TNFα Amplifies DNaseI Expression in Renal Tubular Cells while IL-1β Promotes Nuclear DNaseI Translocation in an Endonuclease-Inactive Form

PLoS One. 2015 Jun 11;10(6):e0129485. doi: 10.1371/journal.pone.0129485. eCollection 2015.

Abstract

We have demonstrated that the renal endonuclease DNaseI is up-regulated in mesangial nephritis while down-regulated during progression of the disease. To determine the basis for these reciprocal DNaseI expression profiles we analyse processes accounting for an early increase in renal DNaseI expression. Main hypotheses were that i. the mesangial inflammation and secreted pro-inflammatory cytokines directly increase DNaseI protein expression in tubular cells, ii. the anti-apoptotic protein tumor necrosis factor receptor-associated protein 1 (Trap 1) is down-regulated by increased expression of DNaseI due to transcriptional interference, and iii. pro-inflammatory cytokines promote nuclear translocation of a variant of DNaseI. The latter hypothesis emerges from the fact that anti-DNaseI antibodies stained tubular cell nuclei in murine and human lupus nephritis. The present study was performed on human tubular epithelial cells stimulated with pro-inflammatory cytokines. Expression of the DNaseI and Trap 1 genes was determined by qPCR, confocal microscopy, gel zymography, western blot and by immune electron microscopy. Results from in vitro cell culture experiments were analysed for biological relevance in kidneys from (NZBxNZW)F1 mice and human patients with lupus nephritis. Central data indicate that stimulating the tubular cells with TNFα promoted increased DNaseI and reduced Trap 1 expression, while TNFα and IL-1β stimulation induced nuclear translocation of the DNaseI. TNFα-stimulation resulted in 3 distinct effects; increased DNaseI and IL-1β gene expression, and nuclear translocation of DNaseI. IL-1β-stimulation solely induced nuclear DNaseI translocation. Tubular cells stimulated with TNFα and simultaneously transfected with IL-1β siRNA resulted in increased DNaseI expression but no nuclear translocation. This demonstrates that IL-1β promotes nuclear translocation of a cytoplasmic variant of DNaseI since translocation clearly was not dependent on DNaseI gene activation. Nuclear translocated DNaseI is shown to be enzymatically inactive, which may point at a new, yet unknown function of renal DNaseI.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Amino Acid Sequence
  • Animals
  • Case-Control Studies
  • Cell Nucleus / drug effects
  • Cell Nucleus / metabolism
  • Cells, Cultured
  • Cytokines / metabolism
  • Deoxyribonuclease I / genetics
  • Deoxyribonuclease I / metabolism*
  • Female
  • HSP90 Heat-Shock Proteins / metabolism
  • Humans
  • Interleukin-1beta / genetics
  • Interleukin-1beta / metabolism*
  • Kidney Tubules / cytology
  • Kidney Tubules / drug effects
  • Kidney Tubules / metabolism*
  • Lupus Nephritis / metabolism
  • Lupus Nephritis / pathology
  • Mice, Inbred BALB C
  • Mice, Mutant Strains
  • Molecular Sequence Data
  • Protein Transport
  • Tumor Necrosis Factor-alpha / metabolism*
  • Tumor Necrosis Factor-alpha / pharmacology

Substances

  • Cytokines
  • HSP90 Heat-Shock Proteins
  • Interleukin-1beta
  • TNF protein, human
  • TRAP-1 protein, mouse
  • TRAP1 protein, human
  • Tumor Necrosis Factor-alpha
  • Deoxyribonuclease I

Grants and funding

This study was supported by grants from Northern Norway Regional Health Authority Medical Research Program, and from University of Tromsø as Milieu support given to OPR. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.