechanistic class of antiviral ribonucleotides.


Subject(s)
Antiviral Agents/pharmacology , Nucleosides/pharmacology , Pyrazines/pharmacology , RNA-Dependent RNA Polymerase/metabolism , Viral Proteins/metabolism , Antiviral Agents/chemistry , HeLa Cells , Humans , Imaging, Three-Dimensional , Magnetic Fields , Nucleosides/chemistry , Pyrazines/chemistry , RNA-Dependent RNA Polymerase/antagonists & inhibitors , RNA-Dependent RNA Polymerase/chemistry , Ribonucleotides/chemistry , Ribonucleotides/metabolism , Viral Proteins/antagonists & inhibitors , Viral Proteins/chemistry
3.
J Mol Biol ; 429(1): 88-96, 2017 01 06.
Article in English | MEDLINE | ID: mdl-27887868

ABSTRACT

The human APOBEC3G (A3G) enzyme restricts HIV-1 in the absence of the viral accessory protein viral infectivity factor (Vif) by deaminating viral cDNA cytosines to uracils. These uracil lesions base-pair with adenines during the completion of reverse transcription and result in A3G signature G-to-A mutations in the viral genome. Vif protects HIV-1 from A3G-mediated restriction by forming an E3-ubiquitin ligase complex to polyubiquitinate A3G and trigger its degradation. Prior studies indicated that Vif may also directly block the enzymatic activity of A3G and, provocatively, that Vif-derived peptides, Vif 25-39 and Vif 105-119, are similarly inhibitory. Here, we show that Vif 25-39 does not inhibit A3G enzymatic activity and that the inhibitory effect of Vif 105-119 and that of a shorter derivative Vif 107-115, although recapitulated, are non-specific. We also elaborate a simple method for assaying DNA cytosine deaminase activity that eliminates potential polymerase chain reaction-induced biases. Our results show that these Vif-derived peptides are unlikely to be useful as tools to study A3G function or as leads for the development of future therapeutics.


Subject(s)
APOBEC-3G Deaminase/antagonists & inhibitors , Enzyme Inhibitors/metabolism , Peptides/metabolism , vif Gene Products, Human Immunodeficiency Virus/metabolism , Peptides/genetics , vif Gene Products, Human Immunodeficiency Virus/genetics
4.
Org Biomol Chem ; 14(17): 4103-9, 2016 Apr 26.
Article in English | MEDLINE | ID: mdl-27113574

ABSTRACT

The topoisomerase (topo) I-DNA covalent complex represents an attractive target for developing diagnostic antibodies to measure responsiveness to drugs. We report a new antigen, peptide , and four murine monoclonal antibodies raised against that exhibit excellent specificity for recognition of in comparison to structurally similar peptides by enzyme-linked immunosorbent assays. Although topo I-DNA complex detection was not achieved in cellular samples by these new antibodies, a new strategy for antigen design is reported.


Subject(s)
Antibodies, Monoclonal/chemistry , Antigens/chemistry , DNA Topoisomerases, Type I/chemistry , DNA/chemistry , Nucleotides/chemistry , Peptides/chemistry , Animals , Antibodies, Monoclonal/immunology , Antibody Specificity , Antigen-Antibody Reactions , Antigens/immunology , Cell Line, Tumor , DNA Topoisomerases, Type I/metabolism , Enzyme-Linked Immunosorbent Assay , Humans , Mice , Molecular Structure , Nucleotides/chemical synthesis , Peptides/chemical synthesis
5.
Nucleic Acids Res ; 44(6): 2816-26, 2016 Apr 07.
Article in English | MEDLINE | ID: mdl-26917015

