Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Toxicon ; 238: 107559, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38113945

ABSTRACT

Protein structure determination is a critical aspect of biological research, enabling us to understand protein function and potential applications. Recent advances in deep learning and artificial intelligence have led to the development of several protein structure prediction tools, such as AlphaFold2 and ColabFold. However, their performance has primarily been evaluated on well-characterised proteins and their ability to predict sturtctures of proteins lacking experimental structures, such as many snake venom toxins, has been less scrutinised. In this study, we evaluated three modelling tools on their prediction of over 1000 snake venom toxin structures for which no experimental structures exist. Our findings show that AlphaFold2 (AF2) performed the best across all assessed parameters. We also observed that ColabFold (CF) only scored slightly worse than AF2, while being computationally less intensive. All tools struggled with regions of intrinsic disorder, such as loops and propeptide regions, and performed well in predicting the structure of functional domains. Overall, our study highlights the importance of exercising caution when working with proteins with no experimental structures available, particularly those that are large and contain flexible regions. Nonetheless, leveraging computational structure prediction tools can provide valuable insights into the modelling of protein interactions with different targets and reveal potential binding sites, active sites, and conformational changes, as well as into the design of potential molecular binders for reagent, diagnostic, or therapeutic purposes.


Subject(s)
Artificial Intelligence , Snake Venoms , Binding Sites , Furylfuramide , Proteins/chemistry , Snake Venoms/chemistry
2.
Protein Sci ; 32(12): e4821, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37897425

ABSTRACT

Recycling IgG antibodies bind to their target antigen at physiological pH in the blood stream and release them upon endocytosis when pH levels drop, allowing the IgG antibodies to be recycled into circulation via FcRn-mediated cellular pathways, while the antigens undergo lysosomal degradation. This enables recycling antibodies to achieve comparable therapeutic effect at lower doses than their non-recycling counterparts. The development of such antibodies is typically achieved by histidine doping of their variable regions or by performing in vitro antibody selection campaigns utilizing histidine doped libraries. Both are strategies that may introduce sequence liabilities. Here, we present a methodology that employs a naïve antibody phage display library, consisting of natural variable domains, to discover antibodies that bind α-cobratoxin from the venom of Naja kaouthia in a pH-dependent manner. As a result, an antibody was discovered that exhibits a 7-fold higher off-rate at pH 5.5 than pH 7.4 in bio-layer interferometry experiments. Interestingly, no histidine residues were found in its variable domains, and in addition, the antibody showed pH-dependent binding to a histidine-devoid antigen mutant. As such, the results demonstrate that pH-dependent antigen-antibody binding may not always be driven by histidine residues. By employing molecular dynamics simulations, different protonation states of titratable residues were found, which potentially could be responsible for the observed pH-dependent antigen binding properties of the antibody. Finally, given the typically high diversity of naïve antibody libraries, the methodology presented here can likely be applied to discover recycling antibodies against different targets ab initio without the need for histidine doping.


Subject(s)
Bacteriophages , Histidine , Histidine/metabolism , Antigens/metabolism , Immunoglobulin G/genetics , Hydrogen-Ion Concentration , Bacteriophages/metabolism , Peptide Library
3.
Toxicon ; 234: 107307, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37783315

ABSTRACT

Despite the considerable global impact of snakebite envenoming, available treatments remain suboptimal. Here, we report the discovery of a broadly-neutralizing human monoclonal antibody, using a phage display-based cross-panning strategy, capable of reducing the cytotoxic effects of venom phospholipase A2s from three different snake genera from different continents. This highlights the potential of utilizing monoclonal antibodies to develop more effective, safer, and globally accessible polyvalent antivenoms that can be widely used to treat snakebite envenoming.


