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










Publication year range
1.
Nat Biotechnol ; 41(5): 686-697, 2023 05.
Article in English | MEDLINE | ID: mdl-36624149

ABSTRACT

Cytosine base editors (CBEs) enable programmable genomic C·G-to-T·A transition mutations and typically comprise a modified CRISPR-Cas enzyme, a naturally occurring cytidine deaminase, and an inhibitor of uracil repair. Previous studies have shown that CBEs utilizing naturally occurring cytidine deaminases may cause unguided, genome-wide cytosine deamination. While improved CBEs that decrease stochastic genome-wide off-targets have subsequently been reported, these editors can suffer from suboptimal on-target performance. Here, we report the generation and characterization of CBEs that use engineered variants of TadA (CBE-T) that enable high on-target C·G to T·A across a sequence-diverse set of genomic loci, demonstrate robust activity in primary cells and cause no detectable elevation in genome-wide mutation. Additionally, we report cytosine and adenine base editors (CABEs) catalyzing both A-to-I and C-to-U editing (CABE-Ts). Together with ABEs, CBE-Ts and CABE-Ts enable the programmable installation of all transition mutations using laboratory-evolved TadA variants with improved properties relative to previously reported CBEs.


Subject(s)
Cytosine , Gene Editing , Mutation/genetics , Cytidine Deaminase/genetics , Genome , CRISPR-Cas Systems/genetics
2.
Mol Ther ; 29(11): 3125-3139, 2021 11 03.
Article in English | MEDLINE | ID: mdl-34619370

ABSTRACT

The development of CRISPR-derived genome editing technologies has enabled the precise manipulation of DNA sequences within the human genome. In this review, we discuss the initial development and cellular mechanism of action of CRISPR nucleases and DNA base editors. We then describe factors that must be taken into consideration when developing these tools into therapeutic agents, including the potential for unintended and off-target edits when using these genome editing tools, and methods to characterize these types of edits. We finish by considering specific challenges associated with bringing a CRISPR-based therapy to the clinic, including manufacturing, regulatory oversight, and considerations for clinical trials that involve genome editing agents.


Subject(s)
CRISPR-Cas Systems , Gene Editing , Genetic Therapy , Animals , CRISPR-Associated Protein 9 , Clinical Trials as Topic , Clustered Regularly Interspaced Short Palindromic Repeats , Gene Editing/methods , Gene Transfer Techniques , Genetic Engineering , Genetic Therapy/methods , Genetic Therapy/trends , Humans , Models, Animal , RNA, Guide, Kinetoplastida , Recombinational DNA Repair , Translational Research, Biomedical/methods , Translational Research, Biomedical/trends
3.
Curr Opin Struct Biol ; 69: 86-98, 2021 08.
Article in English | MEDLINE | ID: mdl-33964614

ABSTRACT

Since the initial reports describing CRISPR-Cas9, labs across the globe have leveraged this valuable gene editing tool to alter the genomes of living cells. With the goal of generating more precise and efficient genome changes, scientists and engineers have mutated, evolved, and covalently altered Cas9 in order to predictably edit the genetic code. Here, we highlight recent advancements and contributions to the growing field of Cas9 engineering. We present key aspects of Cas9 engineering efforts focused on sgRNA manipulation, PAM-recognition, specificity, deaminase fusions, reverse-transcriptase fusions, and structural rearrangements of this important gene-modifying tool.


Subject(s)
CRISPR-Cas Systems , Gene Editing , CRISPR-Cas Systems/genetics , Humans
4.
Nature ; 593(7859): 429-434, 2021 05.
Article in English | MEDLINE | ID: mdl-34012082

ABSTRACT

Gene-editing technologies, which include the CRISPR-Cas nucleases1-3 and CRISPR base editors4,5, have the potential to permanently modify disease-causing genes in patients6. The demonstration of durable editing in target organs of nonhuman primates is a key step before in vivo administration of gene editors to patients in clinical trials. Here we demonstrate that CRISPR base editors that are delivered in vivo using lipid nanoparticles can efficiently and precisely modify disease-related genes in living cynomolgus monkeys (Macaca fascicularis). We observed a near-complete knockdown of PCSK9 in the liver after a single infusion of lipid nanoparticles, with concomitant reductions in blood levels of PCSK9 and low-density lipoprotein cholesterol of approximately 90% and about 60%, respectively; all of these changes remained stable for at least 8 months after a single-dose treatment. In addition to supporting a 'once-and-done' approach to the reduction of low-density lipoprotein cholesterol and the treatment of atherosclerotic cardiovascular disease (the leading cause of death worldwide7), our results provide a proof-of-concept for how CRISPR base editors can be productively applied to make precise single-nucleotide changes in therapeutic target genes in the liver, and potentially in other organs.


Subject(s)
CRISPR-Cas Systems , Cholesterol, LDL/blood , Gene Editing , Models, Animal , Proprotein Convertase 9/genetics , Adenine/metabolism , Animals , Cells, Cultured , Female , Hepatocytes/metabolism , Humans , Liver/enzymology , Loss of Function Mutation , Macaca fascicularis/blood , Macaca fascicularis/genetics , Male , Mice , Mice, Inbred C57BL , Mutagenesis, Site-Directed , Proprotein Convertase 9/blood , Proprotein Convertase 9/metabolism , Time Factors
6.
Nat Biotechnol ; 38(7): 892-900, 2020 07.
Article in English | MEDLINE | ID: mdl-32284586

ABSTRACT

The foundational adenine base editors (for example, ABE7.10) enable programmable A•T to G•C point mutations but editing efficiencies can be low at challenging loci in primary human cells. Here we further evolve ABE7.10 using a library of adenosine deaminase variants to create ABE8s. At NGG protospacer adjacent motif (PAM) sites, ABE8s result in ~1.5× higher editing at protospacer positions A5-A7 and ~3.2× higher editing at positions A3-A4 and A8-A10 compared with ABE7.10. Non-NGG PAM variants have a ~4.2-fold overall higher on-target editing efficiency than ABE7.10. In human CD34+ cells, ABE8 can recreate a natural allele at the promoter of the γ-globin genes HBG1 and HBG2 with up to 60% efficiency, causing persistence of fetal hemoglobin. In primary human T cells, ABE8s achieve 98-99% target modification, which is maintained when multiplexed across three loci. Delivered as messenger RNA, ABE8s induce no significant levels of single guide RNA (sgRNA)-independent off-target adenine deamination in genomic DNA and very low levels of adenine deamination in cellular mRNA.


Subject(s)
Adenine/metabolism , CRISPR-Cas Systems/genetics , Cytosine/metabolism , RNA, Guide, Kinetoplastida/genetics , Adenosine Deaminase , DNA/genetics , Gene Editing/methods , HEK293 Cells , Humans , Mutation/genetics
7.
Nat Commun ; 11(1): 2052, 2020 04 28.
Article in English | MEDLINE | ID: mdl-32345976

ABSTRACT

Cytosine base editors (CBEs) enable efficient, programmable reversion of T•A to C•G point mutations in the human genome. Recently, cytosine base editors with rAPOBEC1 were reported to induce unguided cytosine deamination in genomic DNA and cellular RNA. Here we report eight next-generation CBEs (BE4 with either RrA3F [wt, F130L], AmAPOBEC1, SsAPOBEC3B [wt, R54Q], or PpAPOBEC1 [wt, H122A, R33A]) that display comparable DNA on-target editing frequencies, whilst eliciting a 12- to 69-fold reduction in C-to-U edits in the transcriptome, and up to a 45-fold overall reduction in unguided off-target DNA deamination relative to BE4 containing rAPOBEC1. Further, no enrichment of genome-wide C•G to T•A edits are observed in mammalian cells following transfection of mRNA encoding five of these next-generation editors. Taken together, these next-generation CBEs represent a collection of base editing tools for applications in which minimized off-target and high on-target activity are required.


Subject(s)
Cytosine/metabolism , DNA/genetics , Gene Editing , RNA/genetics , APOBEC-1 Deaminase/metabolism , Cytosine Deaminase/metabolism , DNA Replication/genetics , Deamination , Genome , HEK293 Cells , Humans , Mutagenesis/genetics , Transcription, Genetic , Transcriptome/genetics
8.
Nat Commun ; 10(1): 3868, 2019 08 27.
Article in English | MEDLINE | ID: mdl-31455765

ABSTRACT

Nonribosomal peptide synthetases (NRPSs) underlie the biosynthesis of many natural products that have important medicinal utility. Protection of the NRPS peptide products from proteolysis is critical to these pathways and is often achieved by structural modification, principally the introduction of D-amino acid residues into the elongating peptide. These amino acids are generally formed in situ from their L-stereoisomers by epimerization domains or dual-function condensation/epimerization domains. In singular contrast, the thioesterase domain of nocardicin biosynthesis mediates both the effectively complete L- to D-epimerization of its C-terminal amino acid residue (≥100:1) and hydrolytic product release. We report herein high-resolution crystal structures of the nocardicin thioesterase domain in ligand-free form and reacted with a structurally precise fluorophosphonate substrate mimic that identify the complete peptide binding pocket to accommodate both stereoisomers. These structures combined with additional functional studies provide detailed mechanistic insight into this unique dual-function NRPS domain.


Subject(s)
Amino Acid Isomerases/metabolism , Bacterial Proteins/metabolism , Hydrolases/metabolism , Lactams/metabolism , Peptide Synthases/metabolism , Amino Acid Isomerases/ultrastructure , Bacterial Proteins/ultrastructure , Crystallography, X-Ray , Hydrolases/ultrastructure , Models, Molecular , Nocardia/enzymology , Organophosphonates/metabolism , Peptide Synthases/ultrastructure , Peptides/metabolism , Protein Structure, Secondary , Stereoisomerism , Substrate Specificity
9.
Nat Biotechnol ; 37(7): 820, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31182863

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

11.
Nat Biotechnol ; 37(6): 626-631, 2019 06.
Article in English | MEDLINE | ID: mdl-31110355

ABSTRACT

Base editing requires that the target sequence satisfy the protospacer adjacent motif requirement of the Cas9 domain and that the target nucleotide be located within the editing window of the base editor. To increase the targeting scope of base editors, we engineered six optimized adenine base editors (ABEmax variants) that use SpCas9 variants compatible with non-NGG protospacer adjacent motifs. To increase the range of target bases that can be modified within the protospacer, we use circularly permuted Cas9 variants to produce four cytosine and four adenine base editors with an editing window expanded from ~4-5 nucleotides to up to ~8-9 nucleotides and reduced byproduct formation. This set of base editors improves the targeting scope of cytosine and adenine base editing.


Subject(s)
CRISPR-Associated Protein 9/genetics , CRISPR-Cas Systems/genetics , Gene Editing/methods , Adenine/chemistry , Cytosine/chemistry , Humans , Nucleotides/chemistry , Nucleotides/genetics , Plasmids/chemistry , Plasmids/genetics
13.
Nature ; 559(7714): E8, 2018 07.
Article in English | MEDLINE | ID: mdl-29720650

ABSTRACT

In this Article, owing to an error during the production process, in Fig. 1a, the dark blue and light blue wedges were incorrectly labelled as 'G•C → T•A' and 'G•C → A•T', instead of 'C•G → T•A' and 'C•G → A•T', respectively. Fig. 1 has been corrected online.

