Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 105
Filter
Add more filters

Publication year range
1.
J Infect Dis ; 228(10): 1421-1429, 2023 11 11.
Article in English | MEDLINE | ID: mdl-37224627

ABSTRACT

BACKGROUND: On May 6, 2022, a powerful outbreak of monkeypox virus (MPXV) had been reported outside of Africa, with many continuing new cases being reported around the world. Analysis of mutations among the 2 different lineages present in the 2021 and 2022 outbreaks revealed the presence of G->A mutations occurring in the 5'GpA context, indicative of APOBEC3 cytidine deaminase activity. METHODS: By using a sensitive polymerase chain reaction (differential DNA denaturation PCR) method allowing differential amplification of AT-rich DNA, we analyzed the level of APOBEC3-induced MPXV editing in infected cells and in patients. RESULTS: We demonstrate that G->A hypermutated MPXV genomes can be recovered experimentally from APOBEC3 transfection followed by MPXV infection. Here, among the 7 human APOBEC3 cytidine deaminases (A3A-A3C, A3DE, A3F-A3H), only APOBEC3F was capable of extensively deaminating cytidine residues in MPXV genomes. Hyperedited genomes were also recovered in ∼42% of analyzed patients. Moreover, we demonstrate that substantial repair of these mutations occurs. Upon selection, corrected G->A mutations escaping drift loss contribute to the MPXV evolution observed in the current epidemic. CONCLUSIONS: Stochastic or transient overexpression of the APOBEC3F gene exposes the MPXV genome to a broad spectrum of mutations that may be modeling the mutational landscape after multiple cycles of viral replication.


Subject(s)
Cytidine Deaminase , Monkeypox virus , Humans , Monkeypox virus/genetics , Cytidine Deaminase/genetics , Mutation , Disease Outbreaks , Cytidine , Cytosine Deaminase/chemistry , Cytosine Deaminase/genetics
2.
J Virol ; 95(12)2021 05 24.
Article in English | MEDLINE | ID: mdl-33789992

ABSTRACT

APOBEC3 proteins play pivotal roles in defenses against retroviruses, including HIV-1, as well as retrotransposons. Presumably due to the evolutionary arms race between the hosts and retroelements, APOBEC3 genes have rapidly evolved in primate lineages through sequence diversification, gene amplification and loss, and gene fusion. Consequently, modern primates possess a unique set or "repertoire" of APOBEC3 genes. The APOBEC3 gene repertoire of humans has been well investigated. There are three types of catalytic domains (Z domain; A3Z1, A3Z2, and A3Z3), 11 Z domains, and 7 independent genes, including 4 genes encoding double Z domains. However, the APOBEC3 gene repertoires of nonhuman primates remain largely unclear. Here, we characterize APOBEC3 gene repertoires among primates and investigated the evolutionary scenario of primate APOBEC3 genes using phylogenetic and comparative genomics approaches. In the 21 primate species investigated, we identified 145 APOBEC3 genes, including 69 double-domain type APOBEC3 genes. We further estimated the ages of the respective APOBEC3 genes and revealed that APOBEC3B, APOBEC3D, and APOBEC3F are the youngest in humans and were generated in the common ancestor of Catarrhini. Notably, invasion of the LINE1 retrotransposon peaked during the same period as the generation of these youngest APOBEC3 genes, implying that LINE1 invasion was one of the driving forces of the generation of these genes. Moreover, we found evidence suggesting that sequence diversification by gene conversions among APOBEC3 paralogs occurred in multiple primate lineages. Together, our analyses reveal the hidden diversity and the complicated evolutionary scenario of APOBEC3 genes in primates.IMPORTANCE In terms of virus-host interactions and coevolution, the APOBEC3 gene family is one of the most important subjects in the field of retrovirology. APOBEC3 genes are composed of a repertoire of subclasses based on sequence similarity, and a paper by LaRue et al. provides the standard guideline for the nomenclature and genomic architecture of APOBEC3 genes. However, it has been more than 10 years since this publication, and new information, including RefSeq, which we used in this study, is accumulating. Based on accumulating knowledge, APOBEC3 genes, particularly those of primates, should be refined and reannotated. This study updates knowledge of primate APOBEC3 genes and their genomic architectures. We further inferred the evolutionary scenario of primate APOBEC3 genes and the potential driving forces of APOBEC3 gene evolution. This study will be a landmark for the elucidation of the multiple aspects of APOBEC3 family genes in the future.


