Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Nucleic Acids Res ; 50(9): 5171-5190, 2022 05 20.
Article in English | MEDLINE | ID: mdl-35511079

ABSTRACT

Bacteriophage exclusion ('BREX') phage restriction systems are found in a wide range of bacteria. Various BREX systems encode unique combinations of proteins that usually include a site-specific methyltransferase; none appear to contain a nuclease. Here we describe the identification and characterization of a Type I BREX system from Acinetobacter and the effect of deleting each BREX ORF on growth, methylation, and restriction. We identified a previously uncharacterized gene in the BREX operon that is dispensable for methylation but involved in restriction. Biochemical and crystallographic analyses of this factor, which we term BrxR ('BREX Regulator'), demonstrate that it forms a homodimer and specifically binds a DNA target site upstream of its transcription start site. Deletion of the BrxR gene causes cell toxicity, reduces restriction, and significantly increases the expression of BrxC. In contrast, the introduction of a premature stop codon into the BrxR gene, or a point mutation blocking its DNA binding ability, has little effect on restriction, implying that the BrxR coding sequence and BrxR protein play independent functional roles. We speculate that elements within the BrxR coding sequence are involved in cis regulation of anti-phage activity, while the BrxR protein itself plays an additional regulatory role, perhaps during horizontal transfer.


Subject(s)
Acinetobacter/physiology , Antiviral Restriction Factors , Bacteriophages , Acinetobacter/genetics , Acinetobacter/virology , Antiviral Restriction Factors/genetics , Bacteriophages/physiology , DNA/metabolism , Methyltransferases/genetics , Operon
2.
Structure ; 28(7): 760-775.e8, 2020 07 07.
Article in English | MEDLINE | ID: mdl-32359399

ABSTRACT

The redesign of a macromolecular binding interface and corresponding alteration of recognition specificity is a challenging endeavor that remains recalcitrant to computational approaches. This is particularly true for the redesign of DNA binding specificity, which is highly dependent upon bending, hydrogen bonds, electrostatic contacts, and the presence of solvent and counterions throughout the molecular interface. Thus, redesign of protein-DNA binding specificity generally requires iterative rounds of amino acid randomization coupled to selections. Here, we describe the importance of scaffold thermostability for protein engineering, coupled with a strategy that exploits the protein's specificity profile, to redesign the specificity of a pair of meganucleases toward three separate genomic targets. We determine and describe a series of changes in protein sequence, stability, structure, and activity that accumulate during the engineering process, culminating in fully retargeted endonucleases.


Subject(s)
DNA-Binding Proteins/chemistry , DNA/chemistry , Animals , Binding Sites , DNA/genetics , DNA/metabolism , DNA-Binding Proteins/metabolism , Gene Editing/methods , Humans , Mutation , Protein Binding , Protein Engineering/methods , Protein Stability
3.
Protein Eng Des Sel ; 30(7): 503-522, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28873986

ABSTRACT

The combination of yeast surface display and flow cytometric analyses and selections is being used with increasing frequency to alter specificity of macromolecular recognition, including both protein-protein and protein-nucleic acid interactions. Here we describe the use of yeast surface display and cleavage-dependent flow cytometric assays to increase the specificity of an engineered meganuclease. The re-engineered meganuclease displays a significantly tightened specificity profile, while binding its cognate target site with a slightly lower, but still sub-nanomolar affinity. When incorporated into otherwise identical megaTAL protein scaffolds, these two nucleases display significantly different activity and toxicity profiles in cellulo. The structural basis for reprogrammed DNA cleavage specificity was further examined via high-resolution X-ray crystal structures of both enzymes. This analysis illustrated the altered protein-DNA contacts produced by mutagenesis and selection, that resulted both in altered readout of those based and a necessary reduction in DNA binding affinity that were necessary to improve specificity across the target site. The results of this study provide an illustrative example of the potential (and the challenges) associated with the use of surface display and flow cytometry for the retargeting and optimization of enzymes that act on nucleic acid substrates in a sequence-specific manner.


