scholarly journals 331. Development of Neisseria meningitidis CRISPR/Cas9 Systems for Efficient and Specific Genome Editing

2015 ◽  
Vol 23 ◽  
pp. S132-S133 ◽  
Author(s):  
Ciaran M. Lee ◽  
Thomas J. Cradick ◽  
Gang Bao
2018 ◽  
Author(s):  
Raed Ibraheim ◽  
Chun-Qing Song ◽  
Aamir Mir ◽  
Nadia Amrani ◽  
Wen Xue ◽  
...  

AbstractClustered, regularly interspaced, short palindromic repeats (CRISPR) and CRISPR-associated proteins (Cas) have recently opened a new avenue for gene therapy. Cas9 nuclease guided by a single-guide RNA (sgRNA) has been extensively used for genome editing. Currently, three Cas9 orthologs have been adapted for in vivo genome engineering applications: SpyCas9, SauCas9 and CjeCas9. However, additional in vivo editing platforms are needed, in part to enable a greater range of sequences to be accessed via viral vectors, especially those in which Cas9 and sgRNA are combined into a single vector genome. Here, we present an additional in vivo editing platform using Neisseria meningitidis Cas9 (NmeCas9). NmeCas9 is compact, edits with high accuracy, and possesses a distinct PAM, making it an excellent candidate for safe gene therapy applications. We find that NmeCas9 can be used to target the Pcsk9 and Hpd genes in mice. Using tail vein hydrodynamic-based delivery of NmeCas9 plasmid to target the Hpd gene, we successfully reprogrammed the tyrosine degradation pathway in Hereditary Tyrosinemia Type I mice. More importantly, we delivered NmeCas9 with its single-guide RNA in a single recombinant adeno-associated vector (rAAV) to target Pcsk9, resulting in lower cholesterol levels in mice. This all-in-one vector yielded >35% gene modification after two weeks of vector administration, with minimal off-target cleavage in vivo. Our findings indicate that NmeCas9 can facilitate future efforts to correct disease-causing mutations by expanding the targeting scope of RNA-guided nucleases.


Author(s):  
Rongfang Xu ◽  
Ruiying Qin ◽  
Hongjun Xie ◽  
Juan Li ◽  
Xiaoshuang Liu ◽  
...  

2020 ◽  
Author(s):  
Mareike D Hoffmann ◽  
Jan Mathony ◽  
Julius Upmeier zu Belzen ◽  
Zander Harteveld ◽  
Sabine Aschenbrenner ◽  
...  

Abstract Optogenetic control of CRISPR–Cas9 systems has significantly improved our ability to perform genome perturbations in living cells with high precision in time and space. As new Cas orthologues with advantageous properties are rapidly being discovered and engineered, the need for straightforward strategies to control their activity via exogenous stimuli persists. The Cas9 from Neisseria meningitidis (Nme) is a particularly small and target-specific Cas9 orthologue, and thus of high interest for in vivo genome editing applications. Here, we report the first optogenetic tool to control NmeCas9 activity in mammalian cells via an engineered, light-dependent anti-CRISPR (Acr) protein. Building on our previous Acr engineering work, we created hybrids between the NmeCas9 inhibitor AcrIIC3 and the LOV2 blue light sensory domain from Avena sativa. Two AcrIIC3-LOV2 hybrids from our collection potently blocked NmeCas9 activity in the dark, while permitting robust genome editing at various endogenous loci upon blue light irradiation. Structural analysis revealed that, within these hybrids, the LOV2 domain is located in striking proximity to the Cas9 binding surface. Together, our work demonstrates optogenetic regulation of a type II-C CRISPR effector and might suggest a new route for the design of optogenetic Acrs.


2016 ◽  
Vol 24 (3) ◽  
pp. 645-654 ◽  
Author(s):  
Ciaran M Lee ◽  
Thomas J Cradick ◽  
Gang Bao

2019 ◽  
Author(s):  
Mareike D. Hoffmann ◽  
Jan Mathony ◽  
Julius Upmeier zu Belzen ◽  
Zander Harteveld ◽  
Christina Stengl ◽  
...  

ABSTRACTOptogenetic control of CRISPR-Cas9 systems has significantly improved our ability to perform genome perturbations in living cells with high precision in time and space. As new Cas orthologues with advantageous properties are rapidly being discovered and engineered, the need for straightforward strategies to control their activity via exogenous stimuli persists. The Cas9 from Neisseria meningitidis (Nme) is a particularly small and target-specific Cas9 orthologue, and thus of high interest for in vivo genome editing applications.Here, we report the first optogenetic tool to control NmeCas9 activity in mammalian cells via an engineered, light-dependent anti-CRISPR (Acr) protein. Building on our previous Acr engineering work, we created hybrids between the NmeCas9 inhibitor AcrIIC3 and the LOV2 blue light sensory domain from Avena sativa. Two AcrIIC3-LOV2 hybrids from our collection potently blocked NmeCas9 activity in the dark, while permitting robust genome editing at various endogenous loci upon blue light irradiation. Structural analysis revealed that, within these hybrids, the LOV2 domain is located in striking proximity to the Cas9 binding surface. Together, our work demonstrates optogenetic regulation of a type II-C CRISPR effector and might suggest a new route for the design of optogenetic Acrs.


2018 ◽  
Vol 19 (1) ◽  
Author(s):  
Raed Ibraheim ◽  
Chun-Qing Song ◽  
Aamir Mir ◽  
Nadia Amrani ◽  
Wen Xue ◽  
...  

2017 ◽  
Vol 6 (3) ◽  
pp. 162-162
Author(s):  
Liane Kaufmann ◽  
Michael von Aster
Keyword(s):  

2018 ◽  
Author(s):  
M Keller ◽  
J Dalla-Riva ◽  
A Kurbasic ◽  
M Al-Majdoub ◽  
P Spegel ◽  
...  

1998 ◽  
Vol 26 (149) ◽  
pp. 34 ◽  
Author(s):  
Mª Amparo Morant Gimeno ◽  
J. Díez Domingo ◽  
C. Gimeno ◽  
N. de la Muela ◽  
I. Pereiró ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document