CRISPR Mouse Models

At the speed of CRISPR. Generate models quickly and efficiently.

Working with ingenious targeting laboratory allows you to obtain high-quality CRISPR mouse models on the C57BL/6 background efficiently and with a guarantee of germline transmission or your money back. We create gene knockouts and knockins through a streamlined process to ensure rapid production, reducing costs and timelines compared to traditional gene targeting methods. Our robust personalized support, from design to delivery, is unparalleled in the industry.

Start your purchase!

“iTL generated a conventional knockout mouse for us, and we were very pleased with the entire process. The project manager was extremely helpful and efficient and promptly notified us whenever a step had been completed. I highly recommend iTL’s services for anyone considering knockout mouse models.”– Edward Harhaj, PhD Penn State College of Medicine

CRISPR Mouse Models

Customized Mouse Model Service Using CRISPR-Cas9 Technology

Germline Transmission Guaranteed

Through our collaboration with Shanghai Model Organisms Center, Inc. (SMOC), ingenious targeting laboratory is proud to offer select, high-quality, custom CRISPR mouse models with germline transmission guaranteed.

This service is offered in the classic C57BL/6 mouse strain. We have also expanded our services to include CRISPR gene editing in the NMG mouse strain. Similar to NOD, the NMG strain is severely immunodeficient, with a lack of mature T, B, & NK cells. These models can be applied to PK/PD assays, assessment of drug safety, study of disease, pathogenic mechanism, biology, & more. They are also applicable to engraftment of human cells and tissues.

Fill out our quote form to find out how we can apply the power of CRISPR/Cas9 genome editing for your next project, whether it is a guide RNA-directed gene inactivation or introduction of a single point mutation.

get a quote

Germline transmission guarantee available for these mouse models:

Conventional Knockout

Cas9 generates targeted cuts in your gene of interest which are repaired by an error-prone DNA repair pathway, i.e., non-homologous end joining (NHEJ). Mutations introduced by this process disrupt the gene either by nucleotide insertions or deletions (INDELs) to create a knockout allele.

Constitutive Point Mutation

Single nucleotide changes are introduced at a target site by combining CRISPR/Cas9 with a modified DNA template (e.g., an oligo) in order to support a repair pathway which utilizes homology-directed repair (HDR). Commonly this strategy applies to the modeling of disease-causing mutations or the humanization of critical amino acids.

The 3 Step ingenious-SMOC Process for Generating Your Custom CRISPR Mouse Model

Scientific experts at ingenious and SMOC will work with you to design and customize your genetically engineered constitutive KO or point mutation KI mouse model, to your specifications.

1st Step: Strategy and Design

Identification of target site, guides, and oligonucleotides, as well as predicted off-target sites. Preparation of sgRNA and Cas9 mRNA

2nd Step: Production of F0 Founder Mice

Injection of CRISPR/Cas9 gene editing material into fertilized embryos to generate F0 mutated candidate animals resulting from embryo transfer into pseudopregnant mice.

3rd Step: Breeding of Founders to Obtain F1 Mice

The new founders are bred with wildtype mice of matching strain background, and their offspring are genotyped to identify and select F1 mice bearing the intended knockout or knockin allele originating from the genetically heterogeneous (i.e., mosaic) F0 founder.

How does the CRISPR-Cas9 system work?

The CRISPR system, originally found in bacteria, utilizes an enzyme called Cas9 nuclease to create double-stranded breaks (DSBs) in DNA.  Cas9 activity relies on a CRISPR RNA (crRNA) and a trans-activating CRISPR RNA (tracrRNA). The crRNA is complementary to a DNA target region, and the tracrRNA serves as a scaffold.  These two RNAs form a complex known as a guide RNA (gRNA).

When the CRISPR-Cas9 system is used to create a gene knockout, Cas9 creates a DSB in DNA which is most likely repaired by non-homologous end joining (NHEJ).  NHEJ is error-prone and usually results in insertions and deletions (indels) in the region being repaired.  This creates a frameshift, rendering the gene non-functional.