Subject(s)
DNA/genetics , Deoxyribonucleases/chemistry , Endonucleases/genetics , Metabolic Engineering , Binding Sites , DNA/chemistry , DNA Cleavage , Deoxyribonucleases/genetics , Endonucleases/chemistry , Flow Cytometry , Substrate Specificity
4.
Nucleic Acids Res ; 45(14): 8621-8634, 2017 Aug 21.
Article in English | MEDLINE | ID: mdl-28637173

ABSTRACT

The retargeting of protein-DNA specificity, outside of extremely modular DNA binding proteins such as TAL effectors, has generally proved to be quite challenging. Here, we describe structural analyses of five different extensively retargeted variants of a single homing endonuclease, that have been shown to function efficiently in ex vivo and in vivo applications. The redesigned proteins harbor mutations at up to 53 residues (18%) of their amino acid sequence, primarily distributed across the DNA binding surface, making them among the most significantly reengineered ligand-binding proteins to date. Specificity is derived from the combined contributions of DNA-contacting residues and of neighboring residues that influence local structural organization. Changes in specificity are facilitated by the ability of all those residues to readily exchange both form and function. The fidelity of recognition is not precisely correlated with the fraction or total number of residues in the protein-DNA interface that are actually involved in DNA contacts, including directional hydrogen bonds. The plasticity of the DNA-recognition surface of this protein, which allows substantial retargeting of recognition specificity without requiring significant alteration of the surrounding protein architecture, reflects the ability of the corresponding genetic elements to maintain mobility and persistence in the face of genetic drift within potential host target sites.


Subject(s)
DNA/chemistry , DNA/metabolism , Endodeoxyribonucleases/chemistry , Endodeoxyribonucleases/metabolism , Amino Acid Sequence , Animals , Animals, Genetically Modified , Base Sequence , Binding Sites/genetics , Crystallography , Culicidae/enzymology , Culicidae/genetics , DNA/genetics , Endodeoxyribonucleases/genetics , Models, Molecular , Mutation , Nucleic Acid Conformation , Protein Binding , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Substrate Specificity
5.
Nucleic Acids Res ; 45(3): e11, 2017 02 17.
Article in English | MEDLINE | ID: mdl-28180328

ABSTRACT

LAGLIDADG homing endonucleases (LHEs) are a class of rare-cleaving nucleases that possess several unique attributes for genome engineering applications. An important approach for advancing LHE technology is the generation of a library of design 'starting points' through the discovery and characterization of natural LHEs with diverse specificities. However, while identification of natural LHE proteins by sequence homology from genomic and metagenomic sequence databases is straightforward, prediction of corresponding target sequences from genomic data remains challenging. Here, we describe a general approach that we developed to circumvent this issue that combines two technologies: yeast surface display (YSD) of LHEs and systematic evolution of ligands via exponential enrichment (SELEX). Using LHEs expressed on the surface of yeast, we show that SELEX can yield binding specificity motifs and identify cleavable LHE targets using a combination of bioinformatics and biochemical cleavage assays. This approach, which we term YSD-SELEX, represents a simple and rapid first principles approach to determining the binding and cleavage specificity of novel LHEs that should also be generally applicable to any type of yeast surface expressible DNA-binding protein. In this marriage, SELEX adds DNA specificity determination to the YSD platform, and YSD brings diagnostics and inexpensive, facile protein-matrix generation to SELEX.


Subject(s)
Endonucleases/metabolism , SELEX Aptamer Technique/methods , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Amino Acid Motifs , Amino Acid Sequence , Catalytic Domain/genetics , DNA, Fungal/genetics , DNA, Fungal/metabolism , Endodeoxyribonucleases/chemistry , Endodeoxyribonucleases/genetics , Endodeoxyribonucleases/metabolism , Endonucleases/chemistry , Endonucleases/genetics , Phylogeny , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Sequence Homology, Amino Acid , Substrate Specificity
6.
Structure ; 24(6): 862-73, 2016 06 07.
Article in English | MEDLINE | ID: mdl-27133026

