Abstract

PurposeCanonical Wnt signaling is associated with glaucoma pathogenesis and intraocular pressure (IOP) regulation. Our goal was to gain insight into the influence of non-pigmented ciliary epithelium (NPCE)-derived exosomes on Wnt signaling by trabecular meshwork (TM) cells. The potential impact of exosomes on Wnt signaling in the ocular drainage system remains poorly understood.MethodsExosomes isolated from media collected from cultured NPCE cells by differential ultracentrifugation were characterized by dynamic light scattering (DLS), tunable resistive pulse sensing (TRPS), and nanoparticle tracking analysis (NTA), sucrose density gradient migration and transmission electron microscopy (TEM). The cellular target specificity of the NPCE-derived exosomes was investigated by confocal microscopy-based monitoring of the uptake of DiD-labeled exosomes over time, as compared to uptake by various cell lines. Changes in Wnt protein levels in TM cells induced by NPCE exosomes were evaluated by Western blot.ResultsExosomes derived from NPCE cells were purified and detected as small rounded 50–140 nm membrane vesicles, as defined by DLS, NTA, TRPS and TEM. Western blot analysis indicated that the nanovesicles were positive for classic exosome markers, including Tsg101 and Alix. Isolated nanoparticles were found in sucrose density fractions typical of exosomes (1.118–1.188 g/mL sucrose). Using confocal microscopy, we demonstrated time-dependent specific accumulation of the NPCE-derived exosomes in NTM cells. Other cell lines investigated hardly revealed any exosome uptake. We further showed that exosomes induced changes in Wnt signaling protein expression in the TM cells. Western blot analysis further revealed decreased phosphorylation of GKS3β and reduced β-catenin levels. Finally, we found that treatment of NTM cells with exosomes resulted in a greater than 2-fold decrease in the level of β-catenin in the cytosolic fraction. In contrast, no remarkable difference in the amount of β-catenin in the nuclear fraction was noted, relative to the control.ConclusionsThe data suggest that NPCE cells release exosome-like vesicles and that these nanoparticles affect canonical Wnt signaling in TM cells. These findings may have therapeutic relevance since canonical Wnt pathway is involved in intra-ocular pressure regulation. Further understanding of NPCE-derived exosome-responsive signaling pathways may reveal new targets for pharmacological intervention within the drainage system as a target for glaucoma therapy.

Highlights

  • Glaucoma and the ocular drainage systemThe privilege of age-related diseases is predicted to increase in the coming year and glaucoma patient number worldwide is expected reach 80 million by 2020, despite current advances in therapy[1].Glaucoma is characterized by the ongoing deterioration of the retinal ganglion layer and worsening of visual field defects, accompanied changes in the optic nerve head

  • Exosomes isolated from media collected from cultured non-pigmented ciliary epithelium (NPCE) cells by differential ultracentrifugation were characterized by dynamic light scattering (DLS), tunable resistive pulse sensing (TRPS), and nanoparticle tracking analysis (NTA), sucrose density gradient migration and transmission electron microscopy (TEM)

  • Exosomes derived from NPCE cells were purified and detected as small rounded 50–140 nm membrane vesicles, as defined by DLS, NTA, TRPS and TEM

Read more

Summary

Introduction

Glaucoma and the ocular drainage systