Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 591
Filter
1.
Nat Commun ; 13(1): 7498, 2022 12 05.
Article in English | MEDLINE | ID: mdl-36470880

ABSTRACT

An essential step in restricting HIV infectivity by the antiviral factor APOBEC3G is its incorporation into progeny virions via binding to HIV RNA. However, the mechanism of APOBEC3G capturing viral RNA is unknown. Here, we report crystal structures of a primate APOBEC3G bound to different types of RNAs, revealing that APOBEC3G specifically recognizes unpaired 5'-AA-3' dinucleotides, and to a lesser extent, 5'-GA-3' dinucleotides. APOBEC3G binds to the common 3'A in the AA/GA motifs using an aromatic/hydrophobic pocket in the non-catalytic domain. It binds to the 5'A or 5'G in the AA/GA motifs using an aromatic/hydrophobic groove conformed between the non-catalytic and catalytic domains. APOBEC3G RNA binding property is distinct from that of the HIV nucleocapsid protein recognizing unpaired guanosines. Our findings suggest that the sequence-specific RNA recognition is critical for APOBEC3G virion packaging and restricting HIV infectivity.


Subject(s)
HIV Infections , HIV-1 , Nucleoside Deaminases , Animals , APOBEC-3G Deaminase/metabolism , Cytidine Deaminase/genetics , HIV-1/genetics , Antiviral Agents/metabolism , Nucleoside Deaminases/metabolism , Virion/metabolism , RNA, Viral/metabolism , HIV Infections/metabolism
2.
Trends Genet ; 38(11): 1147-1169, 2022 11.
Article in English | MEDLINE | ID: mdl-35853769

ABSTRACT

Genome editing continues to revolutionize biological research. Due to its simplicity and flexibility, CRISPR/Cas-based editing has become the preferred technology in most systems. Cas nucleases tolerate fusion to large protein domains, thus allowing combination of their DNA recognition properties with new enzymatic activities. Fusion to nucleoside deaminase or reverse transcriptase domains has produced base editors and prime editors that, instead of generating double-strand breaks in the target sequence, induce site-specific alterations of single (or a few adjacent) nucleotides. The availability of protein-only genome editing reagents based on transcription activator-like effectors has enabled the extension of base editing to the genomes of chloroplasts and mitochondria. In this review, we summarize currently available base editing methods for nuclear and organellar genomes. We highlight recent advances with improving precision, specificity, and efficiency and discuss current limitations and future challenges. We also provide a brief overview of applications in agricultural biotechnology and gene therapy.


Subject(s)
CRISPR-Cas Systems , Nucleoside Deaminases , CRISPR-Cas Systems/genetics , DNA/genetics , DNA Breaks, Double-Stranded , Gene Editing/methods , Nucleoside Deaminases/genetics , Nucleoside Deaminases/metabolism , Nucleotides , RNA-Directed DNA Polymerase/genetics , RNA-Directed DNA Polymerase/metabolism , Transcription Activator-Like Effectors/genetics , Transcription Activator-Like Effectors/metabolism
3.
Arch Microbiol ; 204(7): 383, 2022 Jun 10.
Article in English | MEDLINE | ID: mdl-35689128

ABSTRACT

The control of a pyrimidine ribonucleotide salvage pathway in the bacterium Pseudomonas oleovorans ATCC 8062 was studied. This bacterium is important for its ability to synthesize polyesters as well as for its increasing clinical significance in humans. The pyrimidine salvage pathway enzymes pyrimidine nucleotide N-ribosidase and cytosine deaminase were investigated in P. oleovorans ATCC 8062 under selected culture conditions. Initially, the effect of carbon source on the two pyrimidine salvage enzymes in ATCC 8062 cells was examined and it was observed that cell growth on the carbon source succinate generally produced higher enzyme activities than did glucose or glycerol as a carbon source when ammonium sulfate served as the nitrogen source. Using succinate as a carbon source, growth on dihydrouracil as nitrogen source caused a 1.9-fold increase in the pyrimidine nucleotide N-ribosidase activity and a 4.8-fold increase in cytosine deaminase activity compared to the ammonium sulfate-grown cells. Growth of ATCC 8062 cells on cytosine or dihydrothymine as a nitrogen source elevated deaminase activity by more than double that observed for ammonium sulfate-grown cells. The findings indicated a relationship between this pyrimidine salvage pathway and the pyrimidine reductive catabolic pathway since growth on dihydrouracil appeared to increase the degradation of the pyrimidine ribonucleotide monophosphates to uracil. The uracil produced could be degraded by the pyrimidine base reductive catabolic pathway to ß-alanine as a source of nitrogen. This investigation could prove helpful to future work examining the metabolic relationship between pyrimidine salvage pathways and pyrimidine reductive catabolism in pseudomonads.


