Non-alcoholic fatty liver disease in mice with heterozygous mutation in TMED2

PLoS One. 2017 Aug 10;12(8):e0182995. doi: 10.1371/journal.pone.0182995. eCollection 2017.

Abstract

The transmembrane emp24 domain/p24 (TMED) family are essential components of the vesicular transport machinery. Members of the TMED family serve as cargo receptors implicated in selection and packaging of endoplasmic reticulum (ER) luminal proteins into coatomer (COP) II coated vesicles for anterograde transport to the Golgi. Deletion or mutations of Tmed genes in yeast and Drosophila results in ER-stress and activation of the unfolded protein response (UPR). The UPR leads to expression of genes and proteins important for expanding the folding capacity of the ER, degrading misfolded proteins, and reducing the load of new proteins entering the ER. The UPR is activated in non-alcoholic fatty liver disease (NAFLD) in human and mouse and may contribute to the development and the progression of NAFLD. Tmed2, the sole member of the vertebrate Tmed β subfamily, exhibits tissue and temporal specific patterns of expression in embryos and developing placenta but is ubiquitously expressed in all adult organs. We previously identified a single point mutation, the 99J mutation, in the signal sequence of Tmed2 in an N-ethyl-N-nitrosourea (ENU) mutagenesis screen. Histological and molecular analysis of livers from heterozygous mice carrying the 99J mutation, Tmed299J/+, revealed a requirement for TMED2 in liver health. We show that Tmed299J/+ mice had decreased levels of TMED2 and TMED10, dilated endoplasmic reticulum membrane, and increased phosphorylation of eIF2α, indicating ER-stress and activation of the UPR. Increased expression of Srebp1a and 2 at the newborn stage and increased incidence of NAFLD were also found in Tmed299J/+ mice. Our data establishes Tmed299J/+ mice as a novel mouse model for NAFLD and supports a role for TMED2 in liver health.

MeSH terms

  • Animals
  • Endoplasmic Reticulum Stress
  • Hep G2 Cells
  • Heterozygote
  • Humans
  • Liver / metabolism
  • Liver / pathology*
  • Membrane Proteins
  • Mice
  • Mice, Inbred C57BL
  • Non-alcoholic Fatty Liver Disease / genetics*
  • Point Mutation*
  • Sterol Regulatory Element Binding Protein 1 / analysis
  • Sterol Regulatory Element Binding Protein 1 / genetics
  • Sterol Regulatory Element Binding Protein 2 / analysis
  • Sterol Regulatory Element Binding Protein 2 / genetics
  • Unfolded Protein Response
  • Up-Regulation
  • Vesicular Transport Proteins / analysis
  • Vesicular Transport Proteins / genetics*

Substances

  • Membrane Proteins
  • Srebf1 protein, mouse
  • Srebf2 protein, mouse
  • Sterol Regulatory Element Binding Protein 1
  • Sterol Regulatory Element Binding Protein 2
  • Tmed10 protein, mouse
  • Tmed2 protein, mouse
  • Vesicular Transport Proteins

Grants and funding

This work was supported by the Natural Sciences and Engineering Research Council of Canada (NSERC), http://www.nserc-crsng.gc.ca/index_eng.asp, RGPIN-2015-06699.