Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Adv Drug Deliv Rev ; 168: 99-117, 2021 01.
Article in English | MEDLINE | ID: mdl-32931860

ABSTRACT

Genome-editing tools such as Cre recombinase (Cre), zinc-finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs), and most recently the clustered regularly interspaced short palindromic repeat (CRISPR)-associated protein system have revolutionized biomedical research, agriculture, microbial engineering, and therapeutic development. Direct delivery of genome editing enzymes, as opposed to their corresponding DNA and mRNA precursors, is advantageous since they do not require transcription and/or translation. In addition, prolonged overexpression is a problem when delivering viral vector or plasmid DNA which is bypassed when delivering whole proteins. This lowers the risk of insertional mutagenesis and makes for relatively easier manufacturing. However, a major limitation of utilizing genome editing proteins in vivo is their low delivery efficiency, and currently the most successful strategy involves using potentially immunogenic viral vectors. This lack of safe and effective non-viral delivery systems is still a big hurdle for the clinical translation of such enzymes. This review discusses the challenges of non-viral delivery strategies of widely used genome editing enzymes, including Cre recombinase, ZFNs and TALENs, CRISPR/Cas9, and Cas12a (Cpf1) in their protein format and highlights recent innovations of non-viral delivery strategies which have the potential to overcome current delivery limitations and advance the clinical translation of genome editing.


Subject(s)
Gene Editing/methods , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Bacterial Proteins/administration & dosage , CRISPR-Associated Proteins/administration & dosage , Clustered Regularly Interspaced Short Palindromic Repeats , Dendrimers/chemistry , Endodeoxyribonucleases/administration & dosage , Gold/chemistry , Integrases/administration & dosage , Lipids/chemistry , Nanoparticles/chemistry , Phosphorus/chemistry , Polyethyleneimine/chemistry , Transcription Activator-Like Effector Nucleases/administration & dosage , Zinc Finger Nucleases/administration & dosage
2.
Mol Ther ; 27(4): 866-877, 2019 04 10.
Article in English | MEDLINE | ID: mdl-30902585

ABSTRACT

It has previously been shown that engineered zinc finger nucleases (ZFNs) can be packaged into adeno-associated viruses (AAVs) and delivered intravenously into mice, non-human primates, and most recently, humans to induce highly efficient therapeutic genome editing in the liver. Lipid nanoparticles (LNPs) are synthetic delivery vehicles that enable repeat administration and are not limited by the presence of preexisting neutralizing antibodies in patients. Here, we show that mRNA encoding ZFNs formulated into LNP can enable >90% knockout of gene expression in mice by targeting the TTR or PCSK9 gene, at mRNA doses 10-fold lower than has ever been reported. Additionally, co-delivering mRNA-LNP containing ZFNs targeted to intron 1 of the ALB locus with AAV packaged with a promoterless human IDS or FIX therapeutic transgene can result in high levels of targeted integration and subsequent therapeutically relevant levels of protein expression in mice. Finally, we show repeat administration of ZFN mRNA-LNP after a single AAV donor dose results in significantly increased levels of genome editing and transgene expression compared to a single dose. These results demonstrate LNP-mediated ZFN mRNA delivery can drive highly efficient levels of in vivo genome editing and can potentially offer a new treatment modality for a variety of diseases.


Subject(s)
Drug Delivery Systems/methods , Gene Editing/methods , Nanoparticles/administration & dosage , RNA, Messenger/administration & dosage , Zinc Finger Nucleases/administration & dosage , Animals , Cells, Cultured , Dependovirus/genetics , Female , Gene Knockout Techniques , Genetic Vectors , Hepatocytes/metabolism , Introns/genetics , Lipids/chemistry , Male , Mice , Mice, Inbred C57BL , Prealbumin/genetics , Proprotein Convertase 9/genetics , RNA, Messenger/genetics , Transgenes/genetics , Zinc Finger Nucleases/pharmacology
3.
Methods Mol Biol ; 1867: 215-228, 2018.
Article in English | MEDLINE | ID: mdl-30155826

ABSTRACT

Mitochondrial diseases often result from mutations in the mitochondrial genome (mtDNA). In most cases, mutant mtDNA coexists with wild-type mtDNA, resulting in heteroplasmy. One potential future approach to treat heteroplasmic mtDNA diseases is the specific elimination of pathogenic mtDNA mutations, lowering the level of mutant mtDNA below pathogenic thresholds. Mitochondrially targeted zinc-finger nucleases (mtZFNs) have been demonstrated to specifically target and introduce double-strand breaks in mutant mtDNA, facilitating substantial shifts in heteroplasmy. One application of mtZFN technology, in the context of heteroplasmic mtDNA disease, is delivery into the heteroplasmic oocyte or early embryo to eliminate mutant mtDNA, preventing transmission of mitochondrial diseases through the germline. Here we describe a protocol for efficient production of mtZFN mRNA in vitro, and delivery of these into 0.5 dpc mouse embryos to elicit shifts of mtDNA heteroplasmy.


Subject(s)
DNA Breaks, Double-Stranded , DNA, Mitochondrial/genetics , Embryo, Mammalian/metabolism , Gene Transfer Techniques , Mitochondria/enzymology , Mutation , Zinc Finger Nucleases/administration & dosage , Animals , Cells, Cultured , Embryo, Mammalian/cytology , Female , Genome, Mitochondrial , Male , Mice , Mice, Inbred C57BL , Zinc Finger Nucleases/genetics , Zinc Finger Nucleases/metabolism
4.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...