Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
2.
Nat Commun ; 12(1): 3908, 2021 06 23.
Article in English | MEDLINE | ID: mdl-34162850

ABSTRACT

Though AsCas12a fills a crucial gap in the current genome editing toolbox, it exhibits relatively poor editing efficiency, restricting its overall utility. Here we isolate an engineered variant, "AsCas12a Ultra", that increased editing efficiency to nearly 100% at all sites examined in HSPCs, iPSCs, T cells, and NK cells. We show that AsCas12a Ultra maintains high on-target specificity thereby mitigating the risk for off-target editing and making it ideal for complex therapeutic genome editing applications. We achieved simultaneous targeting of three clinically relevant genes in T cells at >90% efficiency and demonstrated transgene knock-in efficiencies of up to 60%. We demonstrate site-specific knock-in of a CAR in NK cells, which afforded enhanced anti-tumor NK cell recognition, potentially enabling the next generation of allogeneic cell-based therapies in oncology. AsCas12a Ultra is an advanced CRISPR nuclease with significant advantages in basic research and in the production of gene edited cell medicines.


Subject(s)
Acidaminococcus/enzymology , Bacterial Proteins/metabolism , CRISPR-Associated Proteins/metabolism , CRISPR-Cas Systems , Endonucleases/metabolism , Gene Editing/methods , Acidaminococcus/genetics , Bacterial Proteins/genetics , CRISPR-Associated Proteins/genetics , Cells, Cultured , Endonucleases/genetics , HEK293 Cells , Hematopoietic Stem Cells/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Jurkat Cells , Killer Cells, Natural/metabolism , Reproducibility of Results , T-Lymphocytes/metabolism
3.
CRISPR J ; 3(3): 177-187, 2020 06.
Article in English | MEDLINE | ID: mdl-32584143

ABSTRACT

Multiplexed genome editing with DNA endonucleases has broad application, including for cellular therapies, but chromosomal translocations, natural byproducts of inducing simultaneous genomic breaks, have not been explored in detail. Here we apply various CRISPR-Cas nucleases to edit the T cell receptor alpha and beta 2 microglobulin genes in human primary T cells and comprehensively evaluate the frequency and stability of the resulting translocations. A thorough translocation frequency analysis using three orthogonal methods (droplet digital PCR, unidirectional sequencing, and metaphase fluorescence in situ hybridization) yielded comparable results and an overall translocation rate of ∼7% between two simultaneous CRISPR-Cas9 induced edits. In addition, we show that chromosomal translocations can be reduced when using different nuclease combinations, or by the presence of a homologous single stranded oligo donor for multiplexed genome editing. Importantly, the two different approaches for translocation reduction are compatible with cell therapy applications.


Subject(s)
CRISPR-Cas Systems , Gene Editing/methods , T-Lymphocytes , Translocation, Genetic , CD4-Positive T-Lymphocytes , CRISPR-Associated Protein 9/genetics , Clustered Regularly Interspaced Short Palindromic Repeats , DNA/genetics , Endonucleases/genetics , Genome, Human , Humans , In Situ Hybridization, Fluorescence , Multifactorial Inheritance , RNA, Guide, Kinetoplastida , Streptococcus pyogenes
4.
PLoS One ; 15(4): e0231716, 2020.
Article in English | MEDLINE | ID: mdl-32298334

ABSTRACT

RNA-guided endonucleases such as Cas9 provide efficient on-target genome editing in cells but may also cleave at off-target loci throughout the genome. Engineered variants of Streptococcus pyogenes Cas9 (SpCas9) have been developed to globally reduce off-target activity, but individual off-targets may remain, or on-target activity may be compromised. In order to evolve against activity at specific off-targets while maintaining strong on-target editing, we developed a novel M13 bacteriophage-mediated selection method. Using this method, sequential rounds of positive and negative selection are used to identify mutations to Cas9 that enhance or diminish editing activity at particular genomic sequences. We also introduce scanning mutagenesis of oligo-directed targets (SMOOT), a comprehensive mutagenesis method to create highly diverse libraries of Cas9 variants that can be challenged with phage-based selection. Our platform identifies novel SpCas9 mutants which mitigate cleavage against off-targets both in biochemical assays and in T-cells while maintaining higher on-target activity than previously described variants. We describe an evolved variant, S. pyogenes Adapted to Reduce Target Ambiguity Cas9 (SpartaCas), composed of the most enriched mutations, each of unknown function. This evolved Cas9 mutant reduces off-target cleavage while preserving efficient editing at multiple therapeutically relevant targets. Directed evolution of Cas9 using our system demonstrates an improved structure-independent methodology to effectively engineer nuclease activity.


