Periostin overexpression in collecting ducts accelerates renal cyst growth and fibrosis in polycystic kidney disease

Am J Physiol Renal Physiol. 2018 Dec 1;315(6):F1695-F1707. doi: 10.1152/ajprenal.00246.2018. Epub 2018 Oct 17.

Abstract

In polycystic kidney disease (PKD), persistent activation of cell proliferation and matrix production contributes to cyst growth and fibrosis, leading to progressive deterioration of renal function. Previously, we showed that periostin, a matricellular protein involved in tissue repair, is overexpressed by cystic epithelial cells of PKD kidneys. Periostin binds αVβ3-integrins and activates integrin-linked kinase (ILK), leading to Akt/mammalian target of rapamycin (mTOR)-mediated proliferation of human PKD cells. By contrast, periostin does not stimulate the proliferation of normal human kidney cells. This difference in the response to periostin is due to elevated expression of αVβ3-integrins by cystic cells. To determine whether periostin accelerates cyst growth and fibrosis, we generated mice with conditional overexpression of periostin in the collecting ducts (CDs). Ectopic CD expression of periostin was not sufficient to induce cyst formation or fibrosis in wild-type mice. However, periostin overexpression in pcy/pcy ( pcy) kidneys significantly increased mTOR activity, cell proliferation, cyst growth, and interstitial fibrosis; and accelerated the decline in renal function. Moreover, CD-specific overexpression of periostin caused a decrease in the survival of pcy mice. These pathological changes were accompanied by increased renal expression of vimentin, α-smooth muscle actin, and type I collagen. We also found that periostin increased gene expression of pathways involved in repair, including integrin and growth factor signaling and ECM production, and it stimulated focal adhesion kinase, Rho GTPase, cytoskeletal reorganization, and migration of PKD cells. These results suggest that periostin stimulates signaling pathways involved in an abnormal tissue repair process that contributes to cyst growth and fibrosis in PKD.

Keywords: autosomal dominant polycystic kidney disease; extracellular matrix; integrin; matricellular protein; proliferation.

Publication types

  • Research Support, N.I.H., Extramural
  • Research Support, Non-U.S. Gov't

MeSH terms

  • Adult
  • Aged
  • Animals
  • Case-Control Studies
  • Cell Adhesion Molecules / genetics
  • Cell Adhesion Molecules / metabolism*
  • Cell Movement
  • Cell Proliferation*
  • Cells, Cultured
  • Disease Models, Animal
  • Disease Progression
  • Epithelial Cells / metabolism*
  • Epithelial Cells / pathology
  • Extracellular Matrix / metabolism
  • Extracellular Matrix / pathology
  • Female
  • Fibrosis
  • Gene Expression Regulation
  • Genetic Predisposition to Disease
  • Humans
  • Kidney Tubules, Collecting / metabolism*
  • Kidney Tubules, Collecting / pathology
  • Male
  • Mice, Transgenic
  • Middle Aged
  • Phenotype
  • Polycystic Kidney, Autosomal Dominant / genetics
  • Polycystic Kidney, Autosomal Dominant / metabolism*
  • Polycystic Kidney, Autosomal Dominant / pathology
  • Receptors, Cell Surface / genetics
  • Signal Transduction
  • Time Factors
  • Up-Regulation

Substances

  • Cell Adhesion Molecules
  • POSTN protein, human
  • Pkhd1 protein, mouse
  • Postn protein, mouse
  • Receptors, Cell Surface