14.
Nature ; 551(7681): 464-471, 2017 11 23.
Article in English | MEDLINE | ID: mdl-29160308

ABSTRACT

The spontaneous deamination of cytosine is a major source of transitions from C•G to T•A base pairs, which account for half of known pathogenic point mutations in humans. The ability to efficiently convert targeted A•T base pairs to G•C could therefore advance the study and treatment of genetic diseases. The deamination of adenine yields inosine, which is treated as guanine by polymerases, but no enzymes are known to deaminate adenine in DNA. Here we describe adenine base editors (ABEs) that mediate the conversion of A•T to G•C in genomic DNA. We evolved a transfer RNA adenosine deaminase to operate on DNA when fused to a catalytically impaired CRISPR-Cas9 mutant. Extensive directed evolution and protein engineering resulted in seventh-generation ABEs that convert targeted A•T base pairs efficiently to G•C (approximately 50% efficiency in human cells) with high product purity (typically at least 99.9%) and low rates of indels (typically no more than 0.1%). ABEs introduce point mutations more efficiently and cleanly, and with less off-target genome modification, than a current Cas9 nuclease-based method, and can install disease-correcting or disease-suppressing mutations in human cells. Together with previous base editors, ABEs enable the direct, programmable introduction of all four transition mutations without double-stranded DNA cleavage.


Subject(s)
Base Pairing/genetics , Gene Editing/methods , Genome, Human/genetics , Adenosine Deaminase/metabolism , CRISPR-Associated Proteins/metabolism , Cell Line, Tumor , DNA/genetics , DNA/metabolism , DNA Cleavage , HEK293 Cells , Humans , Models, Molecular , Polymorphism, Single Nucleotide/genetics
15.
Sci Adv ; 3(8): eaao4774, 2017 08.
Article in English | MEDLINE | ID: mdl-28875174

ABSTRACT

We recently developed base editing, the programmable conversion of target C:G base pairs to T:A without inducing double-stranded DNA breaks (DSBs) or requiring homology-directed repair using engineered fusions of Cas9 variants and cytidine deaminases. Over the past year, the third-generation base editor (BE3) and related technologies have been successfully used by many researchers in a wide range of organisms. The product distribution of base editing-the frequency with which the target C:G is converted to mixtures of undesired by-products, along with the desired T:A product-varies in a target site-dependent manner. We characterize determinants of base editing outcomes in human cells and establish that the formation of undesired products is dependent on uracil N-glycosylase (UNG) and is more likely to occur at target sites containing only a single C within the base editing activity window. We engineered CDA1-BE3 and AID-BE3, which use cytidine deaminase homologs that increase base editing efficiency for some sequences. On the basis of these observations, we engineered fourth-generation base editors (BE4 and SaBE4) that increase the efficiency of C:G to T:A base editing by approximately 50%, while halving the frequency of undesired by-products compared to BE3. Fusing BE3, BE4, SaBE3, or SaBE4 to Gam, a bacteriophage Mu protein that binds DSBs greatly reduces indel formation during base editing, in most cases to below 1.5%, and further improves product purity. BE4, SaBE4, BE4-Gam, and SaBE4-Gam represent the state of the art in C:G-to-T:A base editing, and we recommend their use in future efforts.


Subject(s)
Bacteriophage mu/physiology , Base Pairing , DNA Repair , DNA-Binding Proteins/metabolism , Viral Proteins/metabolism , Cell Line , Enzyme Activation , Gene Frequency , Gene Order , Humans , INDEL Mutation , Uracil-DNA Glycosidase/metabolism
16.
Nature ; 520(7547): 383-7, 2015 Apr 16.
Article in English | MEDLINE | ID: mdl-25624104

ABSTRACT

Non-ribosomal peptide synthetases are giant enzymes composed of modules that house repeated sets of functional domains, which select, activate and couple amino acids drawn from a pool of nearly 500 potential building blocks. The structurally and stereochemically diverse peptides generated in this manner underlie the biosynthesis of a large sector of natural products. Many of their derived metabolites are bioactive such as the antibiotics vancomycin, bacitracin, daptomycin and the ß-lactam-containing penicillins, cephalosporins and nocardicins. Penicillins and cephalosporins are synthesized from a classically derived non-ribosomal peptide synthetase tripeptide (from δ-(L-α-aminoadipyl)-L-cysteinyl-D-valine synthetase). Here we report an unprecedented non-ribosomal peptide synthetase activity that both assembles a serine-containing peptide and mediates its cyclization to the critical ß-lactam ring of the nocardicin family of antibiotics. A histidine-rich condensation domain, which typically performs peptide bond formation during product assembly, also synthesizes the embedded four-membered ring. We propose a mechanism, and describe supporting experiments, that is distinct from the pathways that have evolved to the three other ß-lactam antibiotic families: penicillin/cephalosporins, clavams and carbapenems. These findings raise the possibility that ß-lactam rings can be regio- and stereospecifically integrated into engineered peptides for application as, for example, targeted protease inactivators.