ABSTRACT

A number of established and investigational anticancer drugs slow the religation step of DNA topoisomerase I (topo I). These agents induce cytotoxicity by stabilizing topo I-DNA covalent complexes, which in turn interact with advancing replication forks or transcription complexes to generate lethal lesions. Despite the importance of topo I-DNA covalent complexes, it has been difficult to detect these lesions within intact cells and tumors. Here, we report development of a monoclonal antibody that specifically recognizes covalent topo I-DNA complexes, but not free topo I or DNA, by immunoblotting, immunofluorescence or flow cytometry. Utilizing this antibody, we demonstrate readily detectable topo I-DNA covalent complexes after treatment with camptothecins, indenoisoquinolines and cisplatin but not nucleoside analogues. Topotecan-induced topo I-DNA complexes peak at 15-30 min after drug addition and then decrease, whereas indotecan-induced complexes persist for at least 4 h. Interestingly, simultaneous staining for covalent topo I-DNA complexes, phospho-H2AX and Rad51 suggests that topotecan-induced DNA double-strand breaks occur at sites distinct from stabilized topo I-DNA covalent complexes. These studies not only provide new insight into the action of topo I-directed agents, but also illustrate a strategy that can be applied to study additional topoisomerases and their inhibitors in vitro and in vivo.


Subject(s)
Antibodies, Monoclonal/biosynthesis , Antineoplastic Agents, Phytogenic/pharmacology , DNA Topoisomerases, Type I/genetics , DNA/genetics , Gene Expression Regulation, Neoplastic , Topoisomerase I Inhibitors/pharmacology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/isolation & purification , Apoptosis/drug effects , Benzodioxoles/pharmacology , Cell Line, Tumor , Cisplatin/pharmacology , DNA/metabolism , DNA Breaks, Double-Stranded , DNA Topoisomerases, Type I/metabolism , HCT116 Cells , Histones/genetics , Histones/metabolism , Humans , Isoquinolines/pharmacology , K562 Cells , Mice , Molecular Sequence Data , Protein Binding/drug effects , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism , Sequence Alignment , Structure-Activity Relationship , Topotecan/pharmacology
6.
J Med Chem ; 58(18): 7419-30, 2015 Sep 24.
Article in English | MEDLINE | ID: mdl-26358009

ABSTRACT

High-throughput screening (HTS) was employed to discover APOBEC3G inhibitors, and multiple 2-furylquinolines (e.g., 1) were found. Dose-response assays with 1 from the HTS sample, as well as commercial material, yielded similar confirmatory results. Interestingly, freshly synthesized and DMSO-solubilized 1 was inactive. Repeated screening of the DMSO aliquot of synthesized 1 revealed increasing APOBEC3G inhibitory activity with age, suggesting that 1 decomposes into an active inhibitor. Laboratory aging of 1 followed by analysis revealed that 1 undergoes oxidative decomposition in air, resulting from a [4 + 2] cycloaddition between the furan of 1 and (1)O2. The resulting endoperoxide then undergoes additional transformations, highlighted by Baeyer-Villager rearrangements, to deliver lactam, carboxylic acid, and aldehyde products. The endoperoxide also undergoes hydrolytic opening followed by further transformations to a bis-enone. Eight structurally related analogues from HTS libraries were similarly reactive. This study constitutes a cautionary tale to validate 2-furylquinolines for structure and stability prior to chemical optimization campaigns.


Subject(s)
Cytidine Deaminase/antagonists & inhibitors , Furans/chemistry , Quinolines/chemistry , Thiadiazoles/chemistry , APOBEC-3G Deaminase , Dimethyl Sulfoxide , Dose-Response Relationship, Drug , Drug Stability , HEK293 Cells , HeLa Cells , High-Throughput Screening Assays , Humans , Oxidation-Reduction , Quinolines/pharmacology , Small Molecule Libraries , Solutions , Structure-Activity Relationship , Thiadiazoles/pharmacology
7.
Proc Natl Acad Sci U S A ; 109(11): 4092-7, 2012 Mar 13.
Article in English | MEDLINE | ID: mdl-22388745