Subject(s)
APOBEC Deaminases/genetics , Evolution, Molecular , Primates/genetics , APOBEC Deaminases/chemistry , Animals , Catalytic Domain , Cytidine Deaminase/chemistry , Cytidine Deaminase/genetics , Cytosine Deaminase/chemistry , Cytosine Deaminase/genetics , Gene Conversion , Humans , Long Interspersed Nucleotide Elements , Minor Histocompatibility Antigens/chemistry , Minor Histocompatibility Antigens/genetics , Phylogeny
3.
Nucleic Acids Res ; 48(3): 1353-1371, 2020 02 20.
Article in English | MEDLINE | ID: mdl-31943071

ABSTRACT

The human apolipoprotein B mRNA editing enzyme, catalytic polypeptide-like 3 (APOBEC3, A3) family member proteins can deaminate cytosines in single-strand (ss) DNA, which restricts human immunodeficiency virus type 1 (HIV-1), retrotransposons, and other viruses such as hepatitis B virus, but can cause a mutator phenotype in many cancers. While structural information exists for several A3 proteins, the precise details regarding deamination target selection are not fully understood. Here, we report the first parallel, comparative analysis of site selection of A3 deamination using six of the seven purified A3 member enzymes, oligonucleotides having 5'TC3' or 5'CT3' dinucleotide target sites, and different flanking bases within diverse DNA secondary structures. A3A, A3F and A3H were observed to have strong preferences toward the TC target flanked by A or T, while all examined A3 proteins did not show a preference for a TC target flanked by a G. We observed that the TC target was strongly preferred in ssDNA regions rather than dsDNA, loop or bulge regions, with flanking bases influencing the degree of preference. CT was also shown to be a potential deamination target. Taken together, our observations provide new insights into A3 enzyme target site selection and how A3 mutagenesis impacts mutation rates.


Subject(s)
Cytidine Deaminase/genetics , DNA, Single-Stranded/genetics , DNA-Binding Proteins/genetics , Deamination/genetics , APOBEC Deaminases , Binding Sites/genetics , Cell Line , Cytidine Deaminase/chemistry , Cytosine Deaminase/chemistry , Cytosine Deaminase/genetics , DNA, Single-Stranded/chemistry , DNA-Binding Proteins/chemistry , HIV-1/genetics , HIV-1/pathogenicity , Hepatitis B virus/genetics , Humans , Mutagenesis/genetics , Nucleic Acid Conformation , Protein Structure, Secondary , Retroelements/genetics
4.
Mol Cell ; 49(4): 632-44, 2013 Feb 21.
Article in English | MEDLINE | ID: mdl-23333304

ABSTRACT

The HIV-1 accessory protein Vif hijacks a cellular Cullin-RING ubiquitin ligase, CRL5, to promote degradation of the APOBEC3 (A3) family of restriction factors. Recently, the cellular transcription cofactor CBFß was shown to form a complex with CRL5-Vif and to be essential for A3 degradation and viral infectivity. We now demonstrate that CBFß is required for assembling a well-ordered CRL5-Vif complex by inhibiting Vif oligomerization and by activating CRL5-Vif via direct interaction. The CRL5-Vif-CBFß holoenzyme forms a well-defined heterohexamer, indicating that Vif simultaneously hijacks CRL5 and CBFß. Heterodimers of CBFß and RUNX transcription factors contribute toward the regulation of genes, including those with immune system functions. We show that binding of Vif to CBFß is mutually exclusive with RUNX heterodimerization and impacts the expression of genes whose regulatory domains are associated with RUNX1. Our results provide a mechanism by which a pathogen with limited coding capacity uses one factor to hijack multiple host pathways.


Subject(s)
CCAAT-Binding Factor/metabolism , Core Binding Factor Alpha 2 Subunit/metabolism , Cytosine Deaminase/metabolism , Gene Expression Regulation , vif Gene Products, Human Immunodeficiency Virus/metabolism , APOBEC Deaminases , Amino Acid Sequence , Base Sequence , CCAAT-Binding Factor/chemistry , CCAAT-Binding Factor/physiology , Consensus Sequence , Core Binding Factor Alpha 2 Subunit/chemistry , Core Binding Factor Alpha 2 Subunit/physiology , Cytidine Deaminase , Cytosine Deaminase/chemistry , Cytosine Deaminase/physiology , Gene Expression , Genes, Reporter , HEK293 Cells , HIV-1/physiology , Host-Pathogen Interactions , Humans , Hydrophobic and Hydrophilic Interactions , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Interaction Domains and Motifs , Protein Multimerization , Protein Processing, Post-Translational , Protein Stability , Protein Structure, Quaternary , T-Lymphocytes/metabolism , T-Lymphocytes/virology , Ubiquitination , vif Gene Products, Human Immunodeficiency Virus/chemistry , vif Gene Products, Human Immunodeficiency Virus/physiology
5.
Int J Mol Sci ; 22(4)2021 Feb 14.
Article in English | MEDLINE | ID: mdl-33672989

ABSTRACT

Epidermal growth factor receptor (EGFR) specific therapeutics is of great importance in cancer treatment. Fcy-hEGF fusion protein, composed of yeast cytosine deaminase (Fcy) and human EGF (hEGF), is capable of binding to EGFR and enzymatically convert 5-fluorocytosine (5-FC) to 1000-fold toxic 5-fluorocuracil (5-FU), thereby inhibiting the growth of EGFR-expressing tumor cells. To develop EGFR-specific therapy, 188Re-liposome-Fcy-hEGF was constructed by insertion of Fcy-hEGF fusion protein onto the surface of liposomes encapsulating of 188Re. Western blotting, MALDI-TOF, column size exclusion and flow cytometry were used to confirm the conjugation and bio-activity of 188Re-liposome-Fcy-hEGF. Cell lines with EGFR expression were subjected to treat with 188Re-liposome-Fcy-hEGF/5-FC in the presence of 5-FC. The 188Re-liposome-Fcy-hEGF/5-FC revealed a better cytotoxic effect for cancer cells than the treatment of liposome-Fcy-hEGF/5-FC or 188Re-liposome-Fcy-hEGF alone. The therapeutics has radio- and chemo-toxicity simultaneously and specifically target to EGFR-expression tumor cells, thereby achieving synergistic anticancer activity.