Subject(s)
Snake Bites , Animals , Humans , Venoms , Antibodies, Monoclonal , Antivenins/pharmacology , Snakes , Phospholipases A2 , Snake Venoms
4.
Sci Rep ; 13(1): 10181, 2023 06 22.
Article in English | MEDLINE | ID: mdl-37349546

ABSTRACT

Antibodies with cross-reactive binding and broad toxin-neutralizing capabilities are advantageous for treating indications such as infectious diseases and animal envenomings. Such antibodies have been successfully selected against closely related antigens using phage display technology. However, the mechanisms driving antibody cross-reactivity typically remain to be elucidated. Therefore, we sought to explore how a previously reported phage display-based cross-panning strategy drives the selection of cross-reactive antibodies using seven different snake toxins belonging to three protein (sub-)families: phospholipases A2, long-chain α-neurotoxins, and short-chain α-neurotoxins. We showcase how cross-panning can increase the chances of discovering cross-reactive single-chain variable fragments (scFvs) from phage display campaigns. Further, we find that the feasibility of discovering cross-reactive antibodies using cross-panning cannot easily be predicted by analyzing the sequence, structural, or surface similarity of the antigens alone. However, when antigens share the (exact) same functions, this seems to increase the chances of selecting cross-reactive antibodies, which may possibly be due to the existence of structurally similar motifs on the antigens.


Subject(s)
Bacteriophages , Single-Chain Antibodies , Animals , Peptide Library , Neurotoxins , Antigens , Bacteriophages/genetics , Snake Venoms
5.
N Biotechnol ; 76: 23-32, 2023 Sep 25.
Article in English | MEDLINE | ID: mdl-37037303

ABSTRACT

Phage display technology is a powerful tool for selecting monoclonal antibodies against a diverse set of antigens. Within toxinology, however, it remains challenging to generate monoclonal antibodies against many animal toxins, as they are difficult to obtain from venom. Recombinant toxins have been proposed as a solution to overcome this challenge, but so far, few have been used as antigens to generate neutralizing antibodies. Here, we describe the recombinant expression of α-cobratoxin in E. coli and its successful application as an antigen in a phage display selection campaign. From this campaign, an scFv (single-chain variable fragment) was isolated with similar binding affinity to a control scFv generated against the native toxin. The selected scFv recognizes a structural epitope, enabling it to inhibit the interaction between the acetylcholine receptor and the native toxin in vitro. This approach represents the first entirely in vitro antibody selection strategy for generating neutralizing monoclonal antibodies against a snake toxin.


Subject(s)
Bacteriophages , Single-Chain Antibodies , Animals , Single-Chain Antibodies/genetics , Epitopes , Peptide Library , Escherichia coli/genetics , Escherichia coli/metabolism , Antibodies, Monoclonal , Snake Venoms/metabolism , Bacteriophages/metabolism
6.
Cancer Cell ; 40(9): 1060-1069.e7, 2022 09 12.
Article in English | MEDLINE | ID: mdl-36099883

ABSTRACT

Immunotargeting of tumor-specific antigens is a powerful therapeutic strategy. Immunotherapies directed at MHC-I complexes have expanded the scope of antigens and enabled the direct targeting of intracellular oncoproteins at the cell surface. We asked whether covalent drugs that alkylate mutated residues on oncoproteins could act as haptens to generate unique MHC-I-restricted neoantigens. Here, we report that KRAS G12C mutant cells treated with the covalent inhibitor ARS1620 present ARS1620-modified peptides in MHC-I complexes. Using ARS1620-specific antibodies identified by phage display, we show that these haptenated MHC-I complexes can serve as tumor-specific neoantigens and that a bispecific T cell engager construct based on a hapten-specific antibody elicits a cytotoxic T cell response against KRAS G12C cells, including those resistant to direct KRAS G12C inhibition. With multiple K-RAS G12C inhibitors in clinical use or undergoing clinical trials, our results present a strategy to enhance their efficacy and overcome the rapidly arising tumor resistance.


Subject(s)
Antineoplastic Agents , Histocompatibility Antigens Class I/immunology , Neoplasms , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Antibodies , Antineoplastic Agents/pharmacology , Humans , Immunologic Factors , Immunotherapy , Peptides/pharmacology , Proto-Oncogene Proteins p21(ras)/genetics
7.
Bioconjug Chem ; 33(8): 1494-1504, 2022 08 17.
Article in English | MEDLINE | ID: mdl-35875886