ABSTRACT

Protein-protein interactions mediated by ubiquitin-like (Ubl) modifications occur as mono-Ubl or poly-Ubl chains. Proteins that regulate poly-SUMO (small ubiquitin-like modifier) chain conjugates play important roles in cellular response to DNA damage, such as those caused by cancer radiation therapy. Additionally, high atomic number metals, such as gold, preferentially absorb much more X-ray energy than soft tissues, and thus augment the effect of ionizing radiation when delivered to cells. In this study, we demonstrate that conjugation of a weak SUMO-2/3 ligand to gold nanoparticles facilitated selective multivalent interactions with poly-SUMO-2/3 chains leading to efficient inhibition of poly-SUMO-chain-mediated protein-protein interactions. The ligand-gold particle conjugate significantly sensitized cancer cells to radiation but was not toxic to normal cells. This study demonstrates a viable approach for selective targeting of poly-Ubl chains through multivalent interactions created by nanoparticles that can be chosen based on their properties, such as abilities to augment radiation effects.


Subject(s)
Gold/pharmacology , Metal Nanoparticles/chemistry , Radiation, Ionizing , Small Ubiquitin-Related Modifier Proteins/antagonists & inhibitors , Amino Acid Motifs , Biological Transport/drug effects , Biological Transport/radiation effects , Biotinylation/drug effects , Biotinylation/radiation effects , Cell Death/drug effects , Cell Death/radiation effects , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Comet Assay , DNA Damage , HeLa Cells , Humans , Ligands , Peptide Library , Peptides/chemistry , Protein Binding/drug effects , Protein Binding/radiation effects , Protein Interaction Maps , Small Ubiquitin-Related Modifier Proteins/metabolism , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism , Subcellular Fractions/radiation effects
8.
ACS Chem Biol ; 7(3): 506-17, 2012 Mar 16.
Article in English | MEDLINE | ID: mdl-22181350

ABSTRACT

APOBEC3G is a single-stranded DNA cytosine deaminase that comprises part of the innate immune response to viruses and transposons. Although APOBEC3G is the prototype for understanding the larger mammalian polynucleotide deaminase family, no specific chemical inhibitors exist to modulate its activity. High-throughput screening identified 34 compounds that inhibit APOBEC3G catalytic activity. Twenty of 34 small molecules contained catechol moieties, which are known to be sulfhydryl reactive following oxidation to the orthoquinone. Located proximal to the active site, C321 was identified as the binding site for the inhibitors by a combination of mutational screening, structural analysis, and mass spectrometry. Bulkier substitutions C321-to-L, F, Y, or W mimicked chemical inhibition. A strong specificity for APOBEC3G was evident, as most compounds failed to inhibit the related APOBEC3A enzyme or the unrelated enzymes E. coli uracil DNA glycosylase, HIV-1 RNase H, or HIV-1 integrase. Partial, but not complete, sensitivity could be conferred to APOBEC3A by introducing the entire C321 loop from APOBEC3G. Thus, a structural model is presented in which the mechanism of inhibition is both specific and competitive, by binding a pocket adjacent to the APOBEC3G active site, reacting with C321, and blocking access to substrate DNA cytosines.


Subject(s)
Cytidine Deaminase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Small Molecule Libraries/pharmacology , APOBEC-3G Deaminase , Cells, Cultured , Crystallography, X-Ray , Cytidine Deaminase/isolation & purification , Cytidine Deaminase/metabolism , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Enzyme Inhibitors/chemistry , HEK293 Cells , HIV Integrase/metabolism , Humans , Models, Molecular , Molecular Structure , Ribonuclease H/antagonists & inhibitors , Ribonuclease H/metabolism , Small Molecule Libraries/chemistry , Structure-Activity Relationship
9.
J Org Chem ; 69(23): 7928-32, 2004 Nov 12.
Article in English | MEDLINE | ID: mdl-15527272

ABSTRACT

The tricyclic core of halichlorine has been synthesized through the use of an alkynyliodonium salt/alkylidenecarbene/1,5 C-H insertion sequence that sets the pivotal quaternary center in the target.


Subject(s)
Alkaloids/chemical synthesis , Spiro Compounds/chemical synthesis , Molecular Structure , Nuclear Magnetic Resonance, Biomolecular , Onium Compounds/chemistry , Salts/chemistry , Stereoisomerism
SELECTION OF CITATIONS
SEARCH DETAIL
...