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










Publication year range
1.
J Biol Chem ; 300(7): 107461, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38876299

ABSTRACT

Theta-mediated end joining (TMEJ) is critical for survival of cancer cells when other DNA double-stranded break repair pathways are impaired. Human DNA polymerase theta (Pol θ) can extend ssDNA oligonucleotides, but little is known about preferred substrates and mechanism. We show that Pol θ can extend both ssDNA and RNA substrates by unimolecular stem-loop synthesis initiated by only two 3' terminal base pairs. Given sufficient time, Pol θ uses alternative pairing configurations that greatly expand the repertoire of sequence outcomes. Further primer-template adjustments yield low-fidelity outcomes when the nucleotide pool is imbalanced. Unimolecular stem-loop synthesis competes with bimolecular end joining, even when a longer terminal microhomology for end joining is available. Both reactions are partially suppressed by the ssDNA-binding protein replication protein A. Protein-primer grasp residues that are specific to Pol θ are needed for rapid stem-loop synthesis. The ability to perform stem-loop synthesis from a minimally paired primer is rare among human DNA polymerases, but we show that human DNA polymerases Pol η and Pol λ can catalyze related reactions. Using purified human Pol θ, we reconstituted in vitro TMEJ incorporating an insertion arising from a stem-loop extension. These activities may help explain TMEJ repair events that include inverted repeat sequences.

2.
Front Immunol ; 14: 1229724, 2023.
Article in English | MEDLINE | ID: mdl-37662930

ABSTRACT

Genogroup II (GII) noroviruses are a major cause of diarrheal disease burden in children in both high- and low-income countries. GII.17 noroviruses are composed of distinct genetic clusters (I, II, IIIa, and IIIb) and have shown potential for replacing historically more prevalent GII.4 strains, but the serological basis for GII.17 antigenic diversity has not been studied in children. Utilizing samples from a birth cohort, we investigated antibody and B-cell responses to GII.17 cluster variants in confirmed GII.17 infections in young children as well as demonstrated that the distinct genetic clusters co-circulate. Polyclonal serum antibodies bound multiple clusters but showed cluster-specific blockade activity in a surrogate virus neutralization assay. Antibodies secreted by immortalized memory B cells (MBCs) from an infant GII.17 case were highly specific to GII.17 and exhibited blockade activity against this genotype. We isolated an MBC-derived GII.17-specific Immunoglobulin A (IgA) monoclonal antibody called NVA.1 that potently and selectively blocked GII.17 cluster IIIb and recognized an epitope targeted in serum from cluster IIIb-infected children. These data indicate that multiple antigenically distinct GII.17 variants co-circulate in young children, suggesting retention of cluster diversity alongside potential for immune escape given the existence of antibody-defined cluster-specific epitopes elicited during infection.


Subject(s)
B-Lymphocytes , Norovirus , Child , Infant , Humans , Child, Preschool , Antibodies, Monoclonal , Memory B Cells , Immunoglobulin A , Paraproteins , Epitopes , Genotype , Norovirus/genetics
3.
Structure ; 29(1): 29-42.e4, 2021 01 07.
Article in English | MEDLINE | ID: mdl-32846144

ABSTRACT

Oxidative damage on DNA arising from both endogenous and exogenous sources can result in base modifications that promote errors in replication as well as generating sites of base loss (abasic sites) that present unique challenges to maintaining genomic integrity. These lesions are excised by DNA glycosylases in the first step of the base excision repair pathway. Here we present the first crystal structure of a NEIL2 glycosylase, an enzyme active on cytosine oxidation products and abasic sites. The structure reveals an unusual "open" conformation not seen in NEIL1 or NEIL3 orthologs. NEIL2 is predicted to adopt a "closed" conformation when bound to its substrate. Combined crystallographic and solution-scattering studies show the enzyme to be conformationally dynamic in a manner distinct among the NEIL glycosylases and provide insight into the unique substrate preference of this enzyme. In addition, we characterized three cancer variants of human NEIL2, namely S140N, G230W, and G303R.