ABSTRACT

LAGLIDADG meganucleases are DNA cleaving enzymes used for genome engineering. While their cleavage specificity can be altered using several protein engineering and selection strategies, their overall targetability is limited by highly specific indirect recognition of the central four base pairs within their recognition sites. In order to examine the physical basis of indirect sequence recognition and to expand the number of such nucleases available for genome engineering, we have determined the target sites, DNA-bound structures, and central four cleavage fidelities of nine related enzymes. Subsequent crystallographic analyses of a meganuclease bound to two noncleavable target sites, each containing a single inactivating base pair substitution at its center, indicates that a localized slip of the mutated base pair causes a small change in the DNA backbone conformation that results in a loss of metal occupancy at one binding site, eliminating cleavage activity.


Subject(s)
DNA/chemistry , DNA/metabolism , Deoxyribonucleases/chemistry , Deoxyribonucleases/metabolism , Base Sequence , Binding Sites , DNA Cleavage , Models, Molecular , Nucleic Acid Conformation , Protein Conformation , Substrate Specificity
7.
Methods Mol Biol ; 1123: 191-221, 2014.
Article in English | MEDLINE | ID: mdl-24510269

ABSTRACT

LAGLIDADG homing endonucleases (LHEs) are valuable tools for genome engineering, and our ability to alter LHE target site specificity is rapidly evolving. However, widespread use of these enzymes is limited due to the small number of available engineering scaffolds, each requiring extensive redesign to target widely varying DNA sequences. Here, we describe a technique for the chimerization of homologous I-OnuI family LHEs. Chimerization greatly expands the pool of unique starting scaffolds, thereby enabling more effective and efficient LHE redesign. I-OnuI family enzymes are divided into N- and C-terminal halves based on sequence alignments, and then combinatorially rejoined with a hybrid linker. The resulting chimeric enzymes are expressed on the surface of yeast where stability, DNA binding affinity, and cleavage activity can be assayed by flow cytometry.


Subject(s)
Endonucleases/metabolism , Flow Cytometry , Protein Engineering , Recombinant Fusion Proteins/metabolism , Cloning, Molecular , Endonucleases/genetics , Gene Expression , Recombinant Fusion Proteins/genetics , Substrate Specificity
8.
Methods Mol Biol ; 978: 45-61, 2013.
Article in English | MEDLINE | ID: mdl-23423888

ABSTRACT

A fast, easy, and scalable method to assess the properties of site-specific nucleases is crucial to -understanding their in cellulo behavior in genome engineering or population-level gene drive applications. Here we describe an analytical platform that enables high-throughput, semiquantitative interrogation of the DNA-binding and catalytic properties of LAGLIDADG homing endonucleases (LHEs). Using this platform, natural or engineered LHEs are expressed on the surface of Saccharomyces cerevisiae yeast where they can be rapidly evaluated against synthetic DNA target sequences using flow cytometry.


Subject(s)
DNA/metabolism , Endonucleases/metabolism , Flow Cytometry/methods , Protein Binding , Saccharomyces cerevisiae
9.
Nat Methods ; 9(10): 973-5, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22941364

ABSTRACT

Targeted DNA double-strand breaks introduced by rare-cleaving designer endonucleases can be harnessed for gene disruption applications by engaging mutagenic nonhomologous end-joining DNA repair pathways. However, endonuclease-mediated DNA breaks are often subject to precise repair, which limits the efficiency of targeted genome editing. To address this issue, we coupled designer endonucleases to DNA end-processing enzymes to drive mutagenic break resolution, achieving up to 25-fold enhancements in gene disruption rates.


Subject(s)
DNA Breaks, Double-Stranded , Endonucleases/physiology , Animals , DNA End-Joining Repair , DNA Repair , Exodeoxyribonucleases/physiology , HEK293 Cells , Humans , Mice , Phosphoproteins/physiology , Receptors, CCR5/physiology
10.
Nucleic Acids Res ; 40(16): 7985-8000, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22684507

ABSTRACT

Although engineered LAGLIDADG homing endonucleases (LHEs) are finding increasing applications in biotechnology, their generation remains a challenging, industrial-scale process. As new single-chain LAGLIDADG nuclease scaffolds are identified, however, an alternative paradigm is emerging: identification of an LHE scaffold whose native cleavage site is a close match to a desired target sequence, followed by small-scale engineering to modestly refine recognition specificity. The application of this paradigm could be accelerated if methods were available for fusing N- and C-terminal domains from newly identified LHEs into chimeric enzymes with hybrid cleavage sites. Here we have analyzed the structural requirements for fusion of domains extracted from six single-chain I-OnuI family LHEs, spanning 40-70% amino acid identity. Our analyses demonstrate that both the LAGLIDADG helical interface residues and the linker peptide composition have important effects on the stability and activity of chimeric enzymes. Using a simple domain fusion method in which linker peptide residues predicted to contact their respective domains are retained, and in which limited variation is introduced into the LAGLIDADG helix and nearby interface residues, catalytically active enzymes were recoverable for ≈ 70% of domain chimeras. This method will be useful for creating large numbers of chimeric LHEs for genome engineering applications.


Subject(s)
Endodeoxyribonucleases/chemistry , Protein Engineering/methods , Amino Acid Motifs , Amino Acid Sequence , Base Pairing , DNA/chemistry , Endodeoxyribonucleases/genetics , Endodeoxyribonucleases/metabolism , Enzyme Stability , HEK293 Cells , Humans , Molecular Sequence Data , Peptides/chemistry , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism
11.
Nucleic Acids Res ; 40(Web Server issue): W110-6, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22570419

ABSTRACT

LAGLIDADG homing endonucleases (LHEs) are DNA cleaving enzymes, also termed 'meganucleases' that are employed as gene-targeting reagents. This use of LHEs requires that their DNA specificity be altered to match sequences in genomic targets. The choice of the most appropriate LHE to target a particular gene is facilitated by the growing number of such enzymes with well-characterized activities and structures. 'LAHEDES' (The LAGLIDADG Homing Endonuclease Database and Engineering Server) provides both an online archive of LHEs with validated DNA cleavage specificities and DNA-binding interactions, as well as a tool for the identification of DNA sequences that might be targeted by various LHEs. Searches can be performed using four separate scoring algorithms and user-defined choices of LHE scaffolds. The webserver subsequently provides information regarding clusters of amino acids that should be interrogated during engineering and selection experiments. The webserver is fully open access and can be found at http://homingendonuclease.net.


Subject(s)
Databases, Genetic , Endodeoxyribonucleases/chemistry , Endodeoxyribonucleases/metabolism , Software , DNA/chemistry , DNA/metabolism , DNA Cleavage , Endodeoxyribonucleases/genetics , Gene Targeting , Internet , Mutation , Protein Engineering , Protein Structure, Tertiary , Substrate Specificity
12.
Proc Natl Acad Sci U S A ; 108(32): 13077-82, 2011 Aug 09.
Article in English | MEDLINE | ID: mdl-21784983

ABSTRACT

Homing endonucleases mobilize their own genes by generating double-strand breaks at individual target sites within potential host DNA. Because of their high specificity, these proteins are used for "genome editing" in higher eukaryotes. However, alteration of homing endonuclease specificity is quite challenging. Here we describe the identification and phylogenetic analysis of over 200 naturally occurring LAGLIDADG homing endonucleases (LHEs). Biochemical and structural characterization of endonucleases from one clade within the phylogenetic tree demonstrates strong conservation of protein structure contrasted against highly diverged DNA target sites and indicates that a significant fraction of these proteins are sufficiently stable and active to serve as engineering scaffolds. This information was exploited to create a targeting enzyme to disrupt the endogenous monoamine oxidase B gene in human cells. The ubiquitous presence and diversity of LHEs described in this study may facilitate the creation of many tailored nucleases for genome editing.


