Deficiency of NONO is associated with impaired cardiac function and fibrosis in mice

J Mol Cell Cardiol. 2019 Dec:137:46-58. doi: 10.1016/j.yjmcc.2019.10.004. Epub 2019 Oct 18.

Abstract

Non-POU-domain-containing octamer-binding protein (NONO), a component of multifunctional Drosophila behavior/human splicing (DBHS) family, plays an important role in regulating glucose and fat metabolism, circadian cycles, cell division, collagen formation and fibrosis. Dysfunctional variants of NONO have been described as the cause of congenital heart defects in males. However, the effects of NONO deficiency on the ventricular function and cardiac fibrosis as well as the related mechanisms are not clear. In the present study, we aimed to reveal the overall phenotypes, cardiac function and fibroblasts in NONO knockout (NONO KO) mice compared with the wild-type (WT) male littermates. The results showed that the birth rate of NONOgt/0 mice was much lower than their WT male littermates at the time of weaning. The body weight of NONOgt/0 mice was 19% lower than that of WT male littermates (27.2 ± 1.49 g vs. 22.01 ± 1.20 g, P < .001). NONO KO mice exhibited continuous higher mortality from birth to a year later (P < .05). Compared with those in the WT mice, the heart weight was lower(142.0 ± 8.7 mg vs. 179.0 ± 10.4 mg, P < .001), the heart weight to body weight ratio (HW/BW) was similar, the E/A ratio was higher (1.80 ± 0.47 vs. 1.44 ± 0.26, P < .05), and the left ventricular end diastolic diameter (LVEDd) was significantly lower (2.72 ± 0.51 mm vs.3.54 ± 0.43 mm, P < .001) in the NONO KO mice. We also found excessive matrix deposition in vivo. In vitro, NONO deficiency led to fibroblasts hyperproliferation, while migration was inhibited, which would induce collagen maturation and deposition. Conversely, overexpression of NONO inhibited fibroblasts proliferation and increased migration which reduced collagen deposition. RNA-seq of cardiac fibroblasts further indicated that NONO deficiency upregulated the cell cycle regulators, which included cyclin B2, the origin recognition complex 1 (ORC1) and cell division cycle 6 (CDC6), while downregulated the migration regulators, which included myosins, integrin and coagulation factor II. Overexpression of NONO further verified the effects of these indicators. In conclusion, our study demonstrated that NONO deficiency was associated with developing heart defects in mice. Hyperproliferation of cardiac fibroblasts with dramatically excessive collagen secretion might be the cause of heart defects of NONO KO mice.

Keywords: Cardiac collagen metabolism; Cardiac fibroblasts; Heart defects; NONO; RNA-seq.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Base Sequence
  • Cell Movement
  • Cell Proliferation
  • Collagen / metabolism
  • DNA-Binding Proteins / deficiency*
  • DNA-Binding Proteins / metabolism
  • Diastole
  • Female
  • Fibroblasts / metabolism
  • Fibroblasts / pathology
  • Fibrosis
  • Heart / physiopathology*
  • Heart Ventricles / physiopathology
  • Male
  • Mice, Inbred C57BL
  • Mice, Knockout
  • Phenotype
  • RNA-Binding Proteins / metabolism

Substances

  • DNA-Binding Proteins
  • Nono protein, mouse
  • RNA-Binding Proteins
  • Collagen