Subject(s)
Nucleoside Deaminases , Pseudomonas oleovorans , Ammonium Sulfate , Carbon , Cytosine Deaminase , Humans , Nitrogen , Nucleoside Deaminases/metabolism , Pyrimidine Nucleotides , Pyrimidines/metabolism , Ribonucleotides , Succinic Acid/metabolism , Uracil/metabolism
4.
Chem Commun (Camb) ; 57(76): 9748-9751, 2021 Sep 23.
Article in English | MEDLINE | ID: mdl-34477187

ABSTRACT

Guanosine deaminase (GSDA) in plants specifically deaminates (de)guanosine to produce xanthosine with high specificity, which is further converted to xanthine, a key intermediate in purine metabolism and nitrogen recycling. We solved GSDA's structures from Arabidopsis thaliana in the free and ligand-bound forms at high resolutions. Unlike GDA, the enzyme employs a single-proton shuttle mechanism for catalysis and both the substrate and enzyme undergo structural rearrangements. The last fragment of the enzyme loops back and seals the active site, and the substrate rotates during the reaction, both essential to deamination. We further identified more substrates that could be employed by the enzyme and compare it with other deaminases to reveal the recognition differences of specific substrates. Our studies provide insight into this important enzyme involved in purine metabolism and will potentially aid in the development of deaminase-based gene-editing tools.


Subject(s)
Arabidopsis/enzymology , Nucleoside Deaminases/metabolism , Biocatalysis , Models, Molecular , Nucleoside Deaminases/chemistry
5.
Chembiochem ; 21(18): 2676-2679, 2020 09 14.
Article in English | MEDLINE | ID: mdl-32291886

ABSTRACT

The green and sustainable synthesis of chemicals from renewable feedstocks by a biotransformation approach has gained increasing attention in recent years. In this work, we developed enzymatic cascades to efficiently convert l-phenylalanine into 2-phenylethanol (2-PE) and phenylacetic acid (PAA), l-tyrosine into tyrosol (p-hydroxyphenylethanol, p-HPE) and p-hydroxyphenylacetic acid (p-HPAA). The enzymatic cascade was cast into an aromatic aldehyde formation module, followed by an aldehyde reduction module, or aldehyde oxidation module, to achieve one-pot biotransformation by using recombinant Escherichia coli. Biotransformation of 50 mM l-Phe produced 6.76 g/L PAA with more than 99 % conversion and 5.95 g/L of 2-PE with 97 % conversion. The bioconversion efficiencies of p-HPAA and p-HPE from l-Tyr reached to 88 and 94 %, respectively. In addition, m-fluoro-phenylalanine was further employed as an unnatural aromatic amino acid substrate to obtain m-fluoro-phenylacetic acid; >96 % conversion was achieved. Our results thus demonstrated high-yielding and potential industrial synthesis of above aromatic compounds by one-pot cascade biocatalysis.


Subject(s)
Carboxy-Lyases/metabolism , Nucleoside Deaminases/metabolism , Oxidoreductases/metabolism , Phenylacetates/metabolism , Phenylethyl Alcohol/metabolism , Aldehydes/chemistry , Aldehydes/metabolism , Biocatalysis , Biotransformation , Molecular Structure , Phenylacetates/chemistry , Phenylethyl Alcohol/chemistry
6.
Nature ; 571(7764): 275-278, 2019 07.
Article in English | MEDLINE | ID: mdl-31181567

ABSTRACT

Recently developed DNA base editing methods enable the direct generation of desired point mutations in genomic DNA without generating any double-strand breaks1-3, but the issue of off-target edits has limited the application of these methods. Although several previous studies have evaluated off-target mutations in genomic DNA4-8, it is now clear that the deaminases that are integral to commonly used DNA base editors often bind to RNA9-13. For example, the cytosine deaminase APOBEC1-which is used in cytosine base editors (CBEs)-targets both DNA and RNA12, and the adenine deaminase TadA-which is used in adenine base editors (ABEs)-induces site-specific inosine formation on RNA9,11. However, any potential RNA mutations caused by DNA base editors have not been evaluated. Adeno-associated viruses are the most common delivery system for gene therapies that involve DNA editing; these viruses can sustain long-term gene expression in vivo, so the extent of potential RNA mutations induced by DNA base editors is of great concern14-16. Here we quantitatively evaluated RNA single nucleotide variations (SNVs) that were induced by CBEs or ABEs. Both the cytosine base editor BE3 and the adenine base editor ABE7.10 generated tens of thousands of off-target RNA SNVs. Subsequently, by engineering deaminases, we found that three CBE variants and one ABE variant showed a reduction in off-target RNA SNVs to the baseline while maintaining efficient DNA on-target activity. This study reveals a previously overlooked aspect of off-target effects in DNA editing and also demonstrates that such effects can be eliminated by engineering deaminases.