ABSTRACT

Recombinantly produced biotherapeutics hold promise for improving the current standard of care for snakebite envenoming over conventional serotherapy. Nanobodies have performed well in the clinic, and in the context of antivenom, they have shown the ability to neutralize long α-neurotoxins in vivo. Here, we showcase a protein engineering approach to increase the valence and hydrodynamic size of neutralizing nanobodies raised against a long α-neurotoxin (α-cobratoxin) from the venom of the monocled cobraNaja kaouthia. Based on the p53 tetramerization domain, a panel of anti-α-cobratoxin nanobody-p53 fusion proteins, termed Quads, were produced with different valences, inclusion or exclusion of Fc regions for endosomal recycling purposes, hydrodynamic sizes, and spatial arrangements, comprising up to 16 binding sites. Measurements of binding affinity and stoichiometry showed that the nanobody binding affinity was retained when incorporated into the Quad scaffold, and all nanobody domains were accessible for toxin binding, subsequently displaying increased blocking potency in vitro compared to the monomeric format. Moreover, functional assessment using automated patch-clamp assays demonstrated that the nanobody and Quads displayed neutralizing effects against long α-neurotoxins from both N. kaouthia and the forest cobra N. melanoleuca. This engineering approach offers a means of altering the valence, endosomal recyclability, and hydrodynamic size of existing nanobody-based therapeutics in a simple plug-and-play fashion and can thus serve as a technology for researchers tailoring therapeutic properties for improved neutralization of soluble targets such as snake toxins.


Subject(s)
Elapidae , Single-Domain Antibodies , Animals , Elapid Venoms/chemistry , Elapid Venoms/metabolism , Elapidae/metabolism , Neurotoxins/chemistry , Neurotoxins/metabolism , Single-Domain Antibodies/metabolism , Tumor Suppressor Protein p53/metabolism
8.
Cell Chem Biol ; 29(5): 785-798.e19, 2022 05 19.
Article in English | MEDLINE | ID: mdl-35364007

ABSTRACT

Viruses are responsible for some of the most deadly human diseases, yet available vaccines and antivirals address only a fraction of the potential viral human pathogens. Here, we provide a methodology for managing human herpesvirus (HHV) infection by covalently inactivating the HHV maturational protease via a conserved, non-catalytic cysteine (C161). Using human cytomegalovirus protease (HCMV Pr) as a model, we screened a library of disulfides to identify molecules that tether to C161 and inhibit proteolysis, then elaborated hits into irreversible HCMV Pr inhibitors that exhibit broad-spectrum inhibition of other HHV Pr homologs. We further developed an optimized tool compound targeted toward HCMV Pr and used an integrative structural biology and biochemical approach to demonstrate inhibitor stabilization of HCMV Pr homodimerization, exploiting a conformational equilibrium to block proteolysis. Irreversible HCMV Pr inhibition disrupts HCMV infectivity in cells, providing proof of principle for targeting proteolysis via a non-catalytic cysteine to manage viral infection.


Subject(s)
Cytomegalovirus Infections , Cytomegalovirus , Cysteine , Cytomegalovirus/physiology , Humans , Peptide Hydrolases , Viral Proteases
10.
ACS Nano ; 15(11): 18374-18384, 2021 11 23.
Article in English | MEDLINE | ID: mdl-34694776

ABSTRACT

Inorganic nanocrystals such as quantum dots (QDs) and upconverting nanoparticles (UCNPs) are uniquely suited for quantitative live-cell imaging and are typically functionalized with ligands to study specific receptors or cellular targets. Antibodies (Ab) are among the most useful targeting reagents owing to their high affinities and specificities, but common nanocrystal labeling methods may orient Ab incorrectly, be reversible or denaturing, or lead to Ab-NP complexes too large for some applications. Here, we show that SpyCatcher proteins, which bind and spontaneously form covalent isopeptide bonds with cognate SpyTag peptides, can conjugate engineered Ab to nanoparticle surfaces with control over stability, orientation, and stoichiometry. Compact SpyCatcher-functionalized QDs and UCNPs may be labeled with short-chain variable fragment Ab (scFv) engineered to bind urokinase-type plasminogen activator receptors (uPAR) that are overexpressed in many human cancers. Confocal imaging of anti-uPAR scFv-QD conjugates shows the antibody mediates specific binding and internalization by breast cancer cells expressing uPAR. Time-lapse imaging of photostable scFv-UCNP conjugates shows that Ab binding causes uPAR internalization with a ∼20 min half-life on the cell surface, and uPAR is internalized to endolysosomal compartments distinct from general membrane stains and without significant recycling to the cell surface. The controlled and stable conjugation of engineered Ab to NPs enables targeting of diverse receptors for live-cell study of their distribution, trafficking, and physiology.