When the CRISPR-Cas9 system is used to create a gene knockin, the DSB in DNA caused by Cas9 is repaired using an alternative pathway called homology-directed repair (HDR).  In order for gene knock-in to work, there must be a DNA template available, called a donor template, in addition to Cas9 and the gRNA. The donor template contains the sequence to knock in, flanked by regions of homology that match the area on either side of the cutting region.  Through homologous recombination between the donor and target gene, the knockin piece of DNA is inserted into the target gene.

Advantages of using CRISPR-Cas9 in Mouse Model Generation

CRISPR/Cas9 enables the creation of founder mice for your new line in less time and at a lower cost compared to cell-based approaches. Previously to make a knockout line the modification had to be made and verified in ES cells, which can take three months or more, then potential chimeric founders had to be generated from those cells. CRISPR eliminates this initial step and creates genetic changes directly in embryos, so that screening for potential founders can begin months earlier in relation to ES cell-derived animals. We are looking forward to working with you to create an indispensable mouse line in support of your research.

Frequently asked questions:

Can CRISPR be used in animals?

CRISPR-Cas9 can be used in the generation of transgenic animals. This system is efficient and more straight forward than other traditional techniques.

How is CRISPR used in animal models?

CRISPR-Cas9 is used in mice and rats to knock out a gene or to knock in a new DNA sequence. Source: Three Ways CRISPR Is Making Animal Research Models More Predictive

What diseases could CRISPR get rid of in the future?

CRISPR-Cas9 could be used to treat various human hereditary diseases such as hemophila, β-thalassemia, cystic fibrosis, Alzheimer’s, Huntington’s, Parkinson’s, tyrosinemia, Duchnene muscular dystrophy, Tay-Sachs, and fragile X syndrome disorders. ource: CRISPR-Cas9 for treating hereditary diseases – PubMed

Notable Publications by ingenious Clients

Ojeda-Alonso J, Bégay V, Garcia-Contreras J, Campos-Pérez A, Purfürst B, Lewin G. 2022. Lack of evidence for participation of TMEM150c/TENTONIN3 in sensory mechanotransduction.J Gen Physiol 154(12): e202213098.

Smith TC, Vasilakos G, Shaffer S, Puglise J, Chou C, Barton E, Luna E. 2022. Novel γ-sarcoglycan interactors in murine muscle membranes.Skelet Muscle12(1): 2.

Choi J, Diao H, Faliti CE, Truong J, Rossi M, Bélanger S, Yu B, Goldrath AW, Pipkin ME, Crotty S. 2020. Bcl-6 is the nexus transcription factor of T follicular helper cells via repressor-of-repressor circuits. Nat Immunol21(7): 777-789.

Rothschild G, Zhang W, Lim J, Giri PK, Laffleur B, Chen Y, Fang M, Nair L, Liu ZP, Deng H, Hammarstrom L, Wang J, Basu U. 2020. Noncoding RNA transcription alters chromosomal topology to promote isotype-specific class switch recombination. Sci Immunol5(44).

Notable Publications by SMOC Clients

Zhao H, Huang X, Liu Z, Pu W, Lv Z, He L, Li Y, Zhou Q, Lui KO, Zhou B. 2021. Pre-existing beta cells but not progenitors contribute to new beta cells in the adult pancreas. Nat Metab3(3): 352-365.

Zhou H, Liu J, Zhou C, Gao N, Rao Z, Li H, Hu X, Li C, Yao X, Shen X, Sun Y, Wei Y, Liu F, Ying W, Zhang J, Tang C, Zhang X, Xu H, Shi L, Cheng L, Huang P, Yang H. 2018. In vivo simultaneous transcriptional activation of multiple genes in the brain using CRISPR-dCas9-activator transgenic mice. Nat Neurosci21(3): 440-446.

Huai C, Jia C, Sun R, Xu P, Min T, Wang Q, Zheng C, Chen H, Lu D. 2017. CRISPR/Cas9-mediated somatic and germline gene correction to restore hemostasis in hemophilia B mice. Hum Genet136(7): 875-883.

Xie C, Zhang YP, Song L, Luo J, Qi W, Hu J, Lu D, Yang Z, Zhang J, Xiao J, Zhou B, Du JL, Jing N, Liu Y, Wang Y, Li BL, Song BL, Yan Y. 2016. Genome editing with CRISPR/Cas9 in postnatal mice corrects PRKAG2 cardiac syndrome. Cell Res26(10): 1099-1111.