Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Jul 06.
Article in English | MEDLINE | ID: mdl-39005280

ABSTRACT

Huntington's disease (HD) is an inherited and ultimately fatal neurodegenerative disorder caused by an expanded polyglutamine-encoding CAG repeat within exon 1 of the huntingtin (HTT) gene, which produces a mutant protein that destroys striatal and cortical neurons. Importantly, a critical event in the pathogenesis of HD is the proteolytic cleavage of the mutant HTT protein by caspase-6, which generates fragments of the N-terminal domain of the protein that form highly toxic aggregates. Given the role that proteolysis of the mutant HTT protein plays in HD, strategies for preventing this process hold potential for treating the disorder. By screening 141 CRISPR base editor variants targeting splice elements in the HTT gene, we identified platforms capable of producing HTT protein isoforms resistant to caspase-6-mediated proteolysis via editing of the splice acceptor sequence for exon 13. When delivered to the striatum of a rodent HD model, these base editors induced efficient exon skipping and decreased the formation of the N-terminal fragments, which in turn reduced HTT protein aggregation and attenuated striatal and cortical atrophy. Collectively, these results illustrate the potential for CRISPR base editing to decrease the toxicity of the mutant HTT protein for HD.

2.
bioRxiv ; 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38883727

ABSTRACT

Exon skipping technologies enable exclusion of targeted exons from mature mRNA transcripts, which has broad applications in molecular biology, medicine, and biotechnology. Existing exon skipping techniques include antisense oligonucleotides, targetable nucleases, and base editors, which, while effective for specific applications at some target exons, remain hindered by shortcomings, including transient effects for oligonucleotides, genotoxicity for nucleases and inconsistent exon skipping for base editors. To overcome these limitations, we created SPLICER, a toolbox of next-generation base editors consisting of near-PAMless Cas9 nickase variants fused to adenosine or cytosine deaminases for the simultaneous editing of splice acceptor (SA) and splice donor (SD) sequences. Synchronized SA and SD editing with SPLICER improves exon skipping, reduces aberrant outcomes, including cryptic splicing and intron retention, and enables skipping of exons refractory to single splice-site editing. To demonstrate the therapeutic potential of SPLICER, we targeted APP exon 17, which encodes the amino acid residues that are cleaved to form the Aß plaques in Alzheimer's disease. SPLICER reduced the formation of Aß42 peptides in vitro and enabled efficient exon skipping in a mouse model of Alzheimer's disease. Overall, SPLICER is a widely applicable and efficient toolbox for exon skipping with broad therapeutic applications.

3.
Nat Commun ; 14(1): 6492, 2023 10 14.
Article in English | MEDLINE | ID: mdl-37838698

ABSTRACT

The TDP-43 proteinopathies, which include amyotrophic lateral sclerosis and frontotemporal dementia, are a devastating group of neurodegenerative disorders that are characterized by the mislocalization and aggregation of TDP-43. Here we demonstrate that RNA-targeting CRISPR effector proteins, a programmable class of gene silencing agents that includes the Cas13 family of enzymes and Cas7-11, can be used to mitigate TDP-43 pathology when programmed to target ataxin-2, a modifier of TDP-43-associated toxicity. In addition to inhibiting the aggregation and transit of TDP-43 to stress granules, we find that the in vivo delivery of an ataxin-2-targeting Cas13 system to a mouse model of TDP-43 proteinopathy improved functional deficits, extended survival, and reduced the severity of neuropathological hallmarks. Further, we benchmark RNA-targeting CRISPR platforms against ataxin-2 and find that high-fidelity forms of Cas13 possess improved transcriptome-wide specificity compared to Cas7-11 and a first-generation effector. Our results demonstrate the potential of CRISPR technology for TDP-43 proteinopathies.