Subject(s)
DNA Glycosylases/chemistry , DNA-(Apurinic or Apyrimidinic Site) Lyase/chemistry , Binding Sites , DNA/chemistry , DNA/metabolism , DNA Glycosylases/metabolism , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Humans , Molecular Dynamics Simulation , Protein Binding
4.
J Am Chem Soc ; 142(31): 13283-13287, 2020 08 05.
Article in English | MEDLINE | ID: mdl-32664726

ABSTRACT

MutY glycosylase excises adenines misincorporated opposite the oxidatively damaged lesion, 8-oxo-7,8-dihydroguanine (OG), to initiate base excision repair and prevent G to T transversion mutations. Successful repair requires MutY recognition of the OG:A mispair amidst highly abundant and structurally similar undamaged DNA base pairs. Herein we use a combination of in vitro and bacterial cell repair assays with single-molecule fluorescence microscopy to demonstrate that both a C-terminal domain histidine residue and the 2-amino group of OG base are critical for MutY detection of OG:A sites. These studies are the first to directly link deficiencies in MutY lesion detection with incomplete cellular repair. These results suggest that defects in lesion detection of human MutY (MUTYH) variants may prove predictive of early-onset colorectal cancer known an MUTYH-associated polyposis. Furthermore, unveiling these specific molecular determinants for repair makes it possible to envision new MUTYH-specific cancer therapies.


Subject(s)
DNA Glycosylases/metabolism , Guanine/analogs & derivatives , Histidine/metabolism , DNA Glycosylases/chemistry , Guanine/analysis , Guanine/metabolism , Humans , Microscopy, Fluorescence , Models, Molecular
5.
J Cell Biochem ; 120(8): 13783-13791, 2019 08.
Article in English | MEDLINE | ID: mdl-30938854

ABSTRACT

The chimeric DnaJ-PKAc enzymeresulting from an approximately 400-kb deletion of chromosome 19 is a primary contributor to the oncogenic transformation that occurs in fibrolamellar hepatocellular carcinoma, also called fibrolamellar carcinoma (FLC). This oncogenic deletion juxtaposes exon 1 of the DNAJB1 heat shock protein gene with exon 2 of the PRKACA gene encoding the protein kinase A catalytic subunit, resulting in DnaJ-PKAc fusion under the transcriptional control of the DNAJB1 promoter. The expression of DnaJ-PKAc is approximately 10 times that of wild-type (wt) PKAc catalytic subunits, causing elevated and dysregulated kinase activity that contributes to oncogenic transformation. In normal cells, PKAc activity is regulated by a group of endogenous proteins, termed protein kinase inhibitors (PKI) that competitively inhibit PKAc and assist with the nuclear export of the enzyme. Currently, it is scarcely known whether interactions with PKI are perturbed in DnaJ-PKAc. In this report, we survey existing data sets to assess the expression levels of the various PKI isoforms that exist in humans to identify those that are candidates to encounter DnaJ-PKAc in both normal liver and FLC tumors. We then compare inhibition profiles of wtPKAc and DnaJ-PKAc against PKI and demonstrate that extensive structural homology in the active site clefts of the two enzymes confers similar kinase activities and inhibition by full-length PKI and PKI-derived peptides.


Subject(s)
Cyclic AMP-Dependent Protein Kinase Catalytic Subunits , HSP40 Heat-Shock Proteins , Oncogene Proteins, Fusion , Peptides/chemistry , Protein Kinase Inhibitors/chemistry , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/chemistry , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/genetics , HSP40 Heat-Shock Proteins/antagonists & inhibitors , HSP40 Heat-Shock Proteins/chemistry , HSP40 Heat-Shock Proteins/genetics , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Oncogene Proteins, Fusion/antagonists & inhibitors , Oncogene Proteins, Fusion/chemistry , Oncogene Proteins, Fusion/genetics
6.
Nucleic Acids Res ; 47(6): 3058-3071, 2019 04 08.
Article in English | MEDLINE | ID: mdl-30698731

ABSTRACT

Proper repair of oxidatively damaged DNA bases is essential to maintain genome stability. 8-Oxoguanine (7,8-dihydro-8-oxoguanine, 8-oxoG) is a dangerous DNA lesion because it can mispair with adenine (A) during replication resulting in guanine to thymine transversion mutations. MUTYH DNA glycosylase is responsible for recognizing and removing the adenine from 8-oxoG:adenine (8-oxoG:A) sites. Biallelic mutations in the MUTYH gene predispose individuals to MUTYH-associated polyposis (MAP), and the most commonly observed mutation in some MAP populations is Y165C. Tyr165 is a 'wedge' residue that intercalates into the DNA duplex in the lesion bound state. Here, we utilize single molecule fluorescence microscopy to visualize the real-time search behavior of Escherichia coli and Mus musculus MUTYH WT and wedge variant orthologs on DNA tightropes that contain 8-oxoG:A, 8-oxoG:cytosine, or apurinic product analog sites. We observe that MUTYH WT is able to efficiently find 8-oxoG:A damage and form highly stable bound complexes. In contrast, MUTYH Y150C shows decreased binding lifetimes on undamaged DNA and fails to form a stable lesion recognition complex at damage sites. These findings suggest that MUTYH does not rely upon the wedge residue for damage site recognition, but this residue stabilizes the lesion recognition complex.


