Abstract

BackgroundThe NDUFS4 gene encodes an 18-kD subunit of mitochondria complex I, and mutations in this gene lead to the development of a severe neurodegenerative disease called Leigh syndrome (LS) in humans. To investigate the disease phenotypes and molecular mechanisms of Leigh syndrome, the Ndufs4 knockout (KO) mouse has been widely used as a novel animal model. Because the homozygotes cannot survive beyond child-bearing age, whether Ndufs4 and mitochondrial complex I influence early embryonic development remains unknown. In our study, we attempted to investigate embryonic development in Ndufs4 KO mice, which can be regarded as a Leigh disease model and were created through the CRISPR (clustered regularly interspaced short palindromic repeat) and Cas9 (CRISPR associated)-mediated genome editing system.MethodsWe first designed a single guide RNA (sgRNA) targeting exon 2 of Ndufs4 to delete the NDUFS4 protein in mouse embryos to mimic Leigh syndrome. Then, we described the phenotypes of our mouse model by forced swimming and the open-field test as well as by assessing other behavioral characteristics. Intracytoplasmic sperm injection (ICSI) was performed to obtain KO embryos to test the influence of NDUFS4 deletion on early embryonic development.ResultsIn this study, we first generated Ndufs4 KO mice with physical and behavioral phenotypes similar to Leigh syndrome using the CRISPR/Cas9 system. The low developmental rate of KO embryos that were derived from knockout gametes indicated that the absence of NDUFS4 impaired the development of preimplantation embryos.DiscussionIn this paper, we first obtained Ndufs4 KO mice that could mimic Leigh syndrome using the CRISPR/Cas9 system. Then, we identified the role of NDUFS4 in early embryonic development, shedding light on its roles in the respiratory chain and fertility. Our model provides a useful tool with which to investigate the function of Ndufs4. Although the pathological mechanisms of the disease need to be discovered, it helps to understand the pathogenesis of NDUFS4 deficiency in mice and its effects on human diseases.

Highlights

  • Mitochondria are the major energy-producing organelles in eukaryotes, and the Ndufs4 encodes an 18-kD subunit of mitochondrial complex I (CI), which is the largest protein assembly of the respiratory chain and forms the major entry-point of electrons into the oxidative phosphorylation system (OXPHOS)

  • Researchers found that a higher potential for continued embryogenesis and implantation in humans was associated with embryos that developed from oocytes with greater ATP content (Van Blerkom, Davis & Lee, 1995)

  • Many aberrations during early mouse embryonic development and the increasing health risks in the offspring may be caused by numerous dysregulated genes related to mitochondrial complex I in in vitro fertilization (IVF) embryos (Ren et al, 2015)

Read more

Summary

Introduction

Mitochondria are the major energy-producing organelles in eukaryotes, and the Ndufs encodes an 18-kD subunit of mitochondrial complex I (CI), which is the largest protein assembly of the respiratory chain and forms the major entry-point of electrons into the oxidative phosphorylation system (OXPHOS). Ndufs deletion mice are excellent models with which to study the phenotypes and molecular mechanisms of mitochondrial complex I deficiency and Leigh syndrome. Many aberrations during early mouse embryonic development and the increasing health risks in the offspring may be caused by numerous dysregulated genes related to mitochondrial complex I in IVF (in vitro fertilization) embryos (Ren et al, 2015). These data indicated that both mitochondria and CI play crucial roles in embryonic development. The KO testes contained intact seminiferous epithelium and mature elongated spermatozoa in the seminiferous tubes (Figs. 3F, 3G).

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