Mouse newborn cells allow highly productive mouse cytomegalovirus replication, constituting a novel convenient primary cell culture system

PLoS One. 2017 Mar 24;12(3):e0174695. doi: 10.1371/journal.pone.0174695. eCollection 2017.

Abstract

Mammalian cell culture is indispensable for most aspects of current biomedical research. Immortalized cell lines are very convenient, but transforming principles (e.g. oncogenic viruses or their oncogenes) can heavily influence the experimental outcome. Primary cells do not share this apparent disadvantage but are more laborious to generate. Certain viruses (e.g. mouse cytomegalovirus) do not replicate efficiently in most transformed cell lines. In the past, such viruses have been routinely propagated on primary mouse embryonic fibroblasts (MEF) established around day 17 (d17) of gestation. According to new regulations of the European Union, experiments using gravid mammals and/or their embryos in the last trimester (>d14 in the case of mice) of gestation do require explicit permission of the local authorities responsible for animal care and use. Applying for such permission is time-consuming and often inflexible. Embryonic fibroblasts could also be produced at earlier time points of pregnancy from younger and smaller embryos. Obviously, this approach consumes more pregnant mice and embryos. Newborn mice are larger thus yielding more cells per sacrificed animal and the new Directive (2010/63/EU) excludes the killing of animals solely for the use of their organs or tissues. We established a convenient protocol to generate adherent mouse newborn cells (MNC). A direct comparison of MNC with MEF revealed that MNC fully recapitulate all tested aspects of a broad panel of virological parameters (plaque size, final titers, viral replication kinetics, viral gene expression, drug and interferon susceptibility as well as species specificity). The herein described approach allows researchers the legal use of primary cells and contributes to the 3R (replace, reduce, refine) guiding principles-especially the 'reduce' aspect-for the use of animals in scientific research. Additionally, it offers the option to directly compare in vitro and in vivo experiments when MNC are generated from littermates of animals included in the in vivo experiments.

MeSH terms

  • Animals
  • Cytomegalovirus / physiology*
  • Fibroblasts / cytology
  • Mice
  • Primary Cell Culture / methods*
  • Virus Replication / physiology*

Grants and funding

VTKL-T and MT received funding from the Deutsche Forschungsgemeinschaft (DFG) through TRR60 project A7N and GRK1949 project 13. MT also received intramural support (IFORES Sonderprogramm für Innovative Forschung) from the Medical Faculty of the University Hospital Essen. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.