Subject(s)
Adenomatous Polyposis Coli/genetics , Colorectal Neoplasms/genetics , DNA Damage/genetics , DNA Glycosylases/genetics , Adenine/metabolism , Adenomatous Polyposis Coli/pathology , Animals , Colorectal Neoplasms/pathology , Escherichia coli/genetics , Genomic Instability/genetics , Guanine/analogs & derivatives , Guanine/chemistry , Humans , Mice , Mutation , Oxidative Stress/genetics
7.
DNA Repair (Amst) ; 60: 64-76, 2017 12.
Article in English | MEDLINE | ID: mdl-29100040

ABSTRACT

The human RAD51 recombinase possesses DNA pairing and strand exchange activities that are essential for the error-free, homology-directed repair of DNA double-strand breaks. The recombination activities of RAD51 are activated upon its assembly into presynaptic filaments on single-stranded DNA at resected DSB ends. Defects in filament assembly caused by mutations in RAD51 or its regulators such as BRCA2 are associated with human cancer. Here we describe two novel RAD51 missense variants located in the multimerization/BRCA2 binding region of RAD51. F86L is a breast tumor-derived somatic variant that affects the interface between adjacent RAD51 protomers in the presynaptic filament. E258A is a germline variant that maps to the interface region between the N-terminal and RecA homology domains of RAD51. Both variants exhibit abnormal biochemistry including altered DNA strand exchange activity. Both variants inhibit the DNA strand exchange activity of wild-type RAD51, suggesting a mechanism for negative dominance. The inhibitory effect of F86L on wild-type RAD51 is surprising since F86L alone exhibits robust DNA strand exchange activity. Our findings indicate that even DNA strand exchange-proficient variants can have negative functional interactions with wild-type RAD51. Thus heterozygous F86L or E258 mutations in RAD51 could promote genomic instability, and thereby contribute to tumor progression.


Subject(s)
BRCA2 Protein/metabolism , Mutation, Missense , Protein Interaction Domains and Motifs , Rad51 Recombinase/metabolism , Recombinational DNA Repair , Amino Acid Sequence , DNA/metabolism , DNA Breaks, Double-Stranded , Genetic Predisposition to Disease , Humans , Kinetics , Models, Molecular , Neoplasms/genetics , Neoplasms/metabolism , Protein Binding , Protein Multimerization , Rad51 Recombinase/chemistry , Rad51 Recombinase/genetics , Sequence Alignment
8.
ACS Chem Biol ; 12(6): 1584-1592, 2017 06 16.
Article in English | MEDLINE | ID: mdl-28459528

ABSTRACT

DNA polymerase θ (Pol θ) is implicated in various cellular processes including double-strand break repair and apurinic/apyrimidinic site bypass. Because Pol θ expression correlates with poor cancer prognosis, the ability of Pol θ to bypass the C4'-oxidized abasic site (C4-AP) and 2-deoxyribonolactone (L), which are generated by cytotoxic agents, is of interest. Translesion synthesis and subsequent extension by Pol θ past C4-AP or L and an abasic site (AP) or its tetrahydrofuran analogue (F) was examined. Pol θ conducts translesion synthesis on templates containing AP and F with similar efficiencies and follows the "A-rule," inserting nucleotides in the order A > G > T. Translesion synthesis on templates containing C4-AP and L is less efficient than AP and F, and the preference for A insertion is reduced for L and absent for C4-AP. Extension past all abasic lesions (AP, F, C4-AP, and L) was significantly less efficient than translesion synthesis and yielded deletions caused by the base one or two nucleotides downstream from the lesion being used as a template, with the latter being favored. These results suggest that bypass of abasic lesions by Pol θ is highly mutagenic.