Subject(s)
Bacteriophage M13/genetics , CRISPR-Associated Protein 9/genetics , CRISPR-Associated Protein 9/metabolism , Directed Molecular Evolution/methods , Gene Editing/methods , Streptococcus pyogenes/genetics , CRISPR-Associated Protein 9/chemistry , High-Throughput Screening Assays , Mutagenesis , Mutation , Streptococcus pyogenes/enzymology , Substrate Specificity , T-Lymphocytes/metabolism
5.
CRISPR J ; 2: 172-185, 2019 06.
Article in English | MEDLINE | ID: mdl-31225747

ABSTRACT

Considerable effort has been devoted to developing a comprehensive understanding of CRISPR nuclease specificity. In silico predictions and multiple genome-wide cellular and biochemical approaches have revealed a basic understanding of the Cas9 specificity profile. However, none of these approaches has delivered a model that allows accurate prediction of a CRISPR nuclease's ability to cleave a site based entirely on the sequence of the guide RNA (gRNA) and the target. We describe a library-based biochemical assay that directly reports the cleavage efficiency of a particular Cas9-guide complex by measuring both uncleaved and cleaved target molecules over a wide range of mismatched library members. We applied our assay using libraries of targets to evaluate the specificity of Staphylococcus aureus Cas9 under a variety of experimental conditions. Surprisingly, our data show an unexpectedly high variation in the random gRNA:target DNA mismatch tolerance when cleaving with different gRNAs, indicating guide-intrinsic mismatch permissiveness and challenging the assumption of universal specificity models. We use data generated by our assay to create the first off-target, guide-specific cleavage models. The barcoded libraries of targets approach is rapid, highly modular, and capable of generating protein- and guide-specific models, as well as illuminating the biophysics of Cas9 binding versus cutting. These models may be useful in identifying potential off-targets, and the gRNA-intrinsic nature of mismatch tolerance argues for coupling these specificity models with orthogonal methods for a more complete assessment of gRNA specificity.


Subject(s)
CRISPR-Associated Protein 9/metabolism , Staphylococcus aureus/enzymology , CRISPR-Cas Systems , DNA/metabolism , Substrate Specificity
6.
Nat Med ; 25(2): 229-233, 2019 02.
Article in English | MEDLINE | ID: mdl-30664785

ABSTRACT

Leber congenital amaurosis type 10 is a severe retinal dystrophy caused by mutations in the CEP290 gene1,2. We developed EDIT-101, a candidate genome-editing therapeutic, to remove the aberrant splice donor created by the IVS26 mutation in the CEP290 gene and restore normal CEP290 expression. Key to this therapeutic, we identified a pair of Staphylococcus aureus Cas9 guide RNAs that were highly active and specific to the human CEP290 target sequence. In vitro experiments in human cells and retinal explants demonstrated the molecular mechanism of action and nuclease specificity. Subretinal delivery of EDIT-101 in humanized CEP290 mice showed rapid and sustained CEP290 gene editing. A comparable surrogate non-human primate (NHP) vector also achieved productive editing of the NHP CEP290 gene at levels that met the target therapeutic threshold, and demonstrated the ability of CRISPR/Cas9 to edit somatic primate cells in vivo. These results support further development of EDIT-101 for LCA10 and additional CRISPR-based medicines for other inherited retinal disorders.


Subject(s)
Gene Editing , Leber Congenital Amaurosis/genetics , Leber Congenital Amaurosis/physiopathology , Animals , Cell Line , Gene Knock-In Techniques , Humans , Mice , Primates , Reproducibility of Results , Vision, Ocular
8.
Nat Commun ; 9(1): 3542, 2018 08 28.
Article in English | MEDLINE | ID: mdl-30154463

ABSTRACT

The original HTML version of this Article incorrectly listed an affiliation of Josh Tycko as 'Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA', instead of the correct 'Present address: Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA'. It also incorrectly listed an affiliation of this author as 'Present address: Arrakis Therapeutics, 35 Gatehouse Dr., Waltham, MA, 02451, USA'.The original HTML version incorrectly listed an affiliation of Luis A. Barrera as 'Present address: Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, 06511, USA', instead of the correct 'Present address: Arrakis Therapeutics, 35 Gatehouse Dr., Waltham, MA 02451, USA'.Finally, the original HTML version incorrectly omitted an affiliation of Nicholas C. Huston: 'Present address: Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06511, USA'.This has been corrected in the HTML version of the Article. The PDF version was correct from the time of publication.