Subject(s)
DNA/genetics , Gene Editing/methods , Mutagenesis , Mutation , Nucleoside Deaminases/genetics , Protein Engineering , RNA/genetics , Adenine/metabolism , Aminohydrolases/genetics , Aminohydrolases/metabolism , Cytosine/metabolism , Cytosine Deaminase/genetics , Cytosine Deaminase/metabolism , HEK293 Cells , Humans , Nucleoside Deaminases/metabolism , Substrate Specificity , Transfection
7.
BMC Cancer ; 19(1): 197, 2019 Mar 04.
Article in English | MEDLINE | ID: mdl-30832616

ABSTRACT

BACKGROUND: The cytosine deaminase (CD)/5-fluorocytosine (5-FC) system is among the best explored enzyme/prodrug systems in the field of the suicide gene therapy. Recently, by the screening of the environmental metagenomic libraries we identified a novel isocytosine deaminase (ICD), termed Vcz, which is able of specifically converting a prodrug 5-fluoroisocytosine (5-FIC) into toxic drug 5-fluorouracil (5-FU). The aim of this study is to test the applicability of the ICD Vcz / 5-FIC pair as a potential suicide gene therapy tool. METHODS: Vcz-expressing human glioblastoma U87 and epithelial colorectal adenocarcinoma Caco-2 cells were treated with 5-FIC, and the Vcz-mediated cytotoxicity was evaluated by performing an MTT assay. In order to examine anti-tumor effects of the Vcz/5-FIC system in vivo, murine bone marrow-derived mesenchymal stem cells (MSC) were transduced with the Vcz-coding lentivirus and co-injected with 5-FIC or control reagents into subcutaneous GL261 tumors evoked in C57/BL6 mice. RESULTS: 5-FIC alone showed no significant toxic effects on U87 and Caco-2 cells at 100 µM concentration, whereas the number of cells of both cell lines that express Vcz cytosine deaminase gene decreased by approximately 60% in the presence of 5-FIC. The cytotoxic effects on cells were also induced by media collected from Vcz-expressing cells pre-treated with 5-FIC. The co-injection of the Vcz-transduced mesenchymal stem cells and 5-FIC have been shown to augment tumor necrosis and increase longevity of tumorized mice by 50% in comparison with control group animals. CONCLUSIONS: We have confirmed that the novel ICD Vcz together with the non-toxic prodrug 5-FIC has a potential of being a new enzyme/prodrug system for suicide gene therapy.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Flucytosine/analogs & derivatives , Fluorouracil/pharmacology , Genes, Transgenic, Suicide , Prodrugs/pharmacology , Adenocarcinoma , Animals , Antimetabolites, Antineoplastic/metabolism , Brain Neoplasms , Caco-2 Cells , Cell Line, Tumor , Cell Survival/drug effects , Colorectal Neoplasms , Cytosine/analogs & derivatives , Cytosine/metabolism , Cytosine Deaminase/genetics , Cytosine Deaminase/metabolism , Flucytosine/metabolism , Flucytosine/pharmacology , Fluorouracil/metabolism , Genetic Therapy , Genetic Vectors , Glioblastoma , Humans , Lentivirus , Mesenchymal Stem Cells , Mice , Nucleoside Deaminases/genetics , Nucleoside Deaminases/metabolism , Prodrugs/metabolism
8.
Plant Cell ; 31(3): 734-751, 2019 03.
Article in English | MEDLINE | ID: mdl-30787180

ABSTRACT

Plants can fully catabolize purine nucleotides. A firmly established central intermediate is the purine base xanthine. In the current widely accepted model of plant purine nucleotide catabolism, xanthine can be generated in various ways involving either inosine and hypoxanthine or guanosine and xanthosine as intermediates. In a comprehensive mutant analysis involving single and multiple mutants of urate oxidase, xanthine dehydrogenase, nucleoside hydrolases, guanosine deaminase, and hypoxanthine guanine phosphoribosyltransferase, we demonstrate that purine nucleotide catabolism in Arabidopsis (Arabidopsis thaliana) mainly generates xanthosine, but not inosine and hypoxanthine, and that xanthosine is derived from guanosine deamination and a second source, likely xanthosine monophosphate dephosphorylation. Nucleoside hydrolase 1 (NSH1) is known to be essential for xanthosine hydrolysis, but the in vivo function of a second cytosolic nucleoside hydrolase, NSH2, is unclear. We demonstrate that NSH1 activates NSH2 in vitro and in vivo, forming a complex with almost two orders of magnitude higher catalytic efficiency for xanthosine hydrolysis than observed for NSH1 alone. Remarkably, an inactive NSH1 point mutant can activate NSH2 in vivo, fully preventing purine nucleoside accumulation in nsh1 background. Our data lead to an altered model of purine nucleotide catabolism that includes an NSH heterocomplex as a central component.


