Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 3 de 3
1.
Cell Rep Med ; : 101619, 2024 Jun 11.
Article En | MEDLINE | ID: mdl-38897206

Liver-directed adeno-associated viral (AAV) vector-mediated homology-independent targeted integration (AAV-HITI) by CRISPR-Cas9 at the highly transcribed albumin locus is under investigation to provide sustained transgene expression following neonatal treatment. We show that targeting the 3' end of the albumin locus results in productive integration in about 15% of mouse hepatocytes achieving therapeutic levels of systemic proteins in two mouse models of inherited diseases. We demonstrate that full-length HITI donor DNA is preferentially integrated upon nuclease cleavage and that, despite partial AAV genome integrations in the target locus, no gross chromosomal rearrangements or insertions/deletions at off-target sites are found. In line with this, no evidence of hepatocellular carcinoma is observed within the 1-year follow-up. Finally, AAV-HITI is effective at vector doses considered safe if directly translated to humans providing therapeutic efficacy in the adult liver in addition to newborn. Overall, our data support the development of this liver-directed AAV-based knockin strategy.

2.
Mol Ther ; 32(5): 1298-1310, 2024 May 01.
Article En | MEDLINE | ID: mdl-38459694

Undesired on- and off-target effects of CRISPR-Cas nucleases remain a challenge in genome editing. While the use of Cas9 nickases has been shown to minimize off-target mutagenesis, their use in therapeutic genome editing has been hampered by a lack of efficacy. To overcome this limitation, we and others have developed double-nickase-based strategies to generate staggered DNA double-strand breaks to mediate gene disruption or gene correction with high efficiency. However, the impact of paired single-strand nicks on genome integrity has remained largely unexplored. Here, we developed a novel CAST-seq pipeline, dual CAST, to characterize chromosomal aberrations induced by paired CRISPR-Cas9 nickases at three different loci in primary keratinocytes derived from patients with epidermolysis bullosa. While targeting COL7A1, COL17A1, or LAMA3 with Cas9 nucleases caused previously undescribed chromosomal rearrangements, no chromosomal translocations were detected following paired-nickase editing. While the double-nicking strategy induced large deletions/inversions within a 10 kb region surrounding the target sites at all three loci, similar to the nucleases, the chromosomal on-target aberrations were qualitatively different and included a high proportion of insertions. Taken together, our data indicate that double-nickase approaches combine efficient editing with greatly reduced off-target effects but still leave substantial chromosomal aberrations at on-target sites.


CRISPR-Cas Systems , Deoxyribonuclease I , Gene Editing , Keratinocytes , Humans , Gene Editing/methods , Deoxyribonuclease I/metabolism , Deoxyribonuclease I/genetics , Keratinocytes/metabolism , DNA Breaks, Double-Stranded , Chromosome Aberrations , Collagen Type VII/genetics , Collagen Type VII/metabolism , Cells, Cultured
3.
Viruses ; 13(7)2021 07 12.
Article En | MEDLINE | ID: mdl-34372550

Persistent hepatitis B virus (HBV) infection remains a serious medical problem worldwide, with an estimated global burden of 257 million carriers. Prophylactic and therapeutic interventions, in the form of a vaccine, immunomodulators, and nucleotide and nucleoside analogs, are available. Vaccination, however, offers no therapeutic benefit to chronic sufferers and has had a limited impact on infection rates. Although immunomodulators and nucleotide and nucleoside analogs have been licensed for treatment of chronic HBV, cure rates remain low. Transcription activator-like effector nucleases (TALENs) designed to bind and cleave viral DNA offer a novel therapeutic approach. Importantly, TALENs can target covalently closed circular DNA (cccDNA) directly with the potential of permanently disabling this important viral replicative intermediate. Potential off-target cleavage by engineered nucleases leading to toxicity presents a limitation of this technology. To address this, in the context of HBV gene therapy, existing TALENs targeting the viral core and surface open reading frames were modified with second- and third-generation FokI nuclease domains. As obligate heterodimers these TALENs prevent target cleavage as a result of FokI homodimerization. Second-generation obligate heterodimeric TALENs were as effective at silencing viral gene expression as first-generation counterparts and demonstrated an improved specificity in a mouse model of HBV replication.


Hepatitis B virus/genetics , Hepatitis B/drug therapy , Transcription Activator-Like Effector Nucleases/genetics , Animals , Animals, Outbred Strains , Antiviral Agents/therapeutic use , Cell Line , DNA Viruses/genetics , DNA, Circular , DNA, Viral/genetics , Deoxyribonucleases, Type II Site-Specific/genetics , Deoxyribonucleases, Type II Site-Specific/metabolism , Disease Models, Animal , Endonucleases/genetics , Female , Genetic Therapy/methods , HEK293 Cells , Hep G2 Cells , Hepatitis B/genetics , Hepatitis B/immunology , Hepatitis B, Chronic/genetics , Hepatitis B, Chronic/virology , Humans , Mice , Transcription Activator-Like Effector Nucleases/therapeutic use , Virus Replication/genetics
...