9.
Nat Commun ; 9(1): 2962, 2018 07 27.
Article in English | MEDLINE | ID: mdl-30054474

ABSTRACT

Therapeutic genome editing with Staphylococcus aureus Cas9 (SaCas9) requires a rigorous understanding of its potential off-target activity in the human genome. Here we report a high-throughput screening approach to measure SaCas9 genome editing variation in human cells across a large repertoire of 88,692 single guide RNAs (sgRNAs) paired with matched or mismatched target sites in a synthetic cassette. We incorporate randomized barcodes that enable whitelisting of correctly synthesized molecules for further downstream analysis, in order to circumvent the limitation of oligonucleotide synthesis errors. We find SaCas9 sgRNAs with 21-mer or 22-mer spacer sequences are generally more active, although high efficiency 20-mer spacers are markedly less tolerant of mismatches. Using this dataset, we developed an SaCas9 specificity model that performs robustly in ranking off-target sites. The barcoded pairwise library screen enabled high-fidelity recovery of guide-target relationships, providing a scalable framework for the investigation of CRISPR enzyme properties and general nucleic acid interactions.


Subject(s)
CRISPR-Associated Protein 9/genetics , Gene Editing/methods , Gene Library , Staphylococcus aureus/genetics , Bacterial Proteins/genetics , Base Sequence , CRISPR-Cas Systems , Cloning, Molecular , Clustered Regularly Interspaced Short Palindromic Repeats , Genes, Bacterial/genetics , HEK293 Cells , Humans , RNA, Guide, Kinetoplastida/genetics
11.
BMC Genomics ; 19(1): 212, 2018 03 21.
Article in English | MEDLINE | ID: mdl-29562890

ABSTRACT

BACKGROUND: Understanding the diversity of repair outcomes after introducing a genomic cut is essential for realizing the therapeutic potential of genomic editing technologies. Targeted PCR amplification combined with Next Generation Sequencing (NGS) or enzymatic digestion, while broadly used in the genome editing field, has critical limitations for detecting and quantifying structural variants such as large deletions (greater than approximately 100 base pairs), inversions, and translocations. RESULTS: To overcome these limitations, we have developed a Uni-Directional Targeted Sequencing methodology, UDiTaS, that is quantitative, removes biases associated with variable-length PCR amplification, and can measure structural changes in addition to small insertion and deletion events (indels), all in a single reaction. We have applied UDiTaS to a variety of samples, including those treated with a clinically relevant pair of S. aureus Cas9 single guide RNAs (sgRNAs) targeting CEP290, and a pair of S. pyogenes Cas9 sgRNAs at T-cell relevant loci. In both cases, we have simultaneously measured small and large edits, including inversions and translocations, exemplifying UDiTaS as a valuable tool for the analysis of genome editing outcomes. CONCLUSIONS: UDiTaS is a robust and streamlined sequencing method useful for measuring small indels as well as structural rearrangements, like translocations, in a single reaction. UDiTaS is especially useful for pre-clinical and clinical application of gene editing to measure on- and off-target editing, large and small.


Subject(s)
CRISPR-Cas Systems , Gene Editing , Gene Rearrangement , Genome, Human , INDEL Mutation , Osteosarcoma/diagnosis , Antigens, Neoplasm/genetics , Bone Neoplasms/diagnosis , Bone Neoplasms/genetics , Cell Cycle Proteins , Cells, Cultured , Cytoskeletal Proteins , Genomics/methods , Humans , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Osteosarcoma/genetics , Sequence Deletion , T-Lymphocytes/metabolism , T-Lymphocytes/pathology
12.
Nat Commun ; 8: 13905, 2017 01 09.
Article in English | MEDLINE | ID: mdl-28067217

ABSTRACT

The CRISPR-Cas9 system provides a versatile toolkit for genome engineering that can introduce various DNA lesions at specific genomic locations. However, a better understanding of the nature of these lesions and the repair pathways engaged is critical to realizing the full potential of this technology. Here we characterize the different lesions arising from each Cas9 variant and the resulting repair pathway engagement. We demonstrate that the presence and polarity of the overhang structure is a critical determinant of double-strand break repair pathway choice. Similarly, single nicks deriving from different Cas9 variants differentially activate repair: D10A but not N863A-induced nicks are repaired by homologous recombination. Finally, we demonstrate that homologous recombination is required for repairing lesions using double-stranded, but not single-stranded DNA as a template. This detailed characterization of repair pathway choice in response to CRISPR-Cas9 enables a more deterministic approach for designing research and therapeutic genome engineering strategies.