Subject(s)
Amyotrophic Lateral Sclerosis , TDP-43 Proteinopathies , Mice , Animals , Ataxin-2/genetics , RNA/metabolism , TDP-43 Proteinopathies/genetics , TDP-43 Proteinopathies/metabolism , TDP-43 Proteinopathies/pathology , Amyotrophic Lateral Sclerosis/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism
4.
bioRxiv ; 2023 Aug 26.
Article in English | MEDLINE | ID: mdl-37066174

ABSTRACT

The TDP-43 proteinopathies, which include amyotrophic lateral sclerosis and frontotemporal dementia, are a devastating group of neurodegenerative disorders that are characterized by the mislocalization and aggregation of TDP-43. Here we demonstrate that RNA-targeting CRISPR effector proteins, a programmable class of gene silencing agents that includes the Cas13 family of enzymes and Cas7-11, can be used to mitigate TDP-43 pathology when programmed to target ataxin-2, a modifier of TDP-43-associated toxicity. In addition to inhibiting the aggregation and transit of TDP-43 to stress granules, we find that the in vivo delivery of an ataxin-2-targeting Cas13 system to a mouse model of TDP-43 proteinopathy improved functional deficits, extended survival, and reduced the severity of neuropathological hallmarks. Further, we benchmark RNA-targeting CRISPR platforms against ataxin-2 and find that high-fidelity forms of Cas13 possess improved transcriptome-wide specificity compared to Cas7-11 and a first-generation effector. Our results demonstrate the potential of CRISPR technology for TDP-43 proteinopathies.

5.
Methods Mol Biol ; 2606: 135-158, 2023.
Article in English | MEDLINE | ID: mdl-36592313

ABSTRACT

CRISPR base editors are genome-modifying proteins capable of creating single-base substitutions in DNA but without the requirement for a DNA double-strand break. Given their ability to precisely edit DNA, they hold tremendous therapeutic potential. Here, we describe procedures for delivering base editors in vivo via adeno-associated virus (AAV) vectors, a promising engineered gene delivery vehicle capable of transducing a range of cell types and tissues. We provide step by step protocols for (i) designing and validating base editing systems, (ii) packaging base editors into recombinant AAV vector particles, (iii) delivering AAV to the central nervous system via intrathecal injection, and (iv) quantifying base editing frequencies by next-generation sequencing.


Subject(s)
Dependovirus , Genetic Vectors , Dependovirus/genetics , Genetic Vectors/genetics , Gene Transfer Techniques , DNA , Genome , CRISPR-Cas Systems
6.
bioRxiv ; 2023 Dec 12.
Article in English | MEDLINE | ID: mdl-38168370

ABSTRACT

An abnormal expansion of a GGGGCC hexanucleotide repeat in the C9ORF72 gene is the most common genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD), two debilitating neurodegenerative disorders driven in part by gain-of-function mechanisms involving transcribed forms of the repeat expansion. By utilizing a Cas13 variant with reduced collateral effects, we developed a high-fidelity RNA-targeting CRISPR-based system for C9ORF72-linked ALS/FTD. When delivered to the brain of a transgenic rodent model, this Cas13-based platform effectively curbed the expression of the GGGGCC repeat-containing RNA without affecting normal C9ORF72 levels, which in turn decreased the formation of RNA foci and reversed transcriptional deficits. This high-fidelity Cas13 variant possessed improved transcriptome-wide specificity compared to its native form and mediated efficient targeting in motor neuron-like cells derived from a patient with ALS. Our results lay the foundation for the implementation of RNA-targeting CRISPR technologies for C9ORF72-linked ALS/FTD.

7.
Mol Ther ; 30(12): 3619-3631, 2022 12 07.
Article in English | MEDLINE | ID: mdl-35965414

ABSTRACT

CRISPR technology has demonstrated broad utility for controlling target gene expression; however, there remains a need for strategies capable of modulating expression via the precise editing of non-coding regulatory elements. Here, we demonstrate that CRISPR base editors, a class of gene-modifying proteins capable of creating single-base substitutions in DNA, can be used to perturb gene expression via their targeted mutagenesis of cis-acting sequences. Using the promoter region of the human huntingtin (HTT) gene as an initial target, we show that editing of the binding site for the transcription factor NF-κB led to a marked reduction in HTT gene expression in base-edited cell populations. We found that these gene perturbations were persistent and specific, as a transcriptome-wide RNA analysis revealed minimal off-target effects resulting from the action of the base editor protein. We further demonstrate that this base-editing platform could influence gene expression in vivo as its delivery to a mouse model of Huntington's disease led to a potent decrease in HTT mRNA in striatal neurons. Finally, to illustrate the applicability of this concept, we target the amyloid precursor protein, showing that multiplex editing of its promoter region significantly perturbed its expression. These findings demonstrate the potential for base editors to regulate target gene expression.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats , Gene Editing , Humans , Animals , Mice
8.
Sci Adv ; 8(3): eabk2485, 2022 01 21.
Article in English | MEDLINE | ID: mdl-35044815

