Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Adv Sci (Weinh) ; 10(28): e2303496, 2023 10.
Article in English | MEDLINE | ID: mdl-37562980

ABSTRACT

Domain insertion engineering is a promising approach to recombine the functions of evolutionarily unrelated proteins. Insertion of light-switchable receptor domains into a selected effector protein, for instance, can yield allosteric effectors with light-dependent activity. However, the parameters that determine domain insertion tolerance and allostery are poorly understood. Here, an unbiased screen is used to systematically assess the domain insertion permissibility of several evolutionary unrelated proteins. Training machine learning models on the resulting data allow to dissect features informative for domain insertion tolerance and revealed sequence conservation statistics as the strongest indicators of suitable insertion sites. Finally, extending the experimental pipeline toward the identification of switchable hybrids results in opto-chemogenetic derivatives of the transcription factor AraC that function as single-protein Boolean logic gates. The study reveals determinants of domain insertion tolerance and yielded multimodally switchable proteins with unique functional properties.


Subject(s)
Transcription Factors , Allosteric Regulation
2.
Nucleic Acids Res ; 49(5): e29, 2021 03 18.
Article in English | MEDLINE | ID: mdl-33330940

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.


Subject(s)
CRISPR-Associated Protein 9/antagonists & inhibitors , CRISPR-Associated Protein 9/chemistry , CRISPR-Cas Systems , Gene Editing/methods , Neisseria meningitidis/enzymology , Optogenetics/methods , Cell Line , HEK293 Cells , Humans , Light , Models, Molecular , Protein Engineering , Proteins/chemistry , Proteins/radiation effects
3.
Nat Chem Biol ; 16(7): 725-730, 2020 07.
Article in English | MEDLINE | ID: mdl-32284602

ABSTRACT

Anti-CRISPR (Acr) proteins are powerful tools to control CRISPR-Cas technologies. However, the available Acr repertoire is limited to naturally occurring variants. Here, we applied structure-based design on AcrIIC1, a broad-spectrum CRISPR-Cas9 inhibitor, to improve its efficacy on different targets. We first show that inserting exogenous protein domains into a selected AcrIIC1 surface site dramatically enhances inhibition of Neisseria meningitidis (Nme)Cas9. Then, applying structure-guided design to the Cas9-binding surface, we converted AcrIIC1 into AcrIIC1X, a potent inhibitor of the Staphylococcus aureus (Sau)Cas9, an orthologue widely applied for in vivo genome editing. Finally, to demonstrate the utility of AcrIIC1X for genome engineering applications, we implemented a hepatocyte-specific SauCas9 ON-switch by placing AcrIIC1X expression under regulation of microRNA-122. Our work introduces designer Acrs as important biotechnological tools and provides an innovative strategy to safeguard CRISPR technologies.


Subject(s)
CRISPR-Associated Protein 9/genetics , CRISPR-Cas Systems , Clustered Regularly Interspaced Short Palindromic Repeats , Gene Editing/methods , MicroRNAs/genetics , Protein Engineering/methods , Amino Acid Sequence , CRISPR-Associated Protein 9/metabolism , Cell Line, Tumor , Genome, Human , HEK293 Cells , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , MicroRNAs/metabolism , Models, Molecular , Mutagenesis, Insertional , Neisseria meningitidis/enzymology , Neisseria meningitidis/genetics , Plasmids/chemistry , Plasmids/metabolism , Protein Domains , Protein Structure, Secondary , RNA, Guide, Kinetoplastida/genetics , RNA, Guide, Kinetoplastida/metabolism , Staphylococcus aureus/enzymology , Staphylococcus aureus/genetics
4.
Sci Adv ; 6(6): eaay0187, 2020 02.
Article in English | MEDLINE | ID: mdl-32076642

ABSTRACT

The limited target specificity of CRISPR-Cas nucleases poses a challenge with respect to their application in research and therapy. Here, we present a simple and original strategy to enhance the specificity of CRISPR-Cas9 genome editing by coupling Cas9 to artificial inhibitory domains. Applying a combination of mathematical modeling and experiments, we first determined how CRISPR-Cas9 activity profiles relate to Cas9 specificity. We then used artificially weakened anti-CRISPR (Acr) proteins either coexpressed with or directly fused to Cas9 to fine-tune its activity toward selected levels, thereby achieving an effective kinetic insulation of ON- and OFF-target editing events. We demonstrate highly specific genome editing in mammalian cells using diverse single-guide RNAs prone to potent OFF-targeting. Last, we show that our strategy is compatible with different modes of delivery, including transient transfection and adeno-associated viral vectors. Together, we provide a highly versatile approach to reduce CRISPR-Cas OFF-target effects via kinetic insulation.


