CRISPR IVT RNA
CRISPR IVT RNA is rapidly being adopted as a method for genome editing due to its numerous advantages including editing independent of cell-type promoter expression, no risk of genomic integration, and transient expression which reduces off-target effects. In addition, Cas9 mRNA and gRNA can be delivered co-encapsulated in lipid nanoparticles which results in efficient gene delivery and editing in vitro and in vivo. VectorBuilder offers a full range of IVT RNA products for CRISPR-genome editing including Cas9 mRNA, Cas9-ABE mRNA, and guide RNA. To get started with your own CRISPR experiments, click the button below to talk to our design team today.
Talk to Our Experts
Highlights
Robust quality, fast turnaround, and competitive pricing
Powerful technical support for experimental design, data analysis, and troubleshooting
Cas9 mRNA and gRNA LNP co-encapsulation for rapid, efficient editing
Sequence optimized Cas9 with high expression efficiency available for licensing
Offering Details
- Premade CRISPR IVT RNA products.
- Therapeutic RNA development: IVT vector design & cloning, IVT RNA production & purification.
- LNP encapsulation: standard and custom formulations, antibody-conjugated LNP.
- CDMO services: process development, GMP manufacturing, fill/finish.
Technical Information
CRISPR IVT RNA can be applied across a variety of systems including in vitro and in vivo genome editing as well as CRISPR-mediated base editing. The following data validates the effectiveness of CRISPR IVT RNA in various settings.
- In vitro knockout
- Dual-gRNA knockout
- In vivo knockout
- Base editing
Figure 1. Validation of hSpCas9 mRNA in vitro. (A) IVT Cas9 mRNA was transfected into HEK293T-EGFP cells with two types of EGFP-targeting gRNA. EGFP expression in non-treated (NC) and transfected cells was observed by microscopy (B) and quantified using flow cytometry (C, D). (E, F) The editing to EGFP genes on the genome was further confirmed by T7E1 assay and Sanger sequencing.
Figure 2. Validation of hSpCas9 mRNA knockout with dual gRNAs. (A) A vector coding for Cas9 was linearized and subjected to in vitro transcription to produce Cas9 mRNA. The Cas9 mRNA was then co-transfected with two gRNAs targeting the same gene. (B) Detailed experimental design of the gRNAs targeting exons 2-4 of the gene of interest. Primer pairs P1 and P2 were designed for PCR validation to amplify the regions surrounding the target sites. (C) Gel electrophoresis of PCR amplified genomic DNA confirms gene editing.
Figure 3. In vivo CRISPR IVT RNA knockout. (A) Experimental timeline of CRISPR IVT RNA knockout in vivo. Mice were intravenously injected with 3.0 ug/g of Cas9 mRNA/gRNA mixture co-encapsulated in LNPs or PBS. On Day 7, liver genomic DNA was extracted, PCR amplified, and subjected to a T7E1 assay to confirm editing. (B) Gel electrophoresis of the T7E1 assay confirming editing (asterisked ban). Each lane represents one biological replicate.
Figure 4. Validation of adenine base editor (ABE) mRNAs in vitro. mRNAs coding for (A) ABE8.20-m and (B) ABE7.10 were co-transfected with identical gRNAs to HEK293T cells. Genomic DNA was extracted 48 hr post-transfection, and the target region was amplified by PCR and subjected to NGS. A to G editing (asterisks) was detected within the editing windows (dashed box). The highest editing rates of 97.7% (A) and 95.6% (B) were achieved on the 5th nucleotide.
HiExpress™ hSpCas9 IVT mRNA
VectorBuilder has developed a sequence optimized HiExpressTM hSpCas9 IVT mRNA that exhibits increased expression compared to other human codon-optimized Cas9 variants and therefore requires less mRNA to achieve comparable editing efficiency in cells making it ideally suited for gene editing in therapeutic and biotechnology applications. For more information about licensing this product, contact us today.
Figure 5. HiExpress™ hSpCas9 IVT mRNA developed by VectorBuilder has high expression and editing efficiency. (A) Western blot analysis and (B) quantification of normalized Cas9 expression relative to hSpCas9 in HEK293T cells 24 hours post-transfection with 1 ug of either hSpCas9 IVT mRNA, codon-optimized HiExpress™ hSpCas9 IVT mRNA, or a non-treated control (NC). (C) T7E1 editing assay in HEK293T cells 24 hours post-transfection with 1 ug of gRNA and the indicated amounts of hSpCas9 or HiExpress™ hSpCas9 IVT mRNA. The blue star indicates the intact PCR amplicon and the pink arrows indicate the fragments generated by T7E1.