Subject(s)
DNA Polymerase beta/physiology , Mutagenesis , Animals , Base Sequence , DNA Damage , DNA Polymerase beta/metabolism , DNA Repair , Humans , Nucleotides/metabolism , Oxidation-Reduction , Sequence Deletion , Sugar Acids , Templates, Genetic
9.
DNA Repair (Amst) ; 42: 44-55, 2016 06.
Article in English | MEDLINE | ID: mdl-27153211

ABSTRACT

In human cells, error-free repair of DNA double-strand breaks requires the DNA pairing and strand exchange activities of RAD51 recombinase. Activation of RAD51 recombination activities requires the assembly of RAD51 presynaptic filaments on the single-stranded DNA that forms at resected DSB ends. Mutations in proteins that control presynaptic filament assembly, such as BRCA2, and in RAD51 itself, are associated with human breast cancer. Here we describe the properties of two mutations in RAD51 protein that derive from human lung and kidney tumors, respectively. Sequence variants Q268P and Q272L both map to the DNA binding loop 2 (L2) region of RAD51, a motif that is involved in DNA binding and in the allosteric activation of ATP hydrolysis and DNA strand exchange activities. Both mutations alter the thermal stability, DNA binding, and ATPase properties of RAD51, however both variants retain intrinsic DNA strand exchange activity towards oligonucleotide substrates under optimized conditions. In contrast, both Q268P and Q272L variants exhibit drastically reduced DNA strand exchange activity in reaction mixtures containing long homologous ssDNA and dsDNA substrates and human RPA protein. Mixtures of wild-type and variant proteins also exhibit reduced DNA strand exchange activity, suggesting that heterozygous mutations could negatively affect DNA recombination and repair processes in vivo. Together, the findings of this study suggest that hypomorphic missense mutations in RAD51 protein could be drivers of genomic instability in cancer cells, and thereby contribute to the etiology of metastatic disease.


Subject(s)
DNA/genetics , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mutation, Missense , Rad51 Recombinase/genetics , DNA/metabolism , Female , Humans , Middle Aged , Models, Molecular , Protein Conformation , Protein Stability , Rad51 Recombinase/chemistry , Rad51 Recombinase/metabolism , Temperature
11.
Nucleic Acids Res ; 43(8): 4039-54, 2015 Apr 30.
Article in English | MEDLINE | ID: mdl-25813041

ABSTRACT

G-quadruplex is a four-stranded G-rich DNA structure that is highly susceptible to oxidation. Despite the important roles that G-quadruplexes play in telomere biology and gene transcription, neither the impact of guanine lesions on the stability of quadruplexes nor their repair are well understood. Here, we show that the oxidized guanine lesions 8-oxo-7,8-dihydroguanine (8-oxoG), guanidinohydantoin (Gh) and spiroiminodihydantoin (Sp) reduce the thermostability and alter the folding of telomeric quadruplexes in a location-dependent manner. Also, the NEIL1 and NEIL3 DNA glycosylases can remove hydantoin lesions but none of the glycosylases, including OGG1, are able to remove 8-oxoG from telomeric quadruplexes. Interestingly, a hydantoin lesion at the site most prone to oxidation in quadruplex DNA is not efficiently removed by NEIL1 or NEIL3. However, NEIL1, NEIL2 and NEIL3 remove hydantoins from telomeric quadruplexes formed by five TTAGGG repeats much more rapidly than the commonly studied four-repeat quadruplex structures. We also show that APE1 cleaves furan in selected positions in Na(+)-coordinated telomeric quadruplexes. In promoter G-quadruplex DNA, the NEIL glycosylases primarily remove Gh from Na(+)-coordinated antiparallel quadruplexes but not K(+)-coordinated parallel quadruplexes containing VEGF or c-MYC promoter sequences. Thus, the NEIL DNA glycosylases may be involved in both telomere maintenance and in gene regulation.


Subject(s)
DNA Glycosylases/metabolism , G-Quadruplexes , Guanine/metabolism , N-Glycosyl Hydrolases/metabolism , Promoter Regions, Genetic , Telomere/metabolism , DNA/chemistry , DNA/metabolism , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Furans/metabolism , Guanidines/metabolism , Guanine/analogs & derivatives , Guanosine/analogs & derivatives , Guanosine/metabolism , Humans , Hydantoins/metabolism , Oxidation-Reduction , Potassium/chemistry , Sodium/chemistry , Spiro Compounds/metabolism , Telomere/chemistry
12.
Nat Struct Mol Biol ; 22(4): 304-11, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25775267