Subject(s)
BRCA2 Protein/genetics , CRISPR-Cas Systems , DNA/genetics , Gene Editing/methods , Genome, Human , Rad51 Recombinase/genetics , Recombinational DNA Repair , BRCA2 Protein/antagonists & inhibitors , BRCA2 Protein/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , CRISPR-Associated Protein 9 , Cell Line, Tumor , Clustered Regularly Interspaced Short Palindromic Repeats , DNA/metabolism , DNA Breaks, Double-Stranded , Endonucleases/genetics , Endonucleases/metabolism , HEK293 Cells , Humans , K562 Cells , Osteoblasts/cytology , Osteoblasts/metabolism , RNA, Guide, Kinetoplastida/genetics , RNA, Guide, Kinetoplastida/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rad51 Recombinase/antagonists & inhibitors , Rad51 Recombinase/metabolism
13.
Mol Cell ; 63(3): 355-70, 2016 08 04.
Article in English | MEDLINE | ID: mdl-27494557

ABSTRACT

Advances in the development of delivery, repair, and specificity strategies for the CRISPR-Cas9 genome engineering toolbox are helping researchers understand gene function with unprecedented precision and sensitivity. CRISPR-Cas9 also holds enormous therapeutic potential for the treatment of genetic disorders by directly correcting disease-causing mutations. Although the Cas9 protein has been shown to bind and cleave DNA at off-target sites, the field of Cas9 specificity is rapidly progressing, with marked improvements in guide RNA selection, protein and guide engineering, novel enzymes, and off-target detection methods. We review important challenges and breakthroughs in the field as a comprehensive practical guide to interested users of genome editing technologies, highlighting key tools and strategies for optimizing specificity. The genome editing community should now strive to standardize such methods for measuring and reporting off-target activity, while keeping in mind that the goal for specificity should be continued improvement and vigilance.


Subject(s)
CRISPR-Associated Proteins/metabolism , CRISPR-Cas Systems , DNA/metabolism , Endonucleases/metabolism , Gene Editing/methods , Gene Targeting/methods , Genomics/methods , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , CRISPR-Associated Proteins/genetics , Computational Biology , DNA/genetics , Endonucleases/genetics , Humans , Kinetics , Mutation , Protein Engineering , RNA, Guide, Kinetoplastida/genetics , RNA, Guide, Kinetoplastida/metabolism , Substrate Specificity
16.
PLoS One ; 10(9): e0138486, 2015.
Article in English | MEDLINE | ID: mdl-26378449

ABSTRACT

Death Receptor 5 (DR5) agonists demonstrate anti-tumor activity in preclinical models but have yet to demonstrate robust clinical responses. A key limitation may be the lack of patient selection strategies to identify those most likely to respond to treatment. To overcome this limitation, we screened a DR5 agonist Nanobody across >600 cell lines representing 21 tumor lineages and assessed molecular features associated with response. High expression of DR5 and Casp8 were significantly associated with sensitivity, but their expression thresholds were difficult to translate due to low dynamic ranges. To address the translational challenge of establishing thresholds of gene expression, we developed a classifier based on ratios of genes that predicted response across lineages. The ratio classifier outperformed the DR5+Casp8 classifier, as well as standard approaches for feature selection and classification using genes, instead of ratios. This classifier was independently validated using 11 primary patient-derived pancreatic xenograft models showing perfect predictions as well as a striking linearity between prediction probability and anti-tumor response. A network analysis of the genes in the ratio classifier captured important biological relationships mediating drug response, specifically identifying key positive and negative regulators of DR5 mediated apoptosis, including DR5, CASP8, BID, cFLIP, XIAP and PEA15. Importantly, the ratio classifier shows translatability across gene expression platforms (from Affymetrix microarrays to RNA-seq) and across model systems (in vitro to in vivo). Our approach of using gene expression ratios presents a robust and novel method for constructing translatable biomarkers of compound response, which can also probe the underlying biology of treatment response.


Subject(s)
Cell Lineage/genetics , Gene Expression Regulation, Neoplastic/genetics , Gene Expression/genetics , Pancreatic Neoplasms/genetics , Protein Biosynthesis/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Animals , Apoptosis/genetics , Caspase 8/genetics , Cell Line, Tumor , Humans , Mice , Xenograft Model Antitumor Assays/methods
17.
Chem Biol ; 22(9): 1228-37, 2015 Sep 17.
Article in English | MEDLINE | ID: mdl-26364931

ABSTRACT

In an attempt to identify novel therapeutics and mechanisms to differentially kill tumor cells using phenotypic screening, we identified N-benzyl indole carbinols (N-BICs), synthetic analogs of the natural product indole-3-carbinol (I3C). To understand the mode of action for the molecules we employed Cancer Cell Line Encyclopedia viability profiling and correlative informatics analysis to identify and ultimately confirm the phase II metabolic enzyme sulfotransferase 1A1 (SULT1A1) as the essential factor for compound selectivity. Further studies demonstrate that SULT1A1 activates the N-BICs by rendering the compounds strong electrophiles which can alkylate cellular proteins and thereby induce cell death. This study demonstrates that the selectivity profile for N-BICs is through conversion by SULT1A1 from an inactive prodrug to an active species that induces cell death and tumor suppression.