Subject(s)
Nanoparticles , Quantum Dots , Humans , Receptors, Urokinase Plasminogen Activator/metabolism , Antibodies/metabolism , Cell Membrane/metabolism , Urokinase-Type Plasminogen Activator/metabolism
11.
Cell Rep ; 21(11): 3243-3255, 2017 Dec 12.
Article in English | MEDLINE | ID: mdl-29241550

ABSTRACT

The heavy chain IGHV1-69 germline gene exhibits a high level of polymorphism and shows biased use in protective antibody (Ab) responses to infections and vaccines. It is also highly expressed in several B cell malignancies and autoimmune diseases. G6 is an anti-idiotypic monoclonal Ab that selectively binds to IGHV1-69 heavy chain germline gene 51p1 alleles that have been implicated in these Ab responses and disease processes. Here, we determine the co-crystal structure of humanized G6 (hG6.3) in complex with anti-influenza hemagglutinin stem-directed broadly neutralizing Ab D80. The core of the hG6.3 idiotope is a continuous string of CDR-H2 residues starting with M53 and ending with N58. G6 binding studies demonstrate the remarkable breadth of binding to 51p1 IGHV1-69 Abs with diverse CDR-H3, light chain, and antigen binding specificities. These studies detail the broad expression of the G6 cross-reactive idiotype (CRI) that further define its potential role in precision medicine.


Subject(s)
Antibodies, Anti-Idiotypic/chemistry , Antibodies, Monoclonal, Humanized/chemistry , Antibodies, Neutralizing/chemistry , Antibodies, Viral/chemistry , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Receptors, Antigen, B-Cell/chemistry , Amino Acid Sequence , Antibodies, Anti-Idiotypic/genetics , Antibodies, Anti-Idiotypic/immunology , Antibodies, Monoclonal, Humanized/genetics , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Neutralizing/genetics , Antibodies, Neutralizing/immunology , Antibodies, Viral/genetics , Antibodies, Viral/immunology , Antibody Specificity , Binding Sites , Cloning, Molecular , Crystallography, X-Ray , Gene Expression , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Models, Molecular , Orthomyxoviridae/chemistry , Protein Binding , Protein Interaction Domains and Motifs , Protein Structure, Secondary , Receptors, Antigen, B-Cell/genetics , Receptors, Antigen, B-Cell/immunology , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Sequence Alignment , Sequence Homology, Amino Acid
12.
J Am Chem Soc ; 139(34): 11650-11653, 2017 08 30.
Article in English | MEDLINE | ID: mdl-28759216

ABSTRACT

Targeting of cryptic binding sites represents an attractive but underexplored approach to modulating protein function with small molecules. Using the dimeric protease (Pr) from Kaposi's sarcoma-associated herpesvirus (KSHV) as a model system, we sought to dissect a putative allosteric network linking a cryptic site at the dimerization interface to enzyme function. Five cryogenic X-ray structures were solved of the monomeric protease with allosteric inhibitors bound to the dimer interface site. Distinct coordinated movements captured by the allosteric inhibitors were also revealed as alternative states in room-temperature X-ray data and comparative analyses of other dimeric herpesvirus proteases. A two-step mechanism was elucidated through detailed kinetic analyses and suggests an enzyme isomerization model of inhibition. Finally, a representative allosteric inhibitor from this class was shown to be efficacious in a cellular model of viral infectivity. These studies reveal a coordinated dynamic network of atomic communication linking cryptic binding site occupancy and allosteric inactivation of KHSV Pr that can be exploited to target other members of this clinically relevant family of enzymes.


Subject(s)
Allosteric Regulation/drug effects , Herpesviridae Infections/virology , Herpesvirus 8, Human/enzymology , Peptide Hydrolases/metabolism , Protease Inhibitors/pharmacology , Crystallography, X-Ray , Herpesviridae Infections/drug therapy , Herpesvirus 8, Human/chemistry , Herpesvirus 8, Human/drug effects , Humans , Models, Molecular , Peptide Hydrolases/chemistry , Protein Conformation/drug effects , Protein Multimerization/drug effects
13.
Hum Mutat ; 37(9): 893-7, 2016 09.
Article in English | MEDLINE | ID: mdl-27297501