Subject(s)
Adenosine Monophosphate/metabolism , Arabidopsis Proteins/metabolism , Arabidopsis/enzymology , Guanosine Monophosphate/metabolism , Ribonucleosides/metabolism , Arabidopsis/genetics , Arabidopsis Proteins/genetics , Mutation , N-Glycosyl Hydrolases/genetics , N-Glycosyl Hydrolases/metabolism , Nucleoside Deaminases/genetics , Nucleoside Deaminases/metabolism , Plants, Genetically Modified , Xanthines
9.
Scand J Immunol ; 89(5): e12760, 2019 May.
Article in English | MEDLINE | ID: mdl-30802996

ABSTRACT

A new and diverse range of somatic mutation signatures are observed in late-stage cancers, but the underlying reasons are not fully understood. We advance a "combinatorial association model" for deaminase binding domain (DBD) diversification to explain the generation of previously observed cancer-progression associated mutation signatures. We also propose that changes in the polarization of tumour-associated macrophages (TAMs) are accompanied by the expression of deaminases with a new and diverse range of DBDs, and thus accounting for the generation of new somatic mutation signatures. The mechanism proposed is molecularly reminiscent of combinatorial association of heavy (H) and light (L) protein chains following V(D)J recombination of immunoglobulin molecules (and similarly for protein chains in heterodimers α/ß and γ/δ of V(D)Js of T Cell Receptors) required for pathogen antigen recognition by B cells and T cells, respectively. We also discuss whether extracellular vesicles (EVs) emanating from tumour enhancing M2-polarized macrophages represent a likely source of the de novo deaminase DBDs. We conclude that M2-polarized macrophages extruding EVs loaded with deaminase proteins or deaminase-specific transcription/translation regulatory factors and like information may directly trigger deaminase diversification within cancer cells, and thus account for the many new somatic mutation signatures that are indicative of cancer progression. This hypothesis now has a plausible evidentiary base, and it is worth direct testing in future investigations. A long-term objective would be to identify molecular biomarkers predicting cancer progression (or metastatic disease) and to support the development of new drug targets before metastatic pathways are activated.


Subject(s)
Carcinogenesis/genetics , Macrophages/immunology , Models, Immunological , Mutation/genetics , Neoplasms/genetics , Recombination, Genetic , Th2 Cells/immunology , Animals , Cell Differentiation , Cell Movement , DNA Mutational Analysis , Extracellular Vesicles/metabolism , Humans , Lymphocyte Activation , Models, Theoretical , Nucleoside Deaminases/metabolism , Transcriptome
10.
Curr Opin Biotechnol ; 55: 74-80, 2019 02.
Article in English | MEDLINE | ID: mdl-30193161

ABSTRACT

The game-changing role of CRISPR/Cas for genome editing draw interest to programmable RNA-guided tools in general. Currently, we see a wave of papers pioneering the CRISPR/Cas system for RNA targeting, and applying them for site-directed RNA editing. Here, we exemplarily compare three recent RNA editing strategies that rely on three distinct RNA targeting mechanisms. We conclude that the CRISPR/Cas system seems not generally superior to other RNA targeting strategies in solving the most pressing problem in the RNA editing field, which is to obtain high efficiency in combination with high specificity. However, once achieved, RNA editing promises to complement or even outcompete DNA editing approaches in therapy, and also in some fields of basic research.


Subject(s)
Genetic Engineering , Nucleoside Deaminases/metabolism , RNA Editing , Adenosine/metabolism , Humans , Inosine/metabolism
11.
Nature ; 542(7642): 494-497, 2017 02 22.
Article in English | MEDLINE | ID: mdl-28230119