ABSTRACT

DNA polymerase θ protects against genomic instability via an alternative end-joining repair pathway for DNA double-strand breaks. Polymerase θ is overexpressed in breast, lung and oral cancers, and reduction of its activity in mammalian cells increases sensitivity to double-strand break-inducing agents, including ionizing radiation. Reported here are crystal structures of the C-terminal polymerase domain from human polymerase θ, illustrating two potential modes of dimerization. One structure depicts insertion of ddATP opposite an abasic-site analog during translesion DNA synthesis. The second structure describes a cognate ddGTP complex. Polymerase θ uses a specialized thumb subdomain to establish unique upstream contacts to the primer DNA strand, including an interaction with the 3'-terminal phosphate from one of five distinctive insertion loops. These observations demonstrate how polymerase θ grasps the primer to bypass DNA lesions or extend poorly annealed DNA termini to mediate end-joining.


Subject(s)
DNA-Directed DNA Polymerase/physiology , Amino Acid Sequence , Crystallography, X-Ray , DNA-Directed DNA Polymerase/chemistry , Dimerization , Models, Molecular , Molecular Sequence Data , Protein Structure, Tertiary , Sequence Alignment , DNA Polymerase theta
13.
Nucleic Acids Res ; 43(2): 1098-111, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25539919

ABSTRACT

Human RAD51 protein catalyzes DNA pairing and strand exchange reactions that are central to homologous recombination and homology-directed DNA repair. Successful recombination/repair requires the formation of a presynaptic filament of RAD51 on ssDNA. Mutations in BRCA2 and other proteins that control RAD51 activity are associated with human cancer. Here we describe a set of mutations associated with human breast tumors that occur in a common structural motif of RAD51. Tumor-associated D149N, R150Q and G151D mutations map to a Schellman loop motif located on the surface of the RecA homology domain of RAD51. All three variants are proficient in DNA strand exchange, but G151D is slightly more sensitive to salt than wild-type (WT). Both G151D and R150Q exhibit markedly lower catalytic efficiency for adenosine triphosphate hydrolysis compared to WT. All three mutations alter the physical properties of RAD51 nucleoprotein filaments, with G151D showing the most dramatic changes. G151D forms mixed nucleoprotein filaments with WT RAD51 that have intermediate properties compared to unmixed filaments. These findings raise the possibility that mutations in RAD51 itself may contribute to genome instability in tumor cells, either directly through changes in recombinase properties, or indirectly through changes in interactions with regulatory proteins.


Subject(s)
Mutation , Neoplasms/genetics , Rad51 Recombinase/chemistry , Rad51 Recombinase/genetics , Adenosine Triphosphate/metabolism , Amino Acid Motifs/genetics , DNA/metabolism , Humans , Models, Molecular , Protein Folding , Rad51 Recombinase/metabolism
14.
DNA Repair (Amst) ; 12(12): 1062-71, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24120312

ABSTRACT

Assault to DNA that leads to oxidative base damage is repaired by the base excision repair (BER) pathway with specialized enzymes called DNA glycosylases catalyzing the first step of this pathway. These glycosylases can be categorized into two families: the HhH superfamily, which includes endonuclease III (or Nth), and the Fpg/Nei family, which comprises formamidopyrimidine DNA glycosylase (or Fpg) and endonuclease VIII (or Nei). In humans there are three Nei-like (NEIL) glycosylases: NEIL1, 2, and 3. Here we present the first crystal structure of a viral ortholog of the human NEIL2/NEIL3 proteins, Mimivirus Nei2 (MvNei2), determined at 2.04Å resolution. The C-terminal region of the MvNei2 enzyme comprises two conserved DNA binding motifs: the helix-two-turns-helix (H2TH) motif and a C-H-C-C type zinc-finger similar to that of human NEIL2. The N-terminal region of MvNei2 is most closely related to NEIL3. Like NEIL3, MvNei2 bears a valine at position 2 instead of the usual proline and it lacks two of the three conserved void-filling residues present in other members of the Fpg/Nei family. Mutational analysis of the only conserved void-filling residue methionine 72 to alanine yields an MvNei2 variant with impaired glycosylase activity. Mutation of the adjacent His73 causes the enzyme to be more productive thereby suggesting a plausible role for this residue in the DNA lesion search process.