ABSTRACT

Some variants that cause autosomal-recessive congenital adrenal hyperplasia (CAH) also cause hypermobility type Ehlers-Danlos syndrome (EDS) due to the monoallelic presence of a chimera disrupting two flanking genes: CYP21A2, encoding 21-hydroxylase, necessary for cortisol and aldosterone biosynthesis, and TNXB, encoding tenascin-X, an extracellular matrix protein. Two types of CAH tenascin-X (CAH-X) chimeras have been described with a total deletion of CYP21A2 and characteristic TNXB variants. CAH-X CH-1 has a TNXB exon 35 120-bp deletion resulting in haploinsufficiency, and CAH-X CH-2 has a TNXB exon 40 c.12174C>G (p.Cys4058Trp) variant resulting in a dominant-negative effect. We present here three patients with biallelic CAH-X and identify a novel dominant-negative chimera termed CAH-X CH-3. Compared with monoallelic CAH-X, biallelic CAH-X results in a more severe phenotype with skin features characteristic of classical EDS. We present evidence for disrupted tenascin-X function and computational data linking the type of TNXB variant to disease severity.


Subject(s)
Adrenal Hyperplasia, Congenital/genetics , Ehlers-Danlos Syndrome/genetics , Gene Deletion , Steroid 21-Hydroxylase/genetics , Tenascin/genetics , Adolescent , Adrenal Hyperplasia, Congenital/metabolism , Adult , Alleles , Collagen/metabolism , Ehlers-Danlos Syndrome/metabolism , Female , Fibrillin-1/metabolism , Humans , Male , Pedigree , Tenascin/metabolism , Young Adult
14.
Acta Crystallogr D Biol Crystallogr ; 71(Pt 5): 1059-67, 2015 May.
Article in English | MEDLINE | ID: mdl-25945571

ABSTRACT

High-throughput crystallographic approaches require integrated software solutions to minimize the need for manual effort. REdiii is a system that allows fully automated crystallographic structure solution by integrating existing crystallographic software into an adaptive and partly autonomous workflow engine. The program can be initiated after collecting the first frame of diffraction data and is able to perform processing, molecular-replacement phasing, chain tracing, ligand fitting and refinement without further user intervention. Preset values for each software component allow efficient progress with high-quality data and known parameters. The adaptive workflow engine can determine whether some parameters require modifications and choose alternative software strategies in case the preconfigured solution is inadequate. This integrated pipeline is targeted at providing a comprehensive and efficient approach to screening for ligand-bound co-crystal structures while minimizing repetitiveness and allowing a high-throughput scientific discovery process.


Subject(s)
Automation, Laboratory/methods , Data Collection/methods , Macromolecular Substances/chemistry , Software , Algorithms , Crystallography, X-Ray , Humans , Models, Molecular , Solutions
15.
Structure ; 23(5): 903-911, 2015 May 05.
Article in English | MEDLINE | ID: mdl-25914058

ABSTRACT

Deaminase activity mediated by the human APOBEC3 family of proteins contributes to genomic instability and cancer. APOBEC3A is by far the most active in this family and can cause rapid cell death when overexpressed, but in general how the activity of APOBEC3s is regulated on a molecular level is unclear. In this study, the biochemical and structural basis of APOBEC3A substrate binding and specificity is elucidated. We find that specific binding of single-stranded DNA is regulated by the cooperative dimerization of APOBEC3A. The crystal structure elucidates this homodimer as a symmetric domain swap of the N-terminal residues. This dimer interface provides insights into how cooperative protein-protein interactions may affect function in the APOBEC3 enzymes and provides a potential scaffold for strategies aimed at reducing their mutation load.


Subject(s)
Cytidine Deaminase/chemistry , Cytidine Deaminase/metabolism , DNA, Single-Stranded/metabolism , Proteins/chemistry , Proteins/metabolism , Binding Sites , Crystallography, X-Ray , Cytidine Deaminase/genetics , Dimerization , Humans , Models, Molecular , Mutation , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Proteins/genetics , Substrate Specificity , Zinc/metabolism
16.
Virology ; 471-473: 105-16, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25461536