Subject(s)
CRISPR-Associated Protein 9/genetics , CRISPR-Cas Systems , Gene Editing , Gene Targeting , Recombinant Fusion Proteins , CRISPR-Associated Protein 9/chemistry , CRISPR-Associated Protein 9/metabolism , Cell Line , Enzyme Activation , Gene Targeting/methods , Genes, Reporter , Genetic Loci , Humans , Kinetics , Models, Theoretical , Substrate Specificity
5.
Nucleic Acids Res ; 47(13): e75, 2019 07 26.
Article in English | MEDLINE | ID: mdl-30982889

ABSTRACT

The rapid development of CRISPR-Cas technologies brought a personalized and targeted treatment of genetic disorders into closer reach. To render CRISPR-based therapies precise and safe, strategies to confine the activity of Cas(9) to selected cells and tissues are highly desired. Here, we developed a cell type-specific Cas-ON switch based on miRNA-regulated expression of anti-CRISPR (Acr) proteins. We inserted target sites for miR-122 or miR-1, which are abundant specifically in liver and cardiac muscle cells, respectively, into the 3'UTR of Acr transgenes. Co-expressing these with Cas9 and sgRNAs resulted in Acr knockdown and released Cas9 activity solely in hepatocytes or cardiomyocytes, while Cas9 was efficiently inhibited in off-target cells. We demonstrate control of genome editing and gene activation using a miR-dependent AcrIIA4 in combination with different Streptococcus pyogenes (Spy)Cas9 variants (full-length Cas9, split-Cas9, dCas9-VP64). Finally, to showcase its modularity, we adapted our Cas-ON system to the smaller and more target-specific Neisseria meningitidis (Nme)Cas9 orthologue and its cognate inhibitors AcrIIC1 and AcrIIC3. Our Cas-ON switch should facilitate cell-specific activity of any CRISPR-Cas orthologue, for which a potent anti-CRISPR protein is known.


Subject(s)
CRISPR-Associated Protein 9/genetics , CRISPR-Cas Systems , Gene Editing/methods , Gene Expression Regulation , Transgenes , 3' Untranslated Regions/genetics , Binding Sites , CRISPR-Associated Protein 9/antagonists & inhibitors , CRISPR-Associated Protein 9/biosynthesis , Dependovirus/genetics , Enzyme Activation , Enzyme Induction , Genes, Reporter , HEK293 Cells , HeLa Cells , Hepatocytes/metabolism , Humans , Luciferases, Renilla/analysis , Luciferases, Renilla/genetics , MicroRNAs , Myocytes, Cardiac/metabolism , Organ Specificity , Protein Isoforms/antagonists & inhibitors
6.
Nat Methods ; 15(11): 924-927, 2018 11.
Article in English | MEDLINE | ID: mdl-30377362

ABSTRACT

Anti-CRISPR proteins are powerful tools for CRISPR-Cas9 regulation; the ability to precisely modulate their activity could facilitate spatiotemporally confined genome perturbations and uncover fundamental aspects of CRISPR biology. We engineered optogenetic anti-CRISPR variants comprising hybrids of AcrIIA4, a potent Streptococcus pyogenes Cas9 inhibitor, and the LOV2 photosensor from Avena sativa. Coexpression of these proteins with CRISPR-Cas9 effectors enabled light-mediated genome and epigenome editing, and revealed rapid Cas9 genome targeting in human cells.


Subject(s)
Biosensing Techniques , CRISPR-Associated Proteins/antagonists & inhibitors , CRISPR-Cas Systems , Gene Editing , Optogenetics , Phototropins/chemistry , Protein Engineering , Epigenomics , Genome , HEK293 Cells , Humans , Light , Streptococcus pyogenes/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL
...