Subject(s)
Endonucleases/metabolism , Gene Targeting , Amino Acid Sequence , Base Sequence , Endonucleases/chemistry , Humans , Models, Molecular , Molecular Sequence Data , Monoamine Oxidase/genetics , Mutagenesis/genetics , Protein Binding , Protein Engineering , Substrate Specificity
13.
Structure ; 18(10): 1321-31, 2010 Oct 13.
Article in English | MEDLINE | ID: mdl-20800503

ABSTRACT

The GIY-YIG endonuclease family comprises hundreds of diverse proteins and a multitude of functions; none have been visualized bound to DNA. The structure of the GIY-YIG restriction endonuclease R.Eco29kI has been solved both alone and bound to its target site. The protein displays a domain-swapped homodimeric structure with several extended surface loops encircling the DNA. Only three side chains from each protein subunit contact DNA bases, two directly and one via a bridging solvent molecule. Both tyrosine residues within the GIY-YIG motif are positioned in the catalytic center near a putative nucleophilic water; the remainder of the active site resembles the HNH endonuclease family. The structure illustrates how the GIY-YIG scaffold has been adapted for the highly specific recognition of a DNA restriction site, in contrast to nonspecific DNA cleavage by GIY-YIG domains in homing endonucleases or structure-specific cleavage by DNA repair enzymes such as UvrC.


Subject(s)
DNA/chemistry , Deoxyribonucleases, Type II Site-Specific/chemistry , Protein Folding , Protein Structure, Tertiary , Amino Acid Sequence , Base Sequence , Binding Sites/genetics , Crystallization , Crystallography, X-Ray , DNA/metabolism , Deoxyribonucleases, Type II Site-Specific/genetics , Deoxyribonucleases, Type II Site-Specific/metabolism , Models, Molecular , Molecular Sequence Data , Mutation, Missense , Protein Binding , Protein Multimerization , Sequence Homology, Amino Acid
14.
Science ; 329(5989): 309-13, 2010 Jul 16.
Article in English | MEDLINE | ID: mdl-20647463

ABSTRACT

The Diels-Alder reaction is a cornerstone in organic synthesis, forming two carbon-carbon bonds and up to four new stereogenic centers in one step. No naturally occurring enzymes have been shown to catalyze bimolecular Diels-Alder reactions. We describe the de novo computational design and experimental characterization of enzymes catalyzing a bimolecular Diels-Alder reaction with high stereoselectivity and substrate specificity. X-ray crystallography confirms that the structure matches the design for the most active of the enzymes, and binding site substitutions reprogram the substrate specificity. Designed stereoselective catalysts for carbon-carbon bond-forming reactions should be broadly useful in synthetic chemistry.


Subject(s)
Carbon/chemistry , Computer-Aided Design , Enzymes/chemistry , Protein Engineering , Proteins/chemistry , Acrylamides/chemistry , Algorithms , Butadienes/chemistry , Catalysis , Catalytic Domain , Chemical Phenomena , Computer Simulation , Crystallography, X-Ray , Enzymes/genetics , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Kinetics , Models, Molecular , Mutagenesis , Protein Conformation , Proteins/genetics , Software , Stereoisomerism , Substrate Specificity
15.
Structure ; 16(4): 558-69, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18400177

ABSTRACT

The structure of the rare-cutting restriction endonuclease NotI, which recognizes the 8 bp target 5'-GCGGCCGC-3', has been solved with and without bound DNA. Because of its specificity (recognizing a site that occurs once per 65 kb), NotI is used to generate large genomic fragments and to map DNA methylation status. NotI contains a unique metal binding fold, found in a variety of putative endonucleases, occupied by an iron atom coordinated within a tetrahedral Cys4 motif. This domain positions nearby protein elements for DNA recognition, and serves a structural role. While recognition of the central six base pairs of the target is accomplished via a saturated hydrogen bond network typical of restriction enzymes, the most peripheral base pairs are engaged in a single direct contact in the major groove, reflecting reduced pressure to recognize those positions. NotI may represent an evolutionary intermediate between mobile endonucleases (which recognize longer target sites) and canonical restriction endonucleases.


Subject(s)
DNA/chemistry , Deoxyribonucleases, Type II Site-Specific/chemistry , Models, Molecular , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray , Iron/chemistry , Molecular Sequence Data , Protein Binding , Protein Folding , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...