Genetic Engineering Publications - GEG Tech top picks
36.6K views | +0 today
Follow
Genetic Engineering Publications - GEG Tech top picks
Your new post is loading...
Your new post is loading...
Scooped by BigField GEG Tech
Scoop.it!

An RNA-targeting CRISPR–Cas13d system alleviates disease-related phenotypes in Huntington’s disease models | Nature Neuroscience

An RNA-targeting CRISPR–Cas13d system alleviates disease-related phenotypes in Huntington’s disease models | Nature Neuroscience | Genetic Engineering Publications - GEG Tech top picks | Scoop.it
Huntington’s disease (HD) is a fatal, dominantly inherited neurodegenerative disorder caused by CAG trinucleotide expansion in exon 1 of the huntingtin (HTT) gene. Since the reduction of pathogenic mutant HTT messenger RNA is therapeutic, we developed a mutant allele-sensitive CAGEX RNA-targeting CRISPR–Cas13d system (Cas13d–CAGEX) that eliminates toxic CAGEX RNA in fibroblasts derived from patients with HD and induced pluripotent stem cell-derived neurons. We show that intrastriatal delivery of Cas13d–CAGEX via an adeno-associated viral vector selectively reduces mutant HTT mRNA and protein levels in the striatum of heterozygous zQ175 mice, a model of HD. This also led to improved motor coordination, attenuated striatal atrophy and reduction of mutant HTT protein aggregates. These phenotypic improvements lasted for at least eight months without adverse effects and with minimal off-target transcriptomic effects. Taken together, we demonstrate proof of principle of an RNA-targeting CRISPR–Cas13d system as a therapeutic approach for HD, a strategy with implications for the treatment of other dominantly inherited disorders. Leveraging RNA-targeting CRISPR–Cas13d technology, Morelli et al. engineered a novel therapeutic strategy that safely and effectively eliminates toxic expanded huntingtin RNA in multiple models of Huntington’s disease.
BigField GEG Tech's insight:

Huntington's disease (HD) is a neurological disorder that causes progressive loss of movement, coordination and cognitive function. It is caused by a mutation in a single gene called huntingtin or HTT. In a new study, published Dec. 12, 2022, in Nature Neuroscience, researchers describe the use of CRISPR/Cas13d RNA-targeting technology to develop a new therapeutic strategy that specifically removes the toxic RNA that causes HD. They used viral vehicles to deliver the therapy to neuronal cultures, which were developed from stem cells derived from patients with HD, and found that the approach not only targeted and destroyed mutant RNA molecules, but also eliminated the accumulation of toxic proteins. They also demonstrated that the expression of other human genes was generally not disrupted by the therapy.

Race&Herd's comment, January 19, 2023 12:08 AM
NICE
Scooped by BigField GEG Tech
Scoop.it!

Permanent inactivation of Huntington’s disease mutation by personalized allele-specific CRISPR/Cas9

Permanent inactivation of Huntington’s disease mutation by personalized allele-specific CRISPR/Cas9 | Genetic Engineering Publications - GEG Tech top picks | Scoop.it
BigField GEG Tech's insight:

Here, the scientists design a novel strategy of inactivating the mutant allele using haplotype-specific CRISPR/Cas9 target sites in Huntington's disease (HD), a late-onset neurodegenerative disorder due to a toxic dominant gain-of-function CAG expansion mutation.

No comment yet.
Scooped by BigField GEG Tech
Scoop.it!

CRISPR/Cas9-mediated gene editing ameliorates neurotoxicity in mouse model of Huntington’s disease

CRISPR/Cas9-mediated gene editing ameliorates neurotoxicity in mouse model of Huntington’s disease | Genetic Engineering Publications - GEG Tech top picks | Scoop.it

Click here to edit the content

BigField GEG Tech's insight:

Here, the scientists report that permanent suppression of endogenous mHTT expression in the striatum of mHTT-expressing mice (HD140Q-knockin mice) using CRISPR/Cas9-mediated inactivation effectively depleted HTT aggregates and attenuated early neuropathology. The reduction of mHTT expression in striatal neuronal cells in adult HD140Q-knockin mice did not affect viability, but alleviated motor deficits. Their studies suggest that non–allele-specific CRISPR/Cas9-mediated gene editing could be used to efficiently and permanently eliminate polyglutamine expansion–mediated neuronal toxicity in the adult brain.

No comment yet.