Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
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.
Blood ; 140(6): 619-629, 2022 08 11.
Article in English | MEDLINE | ID: mdl-35560156

ABSTRACT

Allogeneic chimeric antigen receptor T-cell (CART) therapies require multiple gene edits to be clinically tractable. Most allogeneic CARTs have been created using gene editing techniques that induce DNA double-stranded breaks (DSBs), resulting in unintended on-target editing outcomes with potentially unforeseen consequences. Cytosine base editors (CBEs) install C•G to T•A point mutations in T cells, with between 90% and 99% efficiency to silence gene expression without creating DSBs, greatly reducing or eliminating undesired editing outcomes following multiplexed editing as compared with clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9). Using CBE, we developed 7CAR8, a CD7-directed allogeneic CART created using 4 simultaneous base edits. We show that CBE, unlike CRISPR-Cas9, does not impact T-cell proliferation, lead to aberrant DNA damage response pathway activation, or result in karyotypic abnormalities following multiplexed editing. We demonstrate 7CAR8 to be highly efficacious against T-cell acute lymphoblastic leukemia (T-ALL) using multiple in vitro and in vivo models. Thus, CBE is a promising technology for applications requiring multiplexed gene editing and can be used to manufacture quadruple-edited 7CAR8 cells, with high potential for clinical translation for relapsed and refractory T-ALL.


Subject(s)
Hematopoietic Stem Cell Transplantation , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma , CRISPR-Cas Systems , Cytosine , Gene Editing/methods , Humans , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics
3.
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
4.
Nat Med ; 26(4): 535-541, 2020 04.
Article in English | MEDLINE | ID: mdl-32284612

ABSTRACT

Base editing by nucleotide deaminases linked to programmable DNA-binding proteins represents a promising approach to permanently remedy blood disorders, although its application in engrafting hematopoietic stem cells (HSCs) remains unexplored. In this study, we purified A3A (N57Q)-BE3 base editor for ribonucleoprotein (RNP) electroporation of human-peripheral-blood-mobilized CD34+ hematopoietic stem and progenitor cells (HSPCs). We observed frequent on-target cytosine base edits at the BCL11A erythroid enhancer at +58 with few indels. Fetal hemoglobin (HbF) induction in erythroid progeny after base editing or nuclease editing was similar. A single therapeutic base edit of the BCL11A enhancer prevented sickling and ameliorated globin chain imbalance in erythroid progeny from sickle cell disease and ß-thalassemia patient-derived HSPCs, respectively. Moreover, efficient multiplex editing could be achieved with combined disruption of the BCL11A erythroid enhancer and correction of the HBB -28A>G promoter mutation. Finally, base edits could be produced in multilineage-repopulating self-renewing human HSCs with high frequency as assayed in primary and secondary recipient animals resulting in potent HbF induction in vivo. Together, these results demonstrate the potential of RNP base editing of human HSPCs as a feasible alternative to nuclease editing for HSC-targeted therapeutic genome modification.


Subject(s)
Anemia, Sickle Cell/pathology , Gene Editing , Genetic Therapy/methods , Hematopoietic Stem Cells/metabolism , Repressor Proteins/genetics , gamma-Globins/genetics , Anemia, Sickle Cell/therapy , Animals , Antigens, CD34/metabolism , CRISPR-Cas Systems , Cells, Cultured , Feasibility Studies , Female , Gene Editing/methods , Gene Targeting/methods , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/pathology , Heterografts , Humans , Mice , Mice, Inbred NOD , Mice, Transgenic , Primary Cell Culture , Repressor Proteins/metabolism , beta-Thalassemia/pathology , beta-Thalassemia/therapy , gamma-Globins/metabolism
6.
Nat Biotechnol ; 36(10): 977-982, 2018 11.
Article in English | MEDLINE | ID: mdl-30059493

ABSTRACT

Base editor technology, which uses CRISPR-Cas9 to direct cytidine deaminase enzymatic activity to specific genomic loci, enables the highly efficient introduction of precise cytidine-to-thymidine DNA alterations. However, existing base editors create unwanted C-to-T alterations when more than one C is present in the enzyme's five-base-pair editing window. Here we describe a strategy for reducing bystander mutations using an engineered human APOBEC3A (eA3A) domain, which preferentially deaminates cytidines in specific motifs according to a TCR>TCY>VCN hierarchy. In direct comparisons with the widely used base editor 3 (BE3) fusion in human cells, our eA3A-BE3 fusion exhibits similar activities on cytidines in TC motifs but greatly reduced editing on cytidines in other sequence contexts. eA3A-BE3 corrects a human ß-thalassemia promoter mutation with much higher (>40-fold) precision than BE3. We also demonstrate that eA3A-BE3 shows reduced mutation frequencies on known off-target sites of BE3, even when targeting promiscuous homopolymeric sites.


Subject(s)
CRISPR-Associated Protein 9/genetics , Cytidine Deaminase/genetics , Gene Editing/methods , Proteins/genetics , Base Sequence , CRISPR-Cas Systems , DNA/genetics , Genome , Humans , Mutation , Promoter Regions, Genetic/genetics , beta-Thalassemia/genetics
7.
Genes Dev ; 29(4): 350-5, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25691465

ABSTRACT

Here we show that the Ino80 chromatin remodeling complex (Ino80C) directly prevents euchromatin from invading transcriptionally silent chromatin within intergenic regions and at the border of euchromatin and heterochromatin. Deletion of Ino80C subunits leads to increased H3K79 methylation and noncoding RNA polymerase II (Pol II) transcription centered at the Ino80C-binding sites. The effect of Ino80C is direct, as it blocks H3K79 methylation by Dot1 in vitro. Heterochromatin stimulates the binding of Ino80C in vitro and in vivo. Our data reveal that Ino80C serves as a general silencing complex that restricts transcription to gene units in euchromatin.


Subject(s)
Chromatin/genetics , Euchromatin/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Binding Sites , Euchromatin/genetics , Gene Expression Regulation, Fungal , Gene Silencing , Histone-Lysine N-Methyltransferase/metabolism , Methylation , Nuclear Proteins/metabolism , Protein Binding , RNA Polymerase II/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...