ABSTRACT

Nucleic acids undergo naturally occurring chemical modifications. Over 100 different modifications have been described and every position in the purine and pyrimidine bases can be modified; often the sugar is also modified. Despite recent progress, the mechanism for the biosynthesis of most modifications is not fully understood, owing, in part, to the difficulty associated with reconstituting enzyme activity in vitro. Whereas some modifications can be efficiently formed with purified components, others may require more intricate pathways. A model for modification interdependence, in which one modification is a prerequisite for another, potentially explains a major hindrance in reconstituting enzymatic activity in vitro. This model was prompted by the earlier discovery of tRNA cytosine-to-uridine editing in eukaryotes, a reaction that has not been recapitulated in vitro and the mechanism of which remains unknown. Here we show that cytosine 32 in the anticodon loop of Trypanosoma brucei tRNAThr is methylated to 3-methylcytosine (m3C) as a pre-requisite for C-to-U deamination. Formation of m3C in vitro requires the presence of both the T. brucei m3C methyltransferase TRM140 and the deaminase ADAT2/3. Once formed, m3C is deaminated to 3-methyluridine (m3U) by the same set of enzymes. ADAT2/3 is a highly mutagenic enzyme, but we also show that when co-expressed with the methyltransferase its mutagenicity is kept in check. This helps to explain how T. brucei escapes 'wholesale deamination' of its genome while harbouring both enzymes in the nucleus. This observation has implications for the control of another mutagenic deaminase, human AID, and provides a rationale for its regulation.


Subject(s)
Methyltransferases/metabolism , Nucleoside Deaminases/metabolism , RNA Editing , RNA, Transfer, Thr/chemistry , RNA, Transfer, Thr/metabolism , Trypanosoma brucei brucei/enzymology , Trypanosoma brucei brucei/genetics , Anticodon/metabolism , Base Sequence , Cytosine/analogs & derivatives , Cytosine/metabolism , Deamination , Methylation , RNA, Transfer, Thr/genetics , Uridine/metabolism
12.
J Bacteriol ; 196(11): 1950-7, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24633874

ABSTRACT

We show here that deoxycytidine deaminase (DCD)-deficient mutants of Escherichia coli are hypersensitive to killing by exogenous cytidine, adenosine, or guanosine, whereas wild-type cells are not. This hypersensitivity is reversed by exogenous thymidine. The mechanism likely involves the allosteric regulation of ribonucleotide reductase and severe limitations of the dTTP pools, resulting in thymineless death, the phenomenon of cell death due to thymidine starvation. We also report here that DCD-deficient mutants of E. coli are more sensitive to a series of different antibiotics, including vancomycin, and we show synergistic killing with the combination of vancomycin and cytidine. One possibility is that a very low, subinhibitory concentration of vancomycin enters Gram-negative cells and that this concentration is potentiated by chromosomal lesions resulting from the thymineless state. A second possibility is that the metabolic imbalance resulting from DCD deficiency affects the assembly of the outer membrane, which normally presents a barrier to drugs such as vancomycin. We consider these findings with regard to ideas of rendering Gram-negative bacteria sensitive to drugs such as vancomycin.


Subject(s)
Anti-Bacterial Agents/pharmacology , Escherichia coli/enzymology , Gene Expression Regulation, Bacterial/physiology , Gene Expression Regulation, Enzymologic/physiology , Nucleoside Deaminases/metabolism , Vancomycin/pharmacology , Adenosine/pharmacology , Cytidine/pharmacology , Cytidine Deaminase , Drug Resistance, Bacterial , Escherichia coli/drug effects , Gene Deletion , Guanosine/pharmacology , Nucleoside Deaminases/genetics
13.
J Agric Food Chem ; 62(8): 1796-801, 2014 Feb 26.
Article in English | MEDLINE | ID: mdl-24475736

ABSTRACT

Brassica oleracea deoxycytidine deaminase (BoDCD), a deoxycytidine deaminase (DCD, EC 3.5.4.14) enzyme, is known to play an important role in the Trichoderma harzianum ETS 323 mediated resistance mechanism in young leaves of B. oleracea var. capitata during Rhizoctonia solani infection. BoDCD potentially neutralizes cytotoxic products of host lipoxygenase activity, and thereby BoDCD restricts the hypersensitivity-related programmed cell death induced in plants during the initial stages of infection. To determine the biochemical characteristics and to partially elucidate the designated functional properties of BoDCD, the enzyme was cloned into an Escherichia coli expression system, and its potential to neutralize the toxic analogues of 2'-deoxycytidine (dC) was examined. BoDCD transformants of E. coli cells were found to be resistant to 2'-deoxycytidine analogues at all of the concentrations tested. The BoDCD enzyme was also overexpressed as a histidine-tagged protein and purified using nickel chelating affinity chromatography. The molecular weight of BoDCD was determined to be 20.8 kDa as visualized by SDS-PAGE. The substrate specificity and other kinetic properties show that BoDCD is more active in neutralizing cytotoxic cytosine ß-d-arabinofuranoside than in deaminating 2'-deoxycytinde to 2'-deoxyuridine in nucleic acids or in metabolizing cytidine to uridine. The optimal temperature and pH of the enzyme were 27 °C and 7.5. The Km and Vmax values of BoDCD were, respectively, 91.3 µM and 1.475 mM for its natural substrate 2'-deoxycytidine and 63 µM and 2.072 mM for cytosine ß-d-arabinofuranoside. The phenomenon of neutralization of cytotoxic dC analogues by BoDCD is discussed in detail on the basis of enzyme biochemical properties.