ABSTRACT

Cas13 nucleases are a class of programmable RNA-targeting CRISPR effector proteins that are capable of silencing target gene expression in mammalian cells. Here, we demonstrate that RfxCas13d, a Cas13 ortholog with favorable characteristics to other family members, can be delivered to the mouse spinal cord and brain to silence neurodegeneration-associated genes. Intrathecally delivering an adeno-associated virus vector encoding an RfxCas13d variant programmed to target superoxide dismutase 1 (SOD1), a protein whose mutation can cause amyotrophic lateral sclerosis, reduced SOD1 mRNA and protein in the spinal cord by >50% and improved outcomes in a mouse model of the disorder. We further show that intrastriatally delivering an RfxCas13d variant programmed to target huntingtin (HTT), a protein whose mutation is causative for Huntington's disease, led to a ~50% reduction in HTT protein in the mouse brain. Our results establish RfxCas13d as a versatile platform for knocking down gene expression in the nervous system.


Subject(s)
Amyotrophic Lateral Sclerosis , CRISPR-Cas Systems , Amyotrophic Lateral Sclerosis/genetics , Animals , Gene Silencing , Mammals , Mice , Spinal Cord , Superoxide Dismutase , Superoxide Dismutase-1/genetics
9.
Trends Biotechnol ; 39(7): 692-705, 2021 07.
Article in English | MEDLINE | ID: mdl-33277043

ABSTRACT

The emergence of clustered regularly interspaced short palindromic repeat (CRISPR) nucleases has transformed biotechnology by providing an easy, efficient, and versatile platform for editing DNA. However, traditional CRISPR-based technologies initiate editing by activating DNA double-strand break (DSB) repair pathways, which can cause adverse effects in cells and restrict certain therapeutic applications of the technology. To this end, several new CRISPR-based modalities have been developed that are capable of catalyzing editing without the requirement for a DSB. Here, we review three of these technologies: base editors, prime editors, and RNA-targeting CRISPR-associated protein (Cas)13 effectors. We discuss their strengths compared to traditional gene-modifying systems, we highlight their emerging therapeutic applications, and we examine challenges facing their safe and effective clinical implementation.


Subject(s)
CRISPR-Cas Systems , Gene Editing , Cell- and Tissue-Based Therapy/trends , Endonucleases/genetics , Humans
10.
Mol Ther ; 28(4): 1177-1189, 2020 04 08.
Article in English | MEDLINE | ID: mdl-31991108

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a debilitating and fatal disorder that can be caused by mutations in the superoxide dismutase 1 (SOD1) gene. Although ALS is currently incurable, CRISPR base editors hold the potential to treat the disease through their ability to create nonsense mutations that can permanently disable the expression of the mutant SOD1 gene. However, the restrictive carrying capacity of adeno-associated virus (AAV) vectors has limited their therapeutic application. In this study, we establish an intein-mediated trans-splicing system that enables in vivo delivery of cytidine base editors (CBEs) consisting of the widely used Cas9 protein from Streptococcus pyogenes. We show that intrathecal injection of dual AAV particles encoding a split-intein CBE engineered to trans-splice and introduce a nonsense-coding substitution into a mutant SOD1 gene prolonged survival and markedly slowed the progression of disease in the G93A-SOD1 mouse model of ALS. Adult animals treated by this split-intein CRISPR base editor had a reduced rate of muscle atrophy, decreased muscle denervation, improved neuromuscular function, and up to 40% fewer SOD1 immunoreactive inclusions at end-stage mice compared to control mice. This work expands the capabilities of single-base editors and demonstrates their potential for gene therapy.


Subject(s)
Amyotrophic Lateral Sclerosis/therapy , CRISPR-Associated Protein 9/metabolism , Dependovirus/genetics , Superoxide Dismutase-1/genetics , Amyotrophic Lateral Sclerosis/genetics , Animals , Codon, Nonsense , Disease Models, Animal , Gene Editing , Genetic Vectors/administration & dosage , HEK293 Cells , Humans , Injections, Spinal , Inteins , Male , Mice , Mice, Transgenic , Streptococcus pyogenes/enzymology , Trans-Splicing , Treatment Outcome
11.
Mol Ther Nucleic Acids ; 17: 829-839, 2019 Sep 06.
Article in English | MEDLINE | ID: mdl-31465962

ABSTRACT

Huntington's disease (HD) is a currently incurable and, ultimately, fatal neurodegenerative disorder caused by a CAG trinucleotide repeat expansion within exon 1 of the huntingtin (HTT) gene, which results in the production of a mutant protein that forms inclusions and selectively destroys neurons in the striatum and other adjacent structures. The RNA-guided Cas9 endonuclease from CRISPR-Cas9 systems is a versatile technology for inducing DNA double-strand breaks that can stimulate the introduction of frameshift-inducing mutations and permanently disable mutant gene function. Here, we show that the Cas9 nuclease from Staphylococcus aureus, a small Cas9 ortholog that can be packaged alongside a single guide RNA into a single adeno-associated virus (AAV) vector, can be used to disrupt the expression of the mutant HTT gene in the R6/2 mouse model of HD following its in vivo delivery to the striatum. Specifically, we found that CRISPR-Cas9-mediated disruption of the mutant HTT gene resulted in a ∼50% decrease in neuronal inclusions and significantly improved lifespan and certain motor deficits. These results thus illustrate the potential for CRISPR-Cas9 technology to treat HD and other autosomal dominant neurodegenerative disorders caused by a trinucleotide repeat expansion via in vivo genome editing.

12.
Genome Biol ; 19(1): 143, 2018 09 25.
Article in English | MEDLINE | ID: mdl-30253789

ABSTRACT

Two articles recently described the development of CRISPR technologies that have the potential to fundamentally transform the barcoding and tracing of mammalian cells.


Subject(s)
CRISPR-Cas Systems , Clustered Regularly Interspaced Short Palindromic Repeats , Animals
13.
Methods Mol Biol ; 1867: 253-273, 2018.
Article in English | MEDLINE | ID: mdl-30155829

ABSTRACT

Genome-editing technologies have revolutionized the biomedical sciences by providing researchers with the ability to quickly and efficiently modify genes. While programmable nucleases can be introduced into cells using a variety of techniques, their delivery as purified proteins is an effective approach for limiting off-target effects. Here, we describe step-by-step procedures for manufacturing and delivering genome-modifying proteins-including Cas9 ribonucleoproteins (RNPs) and TALE and zinc-finger nucleases-into mammalian cells. Protocols for combining Cas9 RNP with naturally recombinogenic adeno-associated virus (AAV) donor vectors for the seamless insertion of transgenes by homology-directed genome editing are also provided.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , CRISPR-Associated Protein 9/genetics , Gene Editing/methods , Gene Transfer Techniques , Genetic Vectors/administration & dosage , Transgenes , Zinc Finger Nucleases/genetics , CD4-Positive T-Lymphocytes/cytology , CRISPR-Associated Protein 9/administration & dosage , Cell Differentiation , Cells, Cultured , Genome, Human , Humans , Transcription Activator-Like Effector Nucleases/genetics , Zinc Finger Nucleases/administration & dosage
14.
Stem Cell Reports ; 10(5): 1481-1491, 2018 05 08.
Article in English | MEDLINE | ID: mdl-29628395

ABSTRACT

Huntington disease (HD) is an inherited, progressive neurological disorder characterized by degenerating striatal medium spiny neurons (MSNs). One promising approach for treating HD is cell replacement therapy, where lost cells are replaced by MSN progenitors derived from human pluripotent stem cells (hPSCs). While there has been remarkable progress in generating hPSC-derived MSNs, current production methods rely on two-dimensional culture systems that can include poorly defined components, limit scalability, and yield differing preclinical results. To facilitate clinical translation, here, we generated striatal progenitors from hPSCs within a fully defined and scalable PNIPAAm-PEG three-dimensional (3D) hydrogel. Transplantation of 3D-derived striatal progenitors into a transgenic mouse model of HD slowed disease progression, improved motor coordination, and increased survival. In addition, the transplanted cells developed an MSN-like phenotype and formed synaptic connections with host cells. Our results illustrate the potential of scalable 3D biomaterials for generating striatal progenitors for HD cell therapy.


Subject(s)
Corpus Striatum/pathology , Huntington Disease/pathology , Huntington Disease/therapy , Hydrogels/pharmacology , Pluripotent Stem Cells/transplantation , Action Potentials/drug effects , Animals , Cell Differentiation/drug effects , Cell Survival/drug effects , Disease Models, Animal , Hedgehog Proteins/metabolism , Humans , Mice , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Phenotype , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/drug effects , Wnt Signaling Pathway/drug effects
15.
Curr Opin Biotechnol ; 52: 95-101, 2018 08.
Article in English | MEDLINE | ID: mdl-29626736

ABSTRACT

CRISPR-Cas9 is a versatile tool for genome engineering that has revolutionized biotechnology and is poised to impact medicine. Recent advances in the identification of unique CRISPR systems, as well as the re-engineering of the Cas9 protein for expanded function, has enabled the diversification of the CRISPR genome engineering toolbox. In this review, we highlight these innovations and discuss how advances in CRISPR technology can lead to breakthroughs in the field of gene therapy.


Subject(s)
CRISPR-Cas Systems/genetics , Genetic Techniques , Inventions , Biotechnology , Gene Editing , Genome , Humans
16.
Neuron ; 97(5): 1168-1176.e4, 2018 03 07.
Article in English | MEDLINE | ID: mdl-29478915

ABSTRACT

Rapid eye movement (REM) and non-REM (NREM) sleep are controlled by specific neuronal circuits. Here we show that galanin-expressing GABAergic neurons in the dorsomedial hypothalamus (DMH) comprise separate subpopulations with opposing effects on REM versus NREM sleep. Microendoscopic calcium imaging revealed diverse sleep-wake activity of DMH GABAergic neurons, but the galanin-expressing subset falls into two distinct groups, either selectively activated (REM-on) or suppressed (REM-off) during REM sleep. Retrogradely labeled, preoptic area (POA)-projecting galaninergic neurons are REM-off, whereas the raphe pallidus (RPA)-projecting neurons are primarily REM-on. Bidirectional optogenetic manipulations showed that the POA-projecting neurons promote NREM sleep and suppress REM sleep, while the RPA-projecting neurons have the opposite effects. Thus, REM/NREM switch is regulated antagonistically by DMH galaninergic neurons with intermingled cell bodies but distinct axon projections.


Subject(s)
Hypothalamus/diagnostic imaging , Hypothalamus/physiology , Sleep, REM/physiology , Sleep, Slow-Wave/physiology , Animals , Female , Hypothalamus/chemistry , Male , Mice , Mice, Transgenic , Optogenetics/methods , Random Allocation
17.
Sci Adv ; 3(12): eaar3952, 2017 12.
Article in English | MEDLINE | ID: mdl-29279867

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a fatal and incurable neurodegenerative disease characterized by the progressive loss of motor neurons in the spinal cord and brain. In particular, autosomal dominant mutations in the superoxide dismutase 1 (SOD1) gene are responsible for ~20% of all familial ALS cases. The clustered regularly interspaced short palindromic repeats (CRISPR)-CRISPR-associated (Cas9) genome editing system holds the potential to treat autosomal dominant disorders by facilitating the introduction of frameshift-induced mutations that can disable mutant gene function. We demonstrate that CRISPR-Cas9 can be harnessed to disrupt mutant SOD1 expression in the G93A-SOD1 mouse model of ALS following in vivo delivery using an adeno-associated virus vector. Genome editing reduced mutant SOD1 protein by >2.5-fold in the lumbar and thoracic spinal cord, resulting in improved motor function and reduced muscle atrophy. Crucially, ALS mice treated by CRISPR-mediated genome editing had ~50% more motor neurons at end stage and displayed a ~37% delay in disease onset and a ~25% increase in survival compared to control animals. Thus, this study illustrates the potential for CRISPR-Cas9 to treat SOD1-linked forms of ALS and other central nervous system disorders caused by autosomal dominant mutations.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Gene Editing/methods , Genetic Therapy/methods , Superoxide Dismutase-1/genetics , Amyotrophic Lateral Sclerosis/mortality , Amyotrophic Lateral Sclerosis/therapy , Animals , CRISPR-Associated Protein 9/genetics , CRISPR-Cas Systems , Disease Models, Animal , Female , Genetic Vectors , Genome , Humans , Locomotion , Male , Mice, Transgenic , Mutation , RNA, Guide, Kinetoplastida , Spinal Cord/cytology , Spinal Cord/physiology
18.
Stem Cell Reports ; 8(6): 1770-1783, 2017 06 06.
Article in English | MEDLINE | ID: mdl-28552605

ABSTRACT

Oligodendrocyte precursor cells (OPCs) offer considerable potential for the treatment of demyelinating diseases and injuries of the CNS. However, generating large quantities of high-quality OPCs remains a substantial challenge that impedes their therapeutic application. Here, we show that OPCs can be generated from human pluripotent stem cells (hPSCs) in a three-dimensional (3D), scalable, and fully defined thermoresponsive biomaterial system. We used CRISPR/Cas9 to create a NKX2.2-EGFP human embryonic stem cell reporter line that enabled fine-tuning of early OPC specification and identification of conditions that markedly increased the number of OLIG2+ and NKX2.2+ cells generated from hPSCs. Transplantation of 50-day-old OPCs into the brains of NOD/SCID mice revealed that progenitors generated in 3D without cell selection or purification subsequently engrafted, migrated, and matured into myelinating oligodendrocytes in vivo. These results demonstrate the potential of harnessing lineage reporter lines to develop 3D platforms for rapid and large-scale production of OPCs.


Subject(s)
Cell Differentiation , Oligodendrocyte Precursor Cells/cytology , Pluripotent Stem Cells/cytology , Animals , Biocompatible Materials/chemistry , Brain/metabolism , CRISPR-Cas Systems/genetics , Cell Culture Techniques , Cell Line , Cellular Reprogramming , Genes, Reporter , Homeobox Protein Nkx-2.2 , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Nuclear Proteins , Oligodendrocyte Precursor Cells/metabolism , Oligodendrocyte Precursor Cells/transplantation , Oligodendrocyte Transcription Factor 2/genetics , Oligodendrocyte Transcription Factor 2/metabolism , Pluripotent Stem Cells/metabolism , Tissue Scaffolds/chemistry , Transcription Factors/genetics , Transcription Factors/metabolism , Transplantation, Heterologous , Zebrafish Proteins
19.
Nucleic Acids Res ; 45(11): e98, 2017 Jun 20.
Article in English | MEDLINE | ID: mdl-28334779

ABSTRACT

Realizing the full potential of genome editing requires the development of efficient and broadly applicable methods for delivering programmable nucleases and donor templates for homology-directed repair (HDR). The RNA-guided Cas9 endonuclease can be introduced into cells as a purified protein in complex with a single guide RNA (sgRNA). Such ribonucleoproteins (RNPs) can facilitate the high-fidelity introduction of single-base substitutions via HDR following co-delivery with a single-stranded DNA oligonucleotide. However, combining RNPs with transgene-containing donor templates for targeted gene addition has proven challenging, which in turn has limited the capabilities of the RNP-mediated genome editing toolbox. Here, we demonstrate that combining RNP delivery with naturally recombinogenic adeno-associated virus (AAV) donor vectors enables site-specific gene insertion by homology-directed genome editing. Compared to conventional plasmid-based expression vectors and donor templates, we show that combining RNP and AAV donor delivery increases the efficiency of gene addition by up to 12-fold, enabling the creation of lineage reporters that can be used to track the conversion of striatal neurons from human fibroblasts in real time. These results thus illustrate the potential for unifying nuclease protein delivery with AAV donor vectors for homology-directed genome editing.


Subject(s)
Bacterial Proteins/chemistry , Dependovirus/genetics , Endonucleases/chemistry , Gene Knock-In Techniques , Base Sequence , CRISPR-Associated Protein 9 , Cell Differentiation , Fibroblasts/physiology , Genetic Engineering/methods , Genetic Vectors , Genome, Human , HEK293 Cells , Humans , Neurons/metabolism , Sequence Homology, Nucleic Acid
20.
Article in English | MEDLINE | ID: mdl-27908936

ABSTRACT

Targeted nucleases have provided researchers with the ability to manipulate virtually any genomic sequence, enabling the facile creation of isogenic cell lines and animal models for the study of human disease, and promoting exciting new possibilities for human gene therapy. Here we review three foundational technologies-clustered regularly interspaced short palindromic repeats (CRISPR)-CRISPR-associated protein 9 (Cas9), transcription activator-like effector nucleases (TALENs), and zinc-finger nucleases (ZFNs). We discuss the engineering advances that facilitated their development and highlight several achievements in genome engineering that were made possible by these tools. We also consider artificial transcription factors, illustrating how this technology can complement targeted nucleases for synthetic biology and gene therapy.


Subject(s)
Gene Editing , Animals , Clustered Regularly Interspaced Short Palindromic Repeats , Endonucleases/metabolism , Humans
SELECTION OF CITATIONS
SEARCH DETAIL