Subject(s)
Arylsulfotransferase/metabolism , Benzyl Compounds/pharmacology , Indoles/pharmacology , Animals , Benzyl Compounds/pharmacokinetics , Cell Line, Tumor , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Female , HCT116 Cells , Humans , Indoles/pharmacokinetics , Mice , Mice, Nude , Random Allocation , Xenograft Model Antitumor Assays
19.
Nat Chem Biol ; 11(7): 511-7, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26030728

ABSTRACT

Spinal muscular atrophy (SMA), which results from the loss of expression of the survival of motor neuron-1 (SMN1) gene, represents the most common genetic cause of pediatric mortality. A duplicate copy (SMN2) is inefficiently spliced, producing a truncated and unstable protein. We describe herein a potent, orally active, small-molecule enhancer of SMN2 splicing that elevates full-length SMN protein and extends survival in a severe SMA mouse model. We demonstrate that the molecular mechanism of action is via stabilization of the transient double-strand RNA structure formed by the SMN2 pre-mRNA and U1 small nuclear ribonucleic protein (snRNP) complex. The binding affinity of U1 snRNP to the 5' splice site is increased in a sequence-selective manner, discrete from constitutive recognition. This new mechanism demonstrates the feasibility of small molecule-mediated, sequence-selective splice modulation and the potential for leveraging this strategy in other splicing diseases.


Subject(s)
Alternative Splicing , Muscular Atrophy, Spinal/drug therapy , RNA, Double-Stranded/agonists , Ribonucleoprotein, U1 Small Nuclear/agonists , Small Molecule Libraries/pharmacology , Survival of Motor Neuron 2 Protein/metabolism , Animals , Binding Sites , Disease Models, Animal , Female , Gene Expression , Humans , Mice , Mice, Transgenic , Models, Molecular , Muscular Atrophy, Spinal/metabolism , Muscular Atrophy, Spinal/mortality , Muscular Atrophy, Spinal/pathology , Protein Binding/drug effects , Protein Stability/drug effects , Proteolysis , RNA Precursors/agonists , RNA Precursors/chemistry , RNA Precursors/metabolism , RNA, Double-Stranded/chemistry , RNA, Double-Stranded/metabolism , Ribonucleoprotein, U1 Small Nuclear/chemistry , Ribonucleoprotein, U1 Small Nuclear/metabolism , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/metabolism , Survival Analysis , Survival of Motor Neuron 2 Protein/chemistry , Survival of Motor Neuron 2 Protein/genetics
20.
Nat Cell Biol ; 16(11): 1069-79, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25327288

ABSTRACT

Cells rely on autophagy to clear misfolded proteins and damaged organelles to maintain cellular homeostasis. In this study we use the new autophagy inhibitor PIK-III to screen for autophagy substrates. PIK-III is a selective inhibitor of VPS34 that binds a unique hydrophobic pocket not present in related kinases such as PI(3)Kα. PIK-III acutely inhibits autophagy and de novo lipidation of LC3, and leads to the stabilization of autophagy substrates. By performing ubiquitin-affinity proteomics on PIK-III-treated cells we identified substrates including NCOA4, which accumulates in ATG7-deficient cells and co-localizes with autolysosomes. NCOA4 directly binds ferritin heavy chain-1 (FTH1) to target the iron-binding ferritin complex with a relative molecular mass of 450,000 to autolysosomes following starvation or iron depletion. Interestingly, Ncoa4(-/-) mice exhibit a profound accumulation of iron in splenic macrophages, which are critical for the reutilization of iron from engulfed red blood cells. Taken together, the results of this study provide a new mechanism for selective autophagy of ferritin and reveal a previously unappreciated role for autophagy and NCOA4 in the control of iron homeostasis in vivo.


Subject(s)
Autophagy/physiology , Class III Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Ferritins/metabolism , Homeostasis/physiology , Iron/metabolism , Nuclear Receptor Coactivators/metabolism , Animals , Autophagy/drug effects , Cells, Cultured , Humans , Lysosomes/metabolism , Mice , Phagosomes/metabolism , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL
...