Subject(s)
Brassica/enzymology , Nucleoside Deaminases/chemistry , Plant Proteins/chemistry , Brassica/chemistry , Brassica/genetics , Cytidine/metabolism , Cytidine Deaminase , Cytosine/metabolism , Kinetics , Molecular Weight , Nucleoside Deaminases/genetics , Nucleoside Deaminases/metabolism , Plant Proteins/genetics , Plant Proteins/metabolism , Substrate Specificity , Uridine/metabolism
14.
RNA Biol ; 11(12): 1608-18, 2014.
Article in English | MEDLINE | ID: mdl-25625329

ABSTRACT

tRNA modifications are crucial for efficient and accurate protein translation, with defects often linked to disease. There are 7 cytoplasmic tRNA modifications in the yeast Saccharomyces cerevisiae that are formed by an enzyme consisting of a catalytic subunit and an auxiliary protein, 5 of which require only a single subunit in bacteria, and 2 of which are not found in bacteria. These enzymes include the deaminase Tad2-Tad3, and the methyltransferases Trm6-Trm61, Trm8-Trm82, Trm7-Trm732, and Trm7-Trm734, Trm9-Trm112, and Trm11-Trm112. We describe the occurrence and biological role of each modification, evidence for a required partner protein in S. cerevisiae and other eukaryotes, evidence for a single subunit in bacteria, and evidence for the role of the non-catalytic binding partner. Although it is unclear why these eukaryotic enzymes require partner proteins, studies of some 2-subunit modification enzymes suggest that the partner proteins help expand substrate range or allow integration of cellular activities.


Subject(s)
Nucleoside Deaminases/metabolism , Protein Subunits/metabolism , RNA Processing, Post-Transcriptional , RNA, Transfer/metabolism , Saccharomyces cerevisiae Proteins/metabolism , tRNA Methyltransferases/metabolism , Binding Sites , Escherichia coli/enzymology , Escherichia coli/genetics , Models, Molecular , Nucleic Acid Conformation , Nucleoside Deaminases/genetics , Protein Binding , Protein Subunits/genetics , RNA, Transfer/chemistry , RNA, Transfer/genetics , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , tRNA Methyltransferases/genetics
15.
Plant Cell ; 25(10): 4101-9, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24130159

ABSTRACT

Purine nucleotide catabolism is common to most organisms and involves a guanine deaminase to convert guanine to xanthine in animals, invertebrates, and microorganisms. Using metabolomic analysis of mutants, we demonstrate that Arabidopsis thaliana uses an alternative catabolic route employing a highly specific guanosine deaminase (GSDA) not reported from any organism so far. The enzyme is ubiquitously expressed and deaminates exclusively guanosine and 2'-deoxyguanosine but no other aminated purines, pyrimidines, or pterines. GSDA belongs to the cytidine/deoxycytidylate deaminase family of proteins together with a deaminase involved in riboflavin biosynthesis, the chloroplastic tRNA adenosine deaminase Arg and a predicted tRNA-specific adenosine deaminase 2 in A. thaliana. GSDA is conserved in plants, including the moss Physcomitrella patens, but is absent in the algae and outside the plant kingdom. Our data show that xanthosine is exclusively generated through the deamination of guanosine by GSDA in A. thaliana, excluding other possible sources like the dephosphorylation of xanthosine monophosphate. Like the nucleoside hydrolases NUCLEOSIDE HYDROLASE1 (NSH1) and NSH2, GSDA is located in the cytosol, indicating that GMP catabolism to xanthine proceeds in a mostly cytosolic pathway via guanosine and xanthosine. Possible implications for the biosynthetic route of purine alkaloids (caffeine and theobromine) and ureides in other plants are discussed.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/enzymology , Nucleoside Deaminases/metabolism , Purines/metabolism , Ribonucleosides/biosynthesis , Arabidopsis/genetics , Arabidopsis Proteins/genetics , Cloning, Molecular , DNA, Bacterial/genetics , Metabolome , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation , Nucleoside Deaminases/genetics , Plants, Genetically Modified/enzymology , Plants, Genetically Modified/genetics , Xanthines
16.
J Am Chem Soc ; 135(37): 13927-33, 2013 Sep 18.
Article in English | MEDLINE | ID: mdl-23968233