ABSTRACT

APOBEC3s (A3) are Zn(2+) dependent cytidine deaminases with diverse biological functions and implications for cancer and immunity. Four of the seven human A3s restrict HIV by 'hypermutating' the reverse-transcribed viral genomic DNA. HIV Virion Infectivity Factor (Vif) counters this restriction by targeting A3s to proteasomal degradation. However, there is no apparent correlation between catalytic activity, Vif binding, and sequence similarity between A3 domains. Our comparative structural analysis reveals features required for binding Vif and features influencing polynucleotide deaminase activity in A3 proteins. All Vif-binding A3s share a negatively charged surface region that includes residues previously implicated in binding the highly-positively charged Vif. Additionally, catalytically active A3s share a positively charged groove near the Zn(2+) coordinating active site, which may accommodate the negatively charged polynucleotide substrate. Our findings suggest surface electrostatics, as well as the spatial extent of substrate accommodating region, are critical determinants of substrate and Vif binding across A3 proteins with implications for anti-retroviral and anti-cancer therapeutic design.


Subject(s)
Cytosine Deaminase/chemistry , Cytosine Deaminase/metabolism , vif Gene Products, Human Immunodeficiency Virus/chemistry , vif Gene Products, Human Immunodeficiency Virus/metabolism , APOBEC Deaminases , Amino Acid Sequence , Cytidine Deaminase , Gene Expression Regulation , Humans , Membrane Potentials , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Sequence Alignment , Static Electricity
17.
Structure ; 21(6): 1042-50, 2013 Jun 04.
Article in English | MEDLINE | ID: mdl-23685212

ABSTRACT

Human APOBEC3F is an antiretroviral single-strand DNA cytosine deaminase, susceptible to degradation by the HIV-1 protein Vif. In this study the crystal structure of the HIV Vif binding, catalytically active, C-terminal domain of APOBEC3F (A3F-CTD) was determined. The A3F-CTD shares structural motifs with portions of APOBEC3G-CTD, APOBEC3C, and APOBEC2. Residues identified to be critical for Vif-dependent degradation of APOBEC3F all fit within a predominantly negatively charged contiguous region on the surface of A3F-CTD. Specific sequence motifs, previously shown to play a role in Vif susceptibility and virion encapsidation, are conserved across APOBEC3s and between APOBEC3s and HIV-1 Vif. In this structure these motifs pack against each other at intermolecular interfaces, providing potential insights both into APOBEC3 oligomerization and Vif interactions.


Subject(s)
Cytosine Deaminase/metabolism , HIV-1/metabolism , vif Gene Products, Human Immunodeficiency Virus/metabolism , Catalysis , Crystallography, X-Ray , Cytosine Deaminase/chemistry , Models, Molecular , Protein Conformation
18.
J Biol Chem ; 287(41): 34801-8, 2012 Oct 05.
Article in English | MEDLINE | ID: mdl-22896697

ABSTRACT

Multiple studies have indicated that the TET oxidases and, more controversially, the activation-induced cytidine deaminase/APOBEC deaminases have the capacity to convert genomic DNA 5-methylcytosine (MeC) into altered nucleobases that provoke excision repair and culminate in the replacement of the original MeC with a normal cytosine (C). We show that human APOBEC3A (A3A) efficiently deaminates both MeC to thymine (T) and normal C to uracil (U) in single-stranded DNA substrates. In comparison, the related enzyme APOBEC3G (A3G) has undetectable MeC to T activity and 10-fold less C to U activity. Upon 100-fold induction of endogenous A3A by interferon, the MeC status of bulk chromosomal DNA is unaltered, whereas both MeC and C nucleobases in transfected plasmid DNA substrates are highly susceptible to editing. Knockdown experiments show that endogenous A3A is the source of both of these cellular DNA deaminase activities. This is the first evidence for nonchromosomal DNA MeC to T editing in human cells. These biochemical and cellular data combine to suggest a model in which the expanded substrate versatility of A3A may be an evolutionary adaptation that occurred to fortify its innate immune function in foreign DNA clearance by myeloid lineage cell types.


Subject(s)
5-Methylcytosine/metabolism , Cytidine Deaminase/metabolism , DNA/metabolism , Immunity, Innate , Proteins/metabolism , 5-Methylcytosine/immunology , Cytidine Deaminase/immunology , DNA/immunology , Deamination , Enzyme Induction/drug effects , Enzyme Induction/immunology , HEK293 Cells , Humans , Interferons/immunology , Interferons/pharmacology , Plasmids/immunology , Plasmids/pharmacology , Proteins/immunology , Thymine/immunology , Thymine/metabolism , Uracil/immunology , Uracil/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...