Subject(s)
Anti-Bacterial Agents/biosynthesis , Anti-Bacterial Agents/chemistry , Lactams/chemistry , Lactams/metabolism , Peptide Synthases/metabolism , beta-Lactams/chemistry , beta-Lactams/metabolism , Biocatalysis , Biosynthetic Pathways , Cyclization , Histidine , Serine/metabolism
17.
Nat Chem Biol ; 10(4): 251-8, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24531841

ABSTRACT

Nonribosomal peptide synthetases are versatile engines of bioactive natural product biosynthesis that function according to the multiple carrier thiotemplate mechanism. C-terminal thioesterase (TE) domains of these giant modular proteins typically catalyze product release by hydrolysis or macrocyclization. We now report an unprecedented, dual-function TE that is involved in the biosynthesis of nocardicin A, which is the paradigm monocyclic ß-lactam antibiotic. Contrary to our expectation, a stereodefined series of potential peptide substrates for the nocardicin TE domain failed to undergo hydrolysis. The stringent discrimination against peptide intermediates was overcome by prior monocyclic ß-lactam formation at an L-seryl site. Kinetic data are interpreted such that the TE domain acts as a gatekeeper to hold the assembling peptide on an upstream domain until ß-lactam formation takes place and then rapidly catalyzes epimerization, which has not been observed previously as a TE catalytic function, and thioesterase cleavage to discharge a fully fledged pentapeptide ß-lactam harboring nocardicin G, the universal precursor of the nocardicins.


Subject(s)
Anti-Bacterial Agents/biosynthesis , Lactams/metabolism , Thiolester Hydrolases/metabolism , beta-Lactams/metabolism , Acetylation , Chromatography, High Pressure Liquid , Cloning, Molecular , Coenzyme A/metabolism , Cyclization , DNA/genetics , Genetic Vectors , Hydrolysis , Kinetics , Magnetic Resonance Spectroscopy , Multigene Family , Penicillins/biosynthesis , Peptide Biosynthesis , Phosphorylation , Polymerase Chain Reaction , Stereoisomerism
18.
J Org Chem ; 78(13): 6412-26, 2013 Jul 05.
Article in English | MEDLINE | ID: mdl-23758494

ABSTRACT

Methods have been developed to synthesize tri- and pentapeptide thioesters containing one or more p-(hydroxyphenyl)glycine (pHPG) residues and L-serine, some where the latter is O-phosphorylated, O-acetylated, or exists as a ß-lactam. Selection of orthogonal protection strategies and development of conditions to achieve seryl O-phosphorylation without ß-elimination and to maintain stereochemical control, especially simultaneously at exceptionally base-labile pHPG α-carbons, are described. Intramolecular closure of a seryl peptide to a ß-lactam-containing peptide and the syntheses of corresponding thioester analogues are also reported. Modification of classical Mitsunobu conditions is described in the synthesis of the ß-lactam-containing products, and in a broadly useful observation, it was found that simple exclusion of light from the P(OEt)3-mediated Mitsunobu ring closure afforded yields of >95%, presumably owing to reduced photodegradation of the azodicarboxylate used. These sensitive potential substrates and products will be used in mechanistic studies of the two nonribosomal peptide synthetases NocA and NocB that lie at the heart of nocardicin biosynthesis, a family of monocyclic ß-lactam antibiotics.


Subject(s)
Anti-Bacterial Agents/biosynthesis , Esters/chemical synthesis , Lactams/metabolism , Oligopeptides/chemistry , Oligopeptides/chemical synthesis , Sulfhydryl Compounds/chemical synthesis , Anti-Bacterial Agents/chemistry , Esters/chemistry , Lactams/chemistry , Molecular Structure , Peptide Synthases/chemistry , Peptide Synthases/metabolism , Serine/chemistry , Stereoisomerism , Sulfhydryl Compounds/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...