Subject(s)
DNA Glycosylases/chemistry , DNA Glycosylases/metabolism , Mimiviridae/enzymology , Viral Proteins/chemistry , Viral Proteins/metabolism , Zinc Fingers , Amino Acid Motifs , Amino Acid Sequence , Cloning, Molecular , Consensus Sequence , Crystallography, X-Ray , DNA Damage/genetics , DNA Glycosylases/genetics , DNA Repair , DNA-(Apurinic or Apyrimidinic Site) Lyase/chemistry , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Humans , Mimiviridae/genetics , Mutagenesis, Site-Directed , N-Glycosyl Hydrolases/chemistry , N-Glycosyl Hydrolases/genetics , N-Glycosyl Hydrolases/metabolism , Protein Structure, Secondary , Protein Structure, Tertiary , Sequence Homology , Viral Proteins/genetics
15.
Structure ; 21(2): 247-56, 2013 Feb 05.
Article in English | MEDLINE | ID: mdl-23313161

ABSTRACT

Endonuclease VIII-like 3 (Neil3) is a DNA glycosylase of the base excision repair pathway that protects cells from oxidative DNA damage by excising a broad spectrum of cytotoxic and mutagenic base lesions. Interestingly, Neil3 exhibits an unusual preference for DNA with single-stranded regions. Here, we report the 2.0 Å crystal structure of a Neil3 enzyme. Although the glycosylase region of mouse Neil3 (MmuNeil3Δ324) exhibits the same overall fold as that of other Fpg/Nei proteins, it presents distinct structural features. First, MmuNeil3Δ324 lacks the αF-ß9/10 loop that caps the flipped-out 8-oxoG in bacterial Fpg, which is consistent with its inability to cleave 8-oxoguanine. Second, Neil3 not only lacks two of the three void-filling residues that stabilize the opposite strand, but it also harbors negatively charged residues that create an unfavorable electrostatic environment for the phosphate backbone of that strand. These structural features provide insight into the substrate specificity and marked preference of Neil3 for ssDNA.


Subject(s)
DNA, Single-Stranded/chemistry , Endodeoxyribonucleases/chemistry , Amino Acid Motifs , Animals , Catalytic Domain , Crystallography, X-Ray , Humans , Hydrogen Bonding , Mice , Models, Molecular , Nucleic Acid Conformation , Protein Binding , Protein Stability , Protein Structure, Secondary , Substrate Specificity , Surface Properties
16.
Protein Expr Purif ; 84(1): 130-9, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22569481

ABSTRACT

Endonuclease VIII-like 3 (Neil3) is one of the five DNA glycosylases found in mammals that recognize and remove oxidized bases, and initiate the base excision repair (BER) pathway. Previous attempts to express and purify the mouse and human orthologs of Neil3 in their active form have not been successful. Here we report the construction of bicistronic expression vectors for expressing in Escherichia coli the full-length mouse Neil3 (MmuNeil3), its glycosylase domain (MmuNeil3Δ324), as well as the glycosylase domain of human Neil3 (NEIL3Δ324). The purified Neil3 proteins are all active, and NEIL3Δ324 exhibits similar glycosylase/lyase activity as MmuNeil3Δ324 on both single-stranded and double-stranded substrates containing thymine glycol (Tg), spiroiminodihydantoin (Sp) or an abasic site (AP). We show that N-terminal initiator methionine processing is critical for the activity of both mouse and human Neil3 proteins. Co-expressing an E. coli methionine aminopeptidase (EcoMap) Y168A variant with MmuNeil3, MmuNeil3Δ324 and NEIL3Δ324 improves the N-terminal methionine processing and increases the percentage of active Neil3 proteins in the preparation. The purified Neil3 proteins are suitable for biochemical, structural and functional studies.


Subject(s)
Endodeoxyribonucleases/biosynthesis , Endodeoxyribonucleases/isolation & purification , N-Glycosyl Hydrolases/biosynthesis , N-Glycosyl Hydrolases/isolation & purification , Recombinant Fusion Proteins/isolation & purification , Animals , Cloning, Molecular , Codon , Electrophoresis, Polyacrylamide Gel , Endodeoxyribonucleases/chemistry , Endodeoxyribonucleases/genetics , Enzyme Stability , Escherichia coli/genetics , Escherichia coli/metabolism , Histidine/chemistry , Histidine/genetics , Histidine/metabolism , Humans , Methionine/metabolism , Mice , N-Glycosyl Hydrolases/chemistry , N-Glycosyl Hydrolases/genetics , Oligopeptides/chemistry , Oligopeptides/genetics , Oligopeptides/metabolism , Protein Structure, Tertiary , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...