Abstract

It is a tempting goal to identify causative genes underlying phenotypic differences among inbred strains of mice, which is a huge reservoir of genetic resources to understand mammalian pathophysiology. In particular, the wild-derived mouse strains harbor enormous genetic variations that have been acquired during evolutionary divergence over 100s of 1000s of years. However, validating the genetic variation in non-classical strains was extremely difficult, until the advent of CRISPR/Cas9 genome editing tools. In this study, we first describe a T cell phenotype in both wild-derived PWD/PhJ parental mice and F1 hybrids, from a cross to C57BL/6 (B6) mice, and we isolate a genetic locus on Chr2, using linkage mapping and chromosome substitution mice. Importantly, we validate the identification of the functional gene controlling this T cell phenotype, Cd44, by allele specific knockout of the PWD copy, leaving the B6 copy completely intact. Our experiments using F1 mice with a dominant phenotype, allowed rapid validation of candidate genes by designing sgRNA PAM sequences that only target the DNA of the PWD genome. We obtained 10 animals derived from B6 eggs fertilized with PWD sperm cells which were subjected to microinjection of CRISPR/Cas9 gene targeting machinery. In the newborns of F1 hybrids, 80% (n = 10) had allele specific knockout of the candidate gene Cd44 of PWD origin, and no mice showed mistargeting of the B6 copy. In the resultant allele-specific knockout F1 mice, we observe full recovery of T cell phenotype. Therefore, our study provided a precise and rapid approach to functionally validate genes that could facilitate gene discovery in classic mouse genetics. More importantly, as we succeeded in genetic manipulation of mice, allele specific knockout could provide the possibility to inactivate disease alleles while keeping the normal allele of the gene intact in human cells.

Highlights

  • Wild mice refer to both inbred lines and individual animals from natural house mouse populations, both of which harbor enormous genetic variations

  • T lymphocytes are pivotal components in the immune system of mammals to fight against pathogens and tumors and their development is under sophisticated molecular control which requires genetic dissection (Liang et al, 2013; Wang et al, 2017)

  • The mean fluorescent intensity (MFI) of CD44 on both CD4 and CD8 T cells was 3-fold higher in PWD mice following T cell receptor (TCR) stimulation (Figures 1H,I). These results showed that PWD mice had significantly higher expression of CD44 in steady-state, and more importantly, CD44 upregulation following TCR stimulation was more potent in T cells of PWD mice

Read more

Summary

Introduction

Wild mice refer to both inbred lines and individual animals from natural house mouse populations, both of which harbor enormous genetic variations. Such models are useful for study of genetic and environmental factors contributing to host immune response to pathogens (Rosshart et al, 2017). Musculus derived PWK mice have highly diverged genomes with over 17 million single nucleotide polymorphisms (SNPs) which far outnumbers the genetic variations between classical laboratory strains (Keane et al, 2011). Musculus derived strain and harness the functional genetic variations which could not be found in classical mice (Gregorova and Forejt, 2000). In the backcrossed population, we found 50% mice (275 out of 559) displaying the PWD T cell phenotype

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call