FAQ
CRISPR-Cas9 can be delivered through a variety of methods including plasmid, recombinant virus, gRNA-Cas9 RNP complex, and as a mixture of gRNA and Cas9 IVT mRNA. Each of these methods has its own advantages and limitations which allows researchers to select the method tailored to their application for maximum efficiency with minimal off-target and undesired effects, with IVT RNA rapidly emerging as one of the most promising approaches.
Plasmids are generally the easiest to generate in large quantities cheaply and can be delivered through chemical transfection and electroporation which also tends to be simple. However, due to their method of delivery, they are largely limited to in vitro applications. In addition, the efficiency of plasmid transfection can vary widely between cell types and expression is dependent on cell-type specific promoter activity, which can hinder their efficiency in certain systems. As a result, plasmids tend to use highly active promoters which can lead to high-expression levels of Cas9 which can increase off-target editing and random integration of plasmid DNA into the host genome.
Recombinant viruses tend to be another popular method for delivery of CRISPR-Cas9 as they can be used in difficult to transfect cells and they can be used to create stably expressing Cas9 cell lines for multiple gRNA experiments. However, this system has many of the same limitations of plasmid delivery including cell-type dependent promoter specificity and elevated risk of off-target effects due to prolonged Cas9 expression. In addition, recombinant viruses pose an elevated risk of insertional mutagenesis, especially in retroviral and lentiviral systems where genomic integration of the Cas9 transgene occurs.
Delivery of a gRNA-Cas9 RNP complex circumvents many of the limitations of the plasmid and recombinant viral systems allowing for rapid editing as neither transcription nor translation needs to occur. However, this method is largely limited to in vitro applications as it requires electroporation for efficient delivery. Editing by this method occurs rapidly and quickly drops off as Cas9 is degraded in host cells and is therefore widely limited by the availability of Cas9 protein.
In contrast to the above methods, delivery of a mixture of gRNA and Cas9 IVT mRNA has rapidly emerged as one of the most efficient methods for genome editing with minimal off-target effects. gRNA and Cas9 IVT mRNA can be delivered together co-encapsulated in lipid nanoparticles or chemical transfection reagents making the system suitable for both in vitro and in vivo applications without risk of genomic integration. Further, no transcription is required, making expression independent of cell-type specific promoter activity and translation occurs relatively quickly, allowing for rapid and efficient editing. Compared to gRNA-Cas9 RNP complex, mRNA promotes prolonged expression of Cas9 allowing for sustained levels of editing, however, the mRNA is eventually degraded and editing only occurs transiently, limiting off-target effects.
In summary, delivery of CRISPR-Cas9 components through plasmids, recombinant virus, and gRNA-Cas9 RNP complex methods have limitations that limit their use to a number of applications and in addition, have downsides including limited efficiency, increased risk of off-target effects, and potential for insertional mutagenesis. Further, delivery of a mixture of gRNA and Cas9 IVT mRNA has emerged as one of the most efficient methods of genome editing due to its numerous advantages and fewer limitations and should be considered for genome editing applications.
For CRISPR-mediated genome editing, Cas9 nuclease is directed to the target site of site-specific guide RNA (gRNA) in the genome to create DNA cleavage. In most cases, to generate simple gene knockout, a single gRNA can be used together with Cas9 to generate a double-strand break (DSB), which is then inefficiently repaired by the non-homologous end joining (NHEJ), resulting in permanent mutations, such as small insertions or deletions, at the site of repair. A subset of these mutations will result in loss of function of the gene of interest due to frameshifts, premature stop codons, etc.
Dual gRNAs can be used if Cas9_D10A nickase is being used to target the two opposite strands of a single target site. In this approach, the nickase enzyme will generate single strand cuts on both strands, one guided by each of the two gRNAs, resulting in DSBs at the target site. Generally, this method reduces off-target effects of CRISPR/Cas9 expression because targeting by both gRNAs is necessary for DSBs to be generated.
Dual gRNAs can also be used when Cas9_D10A nickase and an exogenous donor DNA template are being used to introduce specific base-changes (e.g. knockins) into a gene of interest. In this approach, the two opposite strands would be targeted by the two gRNAs at two sites flanking the desired mutation site, and homology-directed repair (HDR) pathways make use of the exogenous donor template to repair the excised sequence.
Documents
Brochures & Flyers User InstructionsCertificate of Analysis (COA)
Material Safety Data Sheet (MSDS)