Activation of Prostaglandin FP and EP2 Receptors Differently Modulates Myofibroblast Transition in a Model of Adult Primary Human Trabecular Meshwork Cells

Invest Ophthalmol Vis Sci. 2016 Apr;57(4):1816-25. doi: 10.1167/iovs.15-17693.

Abstract

Purpose: Prostaglandin F2α analogues are the first-line medication for the treatment of ocular hypertension (OHT), and prostanoid EP2 receptor agonists are under clinical development for this indication. The goal of this study was to investigate the effects of F prostanoid (FP) and EP2 receptor activation on the myofibroblast transition of primary trabecular meshwork (TM) cells, which could be a causal mechanism of TM dysfunction in glaucoma.

Methods: Human primary TM cells were treated with either latanoprost or butaprost and TGF-β2. Trabecular meshwork contraction was measured in a three-dimensional (3D) TM cell-populated collagen gel (CPCG) model. Expression of α-smooth muscle actin (α-SMA) and phosphorylation of myosin light chain (MLC) were determined by Western blot. Assembly of actin stress fibers and collagen deposition were evaluated by immunocytochemistry. Involvement of p38, extracellular signal-regulated kinase (ERK), and Rho-associated kinase (ROCK) pathways as well as matrix metalloproteinase activation was tested with specific inhibitors.

Results: In one source of validated adult TM cells, latanoprost induced cell contraction as observed by CPCG surface reduction and increased actin polymerization, α-SMA expression, and MLC phosphorylation, whereas butaprost inhibited TGF-β2-induced CPCG contraction, actin polymerization, and MLC phosphorylation. Both agonists inhibited TGF-β2-dependent collagen deposition. The latanoprost effects were mediated by p38 pathway.

Conclusions: Latanoprost decreased TM collagen accumulation but promoted a contractile phenotype in a source of adult TM cells that could modulate the conventional outflow pathway. In contrast, butaprost attenuated both TM contraction and collagen deposition induced by TGF-β2, thereby inhibiting myofibroblast transition of TM cells. These results open new perspectives for the management of OHT.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Actins / biosynthesis
  • Actins / genetics
  • Adult
  • Alprostadil / analogs & derivatives
  • Alprostadil / pharmacology
  • Animals
  • Antihypertensive Agents / pharmacology
  • Blotting, Western
  • Cell Survival
  • Cells, Cultured
  • Dinoprost
  • Glaucoma / drug therapy*
  • Glaucoma / genetics
  • Glaucoma / metabolism
  • Humans
  • Immunohistochemistry
  • Latanoprost
  • Male
  • Myofibroblasts / drug effects
  • Myofibroblasts / metabolism*
  • Myofibroblasts / pathology
  • Myosin Light Chains / metabolism
  • Neuroprotective Agents
  • Prostaglandins E, Synthetic
  • Prostaglandins F, Synthetic / pharmacology*
  • RNA / genetics*
  • Rats
  • Real-Time Polymerase Chain Reaction
  • Receptors, Prostaglandin / drug effects*
  • Receptors, Prostaglandin / metabolism
  • Receptors, Prostaglandin E, EP2 Subtype / drug effects*
  • Receptors, Prostaglandin E, EP2 Subtype / metabolism
  • Signal Transduction / drug effects
  • Trabecular Meshwork / drug effects
  • Trabecular Meshwork / metabolism*
  • Trabecular Meshwork / pathology

Substances

  • ACTA2 protein, human
  • Actins
  • Antihypertensive Agents
  • Myosin Light Chains
  • Neuroprotective Agents
  • Prostaglandins E, Synthetic
  • Prostaglandins F, Synthetic
  • Receptors, Prostaglandin
  • Receptors, Prostaglandin E, EP2 Subtype
  • prostaglandin F2alpha receptor
  • RNA
  • Latanoprost
  • Dinoprost
  • Alprostadil
  • butaprost