ABSTRACT

A substantial challenge for genomic enzymology is the reliable annotation for proteins of unknown function. Described here is an interrogation of uncharacterized enzymes from the amidohydrolase superfamily using a structure-guided approach that integrates bioinformatics, computational biology, and molecular enzymology. Previously, Tm0936 from Thermotoga maritima was shown to catalyze the deamination of S-adenosylhomocysteine (SAH) to S-inosylhomocysteine (SIH). Homologues of Tm0936 homologues were identified, and substrate profiles were proposed by docking metabolites to modeled enzyme structures. These enzymes were predicted to deaminate analogues of adenosine including SAH, 5'-methylthioadenosine (MTA), adenosine (Ado), and 5'-deoxyadenosine (5'-dAdo). Fifteen of these proteins were purified to homogeneity, and the three-dimensional structures of three proteins were determined by X-ray diffraction methods. Enzyme assays supported the structure-based predictions and identified subgroups of enzymes with the capacity to deaminate various combinations of the adenosine analogues, including the first enzyme (Dvu1825) capable of deaminating 5'-dAdo. One subgroup of proteins, exemplified by Moth1224 from Moorella thermoacetica, deaminates guanine to xanthine, and another subgroup, exemplified by Avi5431 from Agrobacterium vitis S4, deaminates two oxidatively damaged forms of adenine: 2-oxoadenine and 8-oxoadenine. The sequence and structural basis of the observed substrate specificities were proposed, and the substrate profiles for 834 protein sequences were provisionally annotated. The results highlight the power of a multidisciplinary approach for annotating enzymes of unknown function.


Subject(s)
Nucleoside Deaminases/chemistry , Catalytic Domain , Crystallography, X-Ray , Enzyme Assays , Kinetics , Models, Molecular , Molecular Structure , Nucleoside Deaminases/metabolism , Structure-Activity Relationship , Substrate Specificity
17.
Biochemistry ; 52(37): 6525-36, 2013 Sep 17.
Article in English | MEDLINE | ID: mdl-23972005

ABSTRACT

Proteins of unknown function belonging to cog1816 and cog0402 were characterized. Sav2595 from Steptomyces avermitilis MA-4680, Acel0264 from Acidothermus cellulolyticus 11B, Nis0429 from Nitratiruptor sp. SB155-2 and Dr0824 from Deinococcus radiodurans R1 were cloned, purified, and their substrate profiles determined. These enzymes were previously incorrectly annotated as adenosine deaminases or chlorohydrolases. It was shown here that these enzymes actually deaminate 6-aminodeoxyfutalosine. The deamination of 6-aminodeoxyfutalosine is part of an alternative menaquinone biosynthetic pathway that involves the formation of futalosine. 6-Aminodeoxyfutalosine is deaminated by these enzymes with catalytic efficiencies greater than 10(5) M(-1) s(-1), Km values of 0.9-6.0 µM, and kcat values of 1.2-8.6 s(-1). Adenosine, 2'-deoxyadenosine, thiomethyladenosine, and S-adenosylhomocysteine are deaminated at least an order of magnitude slower than 6-aminodeoxyfutalosine. The crystal structure of Nis0429 was determined and the substrate, 6-aminodeoxyfutalosine, was positioned in the active site on the basis of the presence of adventitiously bound benzoic acid. In this model, Ser-145 interacts with the carboxylate moiety of the substrate. The structure of Dr0824 was also determined, but a collapsed active site pocket prevented docking of substrates. A computational model of Sav2595 was built on the basis of the crystal structure of adenosine deaminase and substrates were docked. The model predicted a conserved arginine after ß-strand 1 to be partially responsible for the substrate specificity of Sav2595.


Subject(s)
Nucleoside Deaminases/metabolism , Purine Nucleosides/metabolism , Vitamin K 2/metabolism , Actinomycetales/enzymology , Catalytic Domain , Crystallography, X-Ray , Deamination , Deinococcus/enzymology , Epsilonproteobacteria/enzymology , Epsilonproteobacteria/genetics , Kinetics , Models, Molecular , Molecular Docking Simulation , Nucleoside Deaminases/genetics , Streptomyces/enzymology , Streptomyces/genetics , Substrate Specificity
18.
Anticancer Res ; 33(5): 1899-912, 2013 May.
Article in English | MEDLINE | ID: mdl-23645737

ABSTRACT

BACKGROUND/AIM: GS 9219 is a double prodrug of antiproliferative nucleotide analog 9-(2-Phosphonylmethoxyethyl)guanine (PMEG), with potent in vivo efficacy against various hematological malignancies. This study investigates the role of adenosine deaminase-like (ADAL) protein in the intracellular activation of GS-9219. MATERIALS AND METHODS: A cell line resistant to 9-(2-Phosphonylmethoxyethyl)-N(6)-cyclopropyl-2,6-diaminopurine (cPrPMEDAP), an intermediate metabolite of GS-9219, was generated and characterized. RESULTS: The resistant cell line was cross-resistant to cPrPMEDAP and GS-9219, due to a defect in the deamination of cPrPMEDAP to PMEG. Mutations in the ADAL gene (H286R and S180N) were identified in the resistant cells that adversely-affected its enzymatic activity. Introduction of the wild-type ADAL gene re-sensitized resistant cells to both cPrPMEDAP and GS-9219. CONCLUSION: The ADAL protein plays an essential role in the intracellular activation of GS-9219 by catalyzing the deamination of cPrPMEDAP metabolite to PMEG. Mutations affecting the activity of ADAL confer resistance to both GS-9219 and its metabolite cPrPMEDAP.


Subject(s)
Adenine/analogs & derivatives , Alanine/analogs & derivatives , Drug Resistance, Neoplasm/genetics , Mutation/genetics , Nucleoside Deaminases/genetics , Purines/pharmacology , Uterine Cervical Neoplasms/genetics , Adenine/pharmacology , Alanine/pharmacology , Amino Acid Sequence , Antineoplastic Agents/pharmacology , Blotting, Western , Female , Humans , Molecular Sequence Data , Nucleoside Deaminases/chemistry , Nucleoside Deaminases/metabolism , Prodrugs/pharmacology , Protein Conformation , Sequence Homology, Amino Acid , Tumor Cells, Cultured , Uterine Cervical Neoplasms/drug therapy
19.
Biochemistry ; 51(45): 9094-103, 2012 Nov 13.
Article in English | MEDLINE | ID: mdl-23050701

ABSTRACT

Pseudomonas aeruginosa possesses an unusual pathway for 5'-methylthioadenosine (MTA) metabolism involving deamination to 5'-methylthioinosine (MTI) followed by N-ribosyl phosphorolysis to hypoxanthine and 5-methylthio-α-d-ribose 1-phosphate. The specific MTI phosphorylase of P. aeruginosa has been reported [Guan, R., Ho, M. C., Almo, S. C., and Schramm, V. L. (2011) Biochemistry 50, 1247-1254], and here we characterize MTA deaminase from P. aeruginosa (PaMTADA). Genomic analysis indicated the PA3170 locus to be a candidate for MTA deaminase (MTADA). Protein encoded by PA3170 was expressed and shown to deaminate MTA with 40-fold greater catalytic efficiency for MTA than for adenosine. The k(cat)/K(m) value of 1.6 × 10(7) M(-1) s(-1) for MTA is the highest catalytic efficiency known for an MTA deaminase. 5'-Methylthiocoformycin (MTCF) is a 4.8 pM transition state analogue for PaMTADA but causes no significant inhibition of human adenosine deaminase or MTA phosphorylase. MTCF is permeable to P. aeruginosa and exhibits an IC(50) of 3 nM on cellular PaMTADA activity. PaMTADA is the only activity in P. aeruginosa extracts to act on MTA. MTA and 5-methylthio-α-d-ribose are involved in quorum sensing pathways; thus, PaMTADA is a potential target for quorum sensing. The crystal structure of PaMTADA in complex with MTCF shows the transition state mimic 8(R)-hydroxyl group in contact with a catalytic site Zn(2+), the 5'-methylthio group in a hydrophobic pocket, and the transition state mimic of the diazepine ring in contact with a catalytic site Glu.


Subject(s)
Deoxyadenosines/metabolism , Nucleoside Deaminases/metabolism , Pseudomonas aeruginosa/enzymology , Quorum Sensing , Thionucleosides/metabolism , Adenosine Deaminase/metabolism , Amino Acid Sequence , Coformycin/analogs & derivatives , Coformycin/pharmacology , Crystallography, X-Ray , Humans , Ligases , Methylthioinosine/metabolism , Models, Molecular , Molecular Sequence Data , Nucleoside Deaminases/antagonists & inhibitors , Sequence Alignment , Substrate Specificity
20.
Angew Chem Int Ed Engl ; 51(44): 11166-9, 2012 Oct 29.
Article in English | MEDLINE | ID: mdl-23038402

ABSTRACT

Checking for mistakes: By conjugating a catalytic domain with a guide RNA, deamination activity can be harnessed to repair a specific codon on mRNA. This method can be used for the highly selective repair of point mutations in mRNA by site-selective editing.


Subject(s)
Nucleoside Deaminases/chemistry , Point Mutation , RNA/chemistry , Adenosine Deaminase/chemistry , Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism , Green Fluorescent Proteins/genetics , Humans , Models, Molecular , Molecular Structure , Nucleoside Deaminases/genetics , Nucleoside Deaminases/metabolism , RNA/genetics , RNA/metabolism , RNA, Messenger/genetics , RNA-Binding Proteins , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...