Subject(s)
Cytosine Deaminase/metabolism , Epidermal Growth Factor/metabolism , ErbB Receptors/metabolism , Fluorouracil/pharmacology , Neoplasms/metabolism , Radiopharmaceuticals/pharmacology , Antimetabolites, Antineoplastic/metabolism , Antimetabolites, Antineoplastic/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/radiation effects , Cytosine Deaminase/chemistry , Epidermal Growth Factor/chemistry , Flucytosine/metabolism , Fluorouracil/metabolism , Humans , Liposomes/chemistry , MCF-7 Cells , Neoplasms/pathology , Protein Binding , Radioisotopes/chemistry , Radiopharmaceuticals/chemistry , Rhenium/chemistry
6.
Chembiochem ; 20(15): 1898-1905, 2019 08 01.
Article in English | MEDLINE | ID: mdl-30809902

ABSTRACT

Over the past few decades, various DNA modification detection methods have been developed; many of the high-resolution methods are based on bisulfite treatment, which leads to DNA degradation, to a degree. Thus, novel bisulfite-free approaches have been developed in recent years and shown to be useful for epigenome analysis in otherwise difficult-to-handle, but important, DNA samples, such as hmC-seal and hmC-CATCH. Herein, an overview of advances in the development of epigenome sequencing methods for these important DNA modifications is provided.


Subject(s)
Cytosine/metabolism , Cytosine/chemistry , Cytosine Deaminase/chemistry , Cytosine Deaminase/metabolism , DNA/chemistry , DNA/genetics , DNA/metabolism , Humans , Molecular Structure
7.
Bioconjug Chem ; 30(2): 432-442, 2019 02 20.
Article in English | MEDLINE | ID: mdl-30615416

ABSTRACT

Proteins are ideal candidates for disease treatment because of their high specificity and potency. Despite this potential, delivery of proteins remains a significant challenge due to the intrinsic size, charge, and stability of proteins. Attempts to overcome these challenges have most commonly relied on direct conjugation of polymers and peptides to proteins via reactive groups on naturally occurring residues. While such approaches have shown some success, they allow limited control of the spacing and number of moieties coupled to proteins, which can hinder bioactivity and delivery capabilities of the therapeutic. Here, we describe a strategy to site-specifically conjugate delivery moieties to therapeutic proteins through unnatural amino acid (UAA) incorporation, in order to explore the effect of epidermal growth factor receptor (EGFR)-targeted ligand valency and spacing on internalization of proteins in EGFR-overexpressing inflammatory breast cancer (IBC) cells. Our results demonstrate the ability to enhance targeted protein delivery by tuning a small number of EGFR ligands per protein and clustering these ligands to promote multivalent ligand-receptor interactions. Furthermore, the tailorability of this simple approach was demonstrated through IBC-targeted cell death via the delivery of yeast cytosine deaminase (yCD), a prodrug converting enzyme.


Subject(s)
Amino Acids/metabolism , Cytosine Deaminase/administration & dosage , Luminescent Proteins/administration & dosage , Amino Acids/chemistry , Breast Neoplasms/metabolism , Cell Line, Tumor , Click Chemistry , Cytosine Deaminase/chemistry , Cytosine Deaminase/pharmacokinetics , Drug Delivery Systems , ErbB Receptors/metabolism , Female , Humans , Ligands , Luminescent Proteins/chemistry , Luminescent Proteins/pharmacokinetics , Models, Molecular , Protein Binding , Yeasts/enzymology , Red Fluorescent Protein
8.
Biomacromolecules ; 20(4): 1683-1690, 2019 04 08.
Article in English | MEDLINE | ID: mdl-30884222

ABSTRACT

The simultaneous delivery of multiple therapeutics to a single site has shown promise for cancer targeting and treatment. However, because of the inherent differences in charge and size between drugs and biomolecules, new approaches are required for colocalization of unlike components in one delivery vehicle. In this work, we demonstrate that triblock copolymers containing click nucleic acids (CNAs) can be used to simultaneously load a prodrug enzyme (cytosine deaminase, CodA) and a chemotherapy drug (doxorubicin, DOX) in a single polymer nanoparticle. CNAs are synthetic analogs of DNA comprised of a thiolene backbone and nucleotide bases that can hybridize to complementary strands of DNA. In this study, CodA was appended with complementary DNA sequences and fluorescent dyes to allow its encapsulation in PEG-CNA-PLGA nanoparticles. The DNA-modified CodA was found to retain its enzyme activity for converting prodrug 5-fluorocytosine (5-FC) to active 5-fluorouracil (5-FU) using a modified fluorescent assay. The DNA-conjugated CodA was then loaded into the PEG-CNA-PLGA nanoparticles and tested for cell cytotoxicity in the presence of the 5-FC prodrug. To study the effect of coloading DOX and CodA within a single nanoparticle, cell toxicity assays were run to compare dually loaded nanoparticles with nanoparticles loaded only with either DOX or CodA. We show that the highest level of cell death occurred when both DOX and CodA were simultaneously entrapped and delivered to cells in the presence of 5-FC.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Cytosine Deaminase , DNA , Drug Carriers , Enzymes, Immobilized , Escherichia coli Proteins , Nanoparticles , Neoplasms , Polyesters , Polyethylene Glycols , Prodrugs , Antineoplastic Combined Chemotherapy Protocols/chemistry , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Line, Tumor , Cytosine Deaminase/chemistry , Cytosine Deaminase/pharmacology , DNA/chemistry , DNA/pharmacology , Doxorubicin/chemistry , Doxorubicin/pharmacokinetics , Doxorubicin/pharmacology , Drug Carriers/chemistry , Drug Carriers/therapeutic use , Enzymes, Immobilized/chemistry , Enzymes, Immobilized/pharmacology , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/pharmacology , Flucytosine/chemistry , Flucytosine/pharmacokinetics , Flucytosine/pharmacology , Humans , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Polyesters/chemical synthesis , Polyesters/chemistry , Polyesters/pharmacology , Polyethylene Glycols/chemical synthesis , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacology , Prodrugs/chemistry , Prodrugs/pharmacology
9.
J Am Chem Soc ; 140(1): 14-17, 2018 01 10.
Article in English | MEDLINE | ID: mdl-29251923

ABSTRACT

There is growing interest in designing spatiotemporal control over enzyme activities using noninvasive stimuli such as light. Here, we describe a structure-based, computation-guided predictive method for reversibly controlling enzyme activity using covalently attached photoresponsive azobenzene groups. Applying the method to the therapeutically useful enzyme yeast cytosine deaminase, we obtained a ∼3-fold change in enzyme activity by the photocontrolled modulation of the enzyme's active site lid structure, while fully maintaining thermostability. Multiple cycles of switching, controllable in real time, are possible. The predictiveness of the method is demonstrated by the construction of a variant that does not photoswitch as expected from computational modeling. Our design approach opens new avenues for optically controlling enzyme function. The designed photocontrolled cytosine deaminases may also aid in improving chemotherapy approaches that utilize this enzyme.


Subject(s)
Azo Compounds/chemistry , Cytosine Deaminase/chemistry , Cytosine Deaminase/radiation effects , Photochemical Processes , Azo Compounds/metabolism , Cytosine Deaminase/metabolism , Models, Molecular , Saccharomyces cerevisiae/enzymology
10.
Retrovirology ; 15(1): 31, 2018 04 10.
Article in English | MEDLINE | ID: mdl-29636069

ABSTRACT

BACKGROUND: The apolipoprotein B mRNA-editing enzyme catalytic polypeptide-like 3 (APOBEC3; A3) gene family appears only in mammalian genomes. Some A3 proteins can be incorporated into progeny virions and inhibit lentiviral replication. In turn, the lentiviral viral infectivity factor (Vif) counteracts the A3-mediated antiviral effect by degrading A3 proteins. Recent investigations have suggested that lentiviral vif genes evolved to combat mammalian APOBEC3 proteins, and have further proposed that the Vif-A3 interaction may help determine the co-evolutionary history of cross-species lentiviral transmission in mammals. RESULTS: Here we address the co-evolutionary relationship between two New World felids, the puma (Puma concolor) and the bobcat (Lynx rufus), and their lentiviruses, which are designated puma lentiviruses (PLVs). We demonstrate that PLV-A Vif counteracts the antiviral action of APOBEC3Z3 (A3Z3) of both puma and bobcat, whereas PLV-B Vif counteracts only puma A3Z3. The species specificity of PLV-B Vif is irrespective of the phylogenic relationships of feline species in the genera Puma, Lynx and Acinonyx. We reveal that the amino acid at position 178 in the puma and bobcat A3Z3 is exposed on the protein surface and determines the sensitivity to PLV-B Vif-mediated degradation. Moreover, although both the puma and bobcat A3Z3 genes are polymorphic, their sensitivity/resistance to PLV Vif-mediated degradation is conserved. CONCLUSIONS: To the best of our knowledge, this is the first study suggesting that the host A3 protein potently controls inter-genus lentiviral transmission. Our findings provide the first evidence suggesting that the co-evolutionary arms race between lentiviruses and mammals has occurred in the New World.


Subject(s)
Cytosine Deaminase/genetics , Host-Pathogen Interactions/genetics , Lentivirus Infections/transmission , Lentivirus Infections/virology , Lentivirus/physiology , Animals , Cats , Cytosine Deaminase/chemistry , Cytosine Deaminase/metabolism , Disease Resistance , Evolution, Molecular , Gene Products, vif , Immunodeficiency Virus, Feline/classification , Immunodeficiency Virus, Feline/genetics , Lentivirus/classification , Loss of Function Mutation , Models, Molecular , Phylogeny , Polymorphism, Genetic , Protein Conformation , Proteolysis , Structure-Activity Relationship , Threonine/chemistry , Threonine/genetics
11.
J Virol ; 91(3)2017 Feb 01.
Article in English | MEDLINE | ID: mdl-27881650

ABSTRACT

The APOBEC3 (A3) enzymes, A3G and A3F, are coordinately expressed in CD4+ T cells and can become coencapsidated into HIV-1 virions, primarily in the absence of the viral infectivity factor (Vif). A3F and A3G are deoxycytidine deaminases that inhibit HIV-1 replication by inducing guanine-to-adenine hypermutation through deamination of cytosine to form uracil in minus-strand DNA. The effect of the simultaneous presence of both A3G and A3F on HIV-1 restriction ability is not clear. Here, we used a single-cycle infectivity assay and biochemical analyses to determine if coencapsidated A3G and A3F differ in their restriction capacity from A3G or A3F alone. Proviral DNA sequencing demonstrated that compared to each A3 enzyme alone, A3G and A3F, when combined, had a coordinate effect on hypermutation. Using size exclusion chromatography, rotational anisotropy, and in vitro deamination assays, we demonstrate that A3F promotes A3G deamination activity by forming an A3F/G hetero-oligomer in the absence of RNA which is more efficient at deaminating cytosines. Further, A3F caused the accumulation of shorter reverse transcripts due to decreasing reverse transcriptase efficiency, which would leave single-stranded minus-strand DNA exposed for longer periods of time, enabling more deamination events to occur. Although A3G and A3F are known to function alongside each other, these data provide evidence for an A3F/G hetero-oligomeric A3 with unique properties compared to each individual counterpart. IMPORTANCE: The APOBEC3 enzymes APOBEC3F and APOBEC3G act as a barrier to HIV-1 replication in the absence of the HIV-1 Vif protein. After APOBEC3 enzymes are encapsidated into virions, they deaminate cytosines in minus-strand DNA, which forms promutagenic uracils that induce transition mutations or proviral DNA degradation. Even in the presence of Vif, footprints of APOBEC3-catalyzed deaminations are found, demonstrating that APOBEC3s still have discernible activity against HIV-1 in infected individuals. We undertook a study to better understand the activity of coexpressed APOBEC3F and APOBEC3G. The data demonstrate that an APOBEC3F/APOBEC3G hetero-oligomer can form that has unique properties compared to each APOBEC3 alone. This hetero-oligomer has increased efficiency of virus hypermutation, raising the idea that we still may not fully realize the antiviral mechanisms of endogenous APOBEC3 enzymes. Hetero-oligomerization may be a mechanism to increase their antiviral activity in the presence of Vif.


Subject(s)
APOBEC-3G Deaminase/metabolism , Capsid/metabolism , Cytosine Deaminase/metabolism , HIV Infections/metabolism , HIV Infections/virology , HIV-1/physiology , Virion/physiology , APOBEC-3G Deaminase/chemistry , Cell Line , Cytosine Deaminase/chemistry , Gene Expression , Humans , Intracellular Space , Protein Binding , Protein Multimerization , Protein Transport , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , Recombinant Fusion Proteins , Reverse Transcription , Sequence Deletion , Virus Assembly , Virus Replication , vif Gene Products, Human Immunodeficiency Virus/genetics , vif Gene Products, Human Immunodeficiency Virus/metabolism
12.
Biomacromolecules ; 19(10): 3945-3957, 2018 10 08.
Article in English | MEDLINE | ID: mdl-30160482

ABSTRACT

The simultaneous expression in Escherichia coli cells of the Qß virus-like particle (VLP) capsid protein and protein "cargo" tagged with a positively charged Rev peptide sequence leads to the spontaneous self-assembly of VLPs with multiple copies of the cargo inside. We report the packaging of four new enzymes with potential applications in medicine and chemical manufacturing. The captured enzymes are active while inside the nanoparticle shell and are protected from environmental conditions that lead to free-enzyme destruction. We also describe genetic modifications to the packaging scheme that shed light on the self-assembly mechanism of this system and allow indirect control over the internal packaging density of cargo. The technology was extended to create, via self-assembly, VLPs that simultaneously display protein ligands on the exterior and contain enzymes within. Inverse relationships were observed between the size of both the packaged and externally displayed protein or domains and nanoparticle yield. These results provide a general method for the rapid creation of robust protein nanoparticles with desired catalytic and targeting functionalities.


Subject(s)
Capsid Proteins/metabolism , Gene Products, rev/metabolism , Multifunctional Enzymes/chemistry , Multifunctional Enzymes/metabolism , Nanoparticles/metabolism , RNA, Viral/metabolism , Virus Assembly , Aldehyde-Lyases/chemistry , Aldehyde-Lyases/genetics , Aldehyde-Lyases/metabolism , Capsid Proteins/chemistry , Capsid Proteins/genetics , Catalysis , Cytosine Deaminase/chemistry , Cytosine Deaminase/genetics , Cytosine Deaminase/metabolism , Gene Products, rev/chemistry , Gene Products, rev/genetics , HeLa Cells , Humans , Multifunctional Enzymes/genetics , Nanoparticles/chemistry , RNA, Viral/chemistry , RNA, Viral/genetics
13.
Phys Chem Chem Phys ; 20(5): 3109-3117, 2018 Jan 31.
Article in English | MEDLINE | ID: mdl-28825755

ABSTRACT

APOBEC3F (A3F), an apolipoprotein B mRNA-editing enzyme catalytic polypeptide-like 3 (APOBEC3) family protein, catalyzes cytosine-to-uracil conversion in single-stranded (ss) DNA. A3F acts as an inhibitor of retrovirus replication and exhibits antiviral activity against viral infectivity factor (Vif)-deficient human immunodeficiency virus 1 (HIV-1). Previous studies have mostly been focused on the interaction between A3F and Vif, and the studies on A3F's deamination properties are limited. Here, we report comprehensive characterization of the deaminase activity and ssDNA binding of the C-terminal domain (CTD) of A3F. It was shown that the deaminase activity of A3F-CTD is affected by the nucleic acid residues adjacent to the target sequence, TC, and that TTCA/G are the most preferred sequences. A3F-CTD deaminates the target sequence in longer ssDNAs most efficiently. Mutation analysis identified the amino acid residues that are responsible for the deaminase activity and ssDNA binding in the loops surrounding the catalytic center. The functions of these residues were rationally interpreted on the basis of the co-crystal structure of A3A-ssDNA and the known roles of the equivalent amino acid residues found in other A3s. Furthermore, we demonstrated that the deaminase activity of A3F-CTD could be regulated through phosphorylation of a putative site, S216. Finally, A3F-CTD was found to be active in a wide pH range (5.5 to 9.5) with similar activity. Interestingly, the A3F-CTD N214H mutant exhibited a dramatic increase in activity at pH 5.5.


Subject(s)
Amino Acids/metabolism , Cytosine Deaminase/chemistry , Amino Acid Sequence , Amino Acids/chemistry , Base Sequence , Binding Sites , Biocatalysis , Catalytic Domain , Crystallography, X-Ray , Cytosine Deaminase/genetics , Cytosine Deaminase/metabolism , DNA, Single-Stranded/metabolism , Deamination , Fluorescence Polarization , Humans , Hydrogen-Ion Concentration , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , Phosphorylation , Protein Binding , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification
14.
J Virol ; 90(2): 1034-47, 2016 01 15.
Article in English | MEDLINE | ID: mdl-26537685

ABSTRACT

UNLABELLED: The HIV-1 Vif protein inactivates the cellular antiviral cytidine deaminase APOBEC3F (A3F) in virus-infected cells by specifically targeting it for proteasomal degradation. Several studies identified Vif sequence motifs involved in A3F interaction, whereas a Vif-binding A3F interface was proposed based on our analysis of highly similar APOBEC3C (A3C). However, the structural mechanism of specific Vif-A3F recognition is still poorly understood. Here we report structural features of interaction interfaces for both HIV-1 Vif and A3F molecules. Alanine-scanning analysis of Vif revealed that six residues located within the conserved Vif F1-, F2-, and F3-box motifs are essential for both A3C and A3F degradation, and an additional four residues are uniquely required for A3F degradation. Modeling of the Vif structure on an HIV-1 Vif crystal structure revealed that three discontinuous flexible loops of Vif F1-, F2-, and F3-box motifs sterically cluster to form a flexible A3F interaction interface, which represents hydrophobic and positively charged surfaces. We found that the basic Vif interface patch (R17, E171, and R173) involved in the interactions with A3C and A3F differs. Furthermore, our crystal structure determination and extensive mutational analysis of the A3F C-terminal domain demonstrated that the A3F interface includes a unique acidic stretch (L291, A292, R293, and E324) crucial for Vif interaction, suggesting additional electrostatic complementarity to the Vif interface compared with the A3C interface. Taken together, these findings provide structural insights into the A3F-Vif interaction mechanism, which will provide an important basis for development of novel anti-HIV-1 drugs using cellular cytidine deaminases. IMPORTANCE: HIV-1 Vif targets cellular antiviral APOBEC3F (A3F) enzyme for degradation. However, the details on the structural mechanism for specific A3F recognition remain unclear. This study reports structural features of interaction interfaces for both HIV-1 Vif and A3F molecules. Three discontinuous sequence motifs of Vif, F1, F2, and F3 boxes, assemble to form an A3F interaction interface. In addition, we determined a crystal structure of the wild-type A3F C-terminal domain responsible for the Vif interaction. These results demonstrated that both electrostatic and hydrophobic interactions are the key force driving Vif-A3F binding and that the Vif-A3F interfaces are larger than the Vif-A3C interfaces. These findings will allow us to determine the configurations of the Vif-A3F complex and to construct a structural model of the complex, which will provide an important basis for inhibitor development.


Subject(s)
Cytosine Deaminase/chemistry , Cytosine Deaminase/metabolism , vif Gene Products, Human Immunodeficiency Virus/chemistry , vif Gene Products, Human Immunodeficiency Virus/metabolism , Crystallography, X-Ray , Cytidine Deaminase/chemistry , Cytidine Deaminase/metabolism , DNA Mutational Analysis , Models, Molecular , Mutagenesis, Site-Directed , Protein Conformation , Protein Interaction Mapping , Proteolysis , vif Gene Products, Human Immunodeficiency Virus/genetics
15.
Biochim Biophys Acta Proteins Proteom ; 1865(8): 1020-1029, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28478051

ABSTRACT

Yeast cytosine deaminase (yCD) is critical in gene-directed enzyme prodrug therapy as it catalyzes the hydrolytic deamination of cytosine. The product (uracil) release process is considered as rate-limiting in the whole enzymatic catalysis and includes the cleavage of the uracil-metal bond and the delivery of free uracil out of the reactive site. Herein extensive combined random acceleration molecular dynamics (RAMD) and molecular dynamics (MD) simulations coupled with the umbrella sampling technique have been performed to study the product transport mechanism. Five channels have been identified, and the thermodynamic and dynamic characterizations for the two most favorable channels have been determined and analyzed. The free energy barrier for the most beneficial pathway is about 13kcal/mol and mainly results from the cleavage of hydrogen bonds between the ligand uracil and surrounding residues Asn51, Glu64, and Asp155. The conjugated rings of Phe114 and Trp152 play gating and guiding roles in the product delivery via π⋯π van der Waals interactions with the product. Finally, the full cycle of the enzymatic catalysis has been determined, making the whole process computationally more precise.


Subject(s)
Cytosine Deaminase/chemistry , Cytosine Deaminase/metabolism , Yeasts/metabolism , Binding Sites/physiology , Catalysis , Computational Biology/methods , Cytosine/chemistry , Cytosine/metabolism , Hydrogen Bonding , Molecular Dynamics Simulation , Prodrugs/chemistry , Thermodynamics , Uracil/chemistry , Uracil/metabolism
17.
Biochemistry ; 53(47): 7426-35, 2014 Dec 02.
Article in English | MEDLINE | ID: mdl-25384249

ABSTRACT

5-Methylcytosine is found in all domains of life, but the bacterial cytosine deaminase from Escherichia coli (CodA) will not accept 5-methylcytosine as a substrate. Since significant amounts of 5-methylcytosine are produced in both prokaryotes and eukaryotes, this compound must eventually be catabolized and the fragments recycled by enzymes that have yet to be identified. We therefore initiated a comprehensive phylogenetic screen for enzymes that may be capable of deaminating 5-methylcytosine to thymine. From a systematic analysis of sequence homologues of CodA from thousands of bacterial species, we identified putative cytosine deaminases where a "discriminating" residue in the active site, corresponding to Asp-314 in CodA from E. coli, was no longer conserved. Representative examples from Klebsiella pneumoniae (locus tag: Kpn00632), Rhodobacter sphaeroides (locus tag: Rsp0341), and Corynebacterium glutamicum (locus tag: NCgl0075) were demonstrated to efficiently deaminate 5-methylcytosine to thymine with values of kcat/Km of 1.4 × 10(5), 2.9 × 10(4), and 1.1 × 10(3) M(-1) s(-1), respectively. These three enzymes also catalyze the deamination of 5-fluorocytosine to 5-fluorouracil with values of kcat/Km of 1.2 × 10(5), 6.8 × 10(4), and 2.0 × 10(2) M(-1) s(-1), respectively. The three-dimensional structure of Kpn00632 was determined by X-ray diffraction methods with 5-methylcytosine (PDB id: 4R85 ), 5-fluorocytosine (PDB id: 4R88 ), and phosphonocytosine (PDB id: 4R7W ) bound in the active site. When thymine auxotrophs of E. coli express these enzymes, they are capable of growth in media lacking thymine when supplemented with 5-methylcytosine. Expression of these enzymes in E. coli is toxic in the presence of 5-fluorocytosine, due to the efficient transformation to 5-fluorouracil.


Subject(s)
5-Methylcytosine/metabolism , Bacteria/enzymology , Cytosine Deaminase/metabolism , Amino Acid Sequence , Biocatalysis , Catalytic Domain , Cell Line , Cytosine Deaminase/chemistry , Flucytosine/metabolism , Flucytosine/toxicity , Models, Molecular , Molecular Sequence Data , Phylogeny , Thymine/metabolism
18.
J Phys Chem B ; 128(13): 3102-3112, 2024 Apr 04.
Article in English | MEDLINE | ID: mdl-38516924

ABSTRACT

The yeast cytosine deaminase (yCD) enzyme/5-fluorocytosine prodrug system is a promising candidate for targeted chemotherapeutics. After conversion of the prodrug into the toxic chemotherapeutic drug, 5-fluorouracil (5-FU), the slow product release from the enzyme limits the overall catalytic efficiency of the enzyme/prodrug system. Here, we present a computational study of the product release of the anticancer drug, 5-FU, from yCD using metadynamics. We present a comparison of the 5-FU drug to the natural enzyme product, uracil. We use volume-based metadynamics to compute the free energy landscape for product release and show a modest binding affinity for the product to the enzyme, consistent with experiments. Next, we use infrequent metadynamics to estimate the unbiased release rate from Kramers time-dependent rate theory and find a favorable comparison to experiment with a slower rate of product release for the 5-FU system. Our work demonstrates how adaptive sampling methods can be used to study the protein-ligand unbinding process for engineering enzyme/prodrug systems and gives insights into the molecular mechanism of product release for the yCD/5-FU system.


Subject(s)
Antineoplastic Agents , Prodrugs , Saccharomyces cerevisiae , Cytosine Deaminase/chemistry , Cytosine Deaminase/metabolism , Fluorouracil/metabolism , Flucytosine/chemistry , Flucytosine/metabolism , Prodrugs/chemistry
19.
J Biol Chem ; 287(19): 15544-56, 2012 May 04.
Article in English | MEDLINE | ID: mdl-22411989

ABSTRACT

This study tested whether nonredox metalloenzymes are commonly charged with iron in vivo and are primary targets of oxidative stress because of it. Indeed, three sample mononuclear enzymes, peptide deformylase, threonine dehydrogenase, and cytosine deaminase, were rapidly damaged by micromolar hydrogen peroxide in vitro and in live Escherichia coli. The first two enzymes use a cysteine residue to coordinate the catalytic metal atom; it was quantitatively oxidized by the radical generated by the Fenton reaction. Because oxidized cysteine can be repaired by cellular reductants, the effect was to avoid irreversible damage to other active-site residues. Nevertheless, protracted H(2)O(2) exposure gradually inactivated these enzymes, consistent with the overoxidation of the cysteine residue to sulfinic or sulfonic forms. During H(2)O(2) stress, E. coli defended all three proteins by inducing MntH, a manganese importer, and Dps, an iron-sequestration protein. These proteins appeared to collaborate in replacing the iron atom with nonoxidizable manganese. The implication is that mononuclear metalloproteins are common targets of H(2)O(2) and that both structural and metabolic arrangements exist to protect them.


Subject(s)
Alcohol Oxidoreductases/metabolism , Amidohydrolases/metabolism , Cytosine Deaminase/metabolism , Escherichia coli Proteins/metabolism , Hydrogen Peroxide/pharmacology , Alcohol Oxidoreductases/chemistry , Alcohol Oxidoreductases/genetics , Amidohydrolases/chemistry , Amidohydrolases/genetics , Apoproteins/chemistry , Apoproteins/metabolism , Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/metabolism , Biocatalysis/drug effects , Blotting, Western , Catalytic Domain , Cation Transport Proteins/chemistry , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Cysteine/chemistry , Cysteine/metabolism , Cytosine Deaminase/chemistry , Cytosine Deaminase/genetics , Escherichia coli/enzymology , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Iron/chemistry , Iron/metabolism , Kinetics , Manganese/chemistry , Manganese/metabolism , Models, Chemical , Mutation , Oxidants/pharmacology , Oxidation-Reduction/drug effects
20.
Biol Chem ; 394(11): 1357-70, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23787464

ABSTRACT

The APOBEC3 (A3) family of cytidine deaminases plays a vital role for innate defense against retroviruses. Lentiviruses such as HIV-1 evolved the Vif protein that triggers A3 protein degradation. There are seven A3 proteins, A3A-A3H, found in humans. All A3 proteins can deaminate cytidines to uridines in single-stranded DNA (ssDNA), generated during viral reverse transcription. A3 proteins have either one or two cytidine deaminase domains (CD). The CDs coordinate a zinc ion, and their amino acid specificity classifies the A3s into A3Z1, A3Z2, and A3Z3. A3 proteins occur as monomers, dimers, and large oligomeric complexes. Studies on the nature of A3 oligomerization, as well as the mode of interaction of A3s with RNA and ssDNA are partially controversial. High-resolution structures of the catalytic CD2 of A3G and A3F as well as of the single CD proteins A3A and A3C have been published recently. The NMR and X-ray crystal structures show globular proteins with six α-helices and five ß sheets arranged in a characteristic motif (α1-ß1-ß2/2'-α2-ß3-α3-ß4-α4-ß5-α5-α6). However, the detailed arrangement and extension of individual structure elements and their relevance for A3 complex formation and activity remains a matter of debate and will be highlighted in this review.


Subject(s)
Cytosine Deaminase/chemistry , DNA, Viral/chemistry , APOBEC Deaminases , Catalysis , Cytidine Deaminase , Cytosine Deaminase/metabolism , DNA, Single-Stranded/chemistry , DNA, Single-Stranded/metabolism , DNA, Viral/antagonists & inhibitors , Humans , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Protein Structure, Tertiary , Retroviridae/enzymology , Retroviridae/metabolism , Scattering, Small Angle
SELECTION OF CITATIONS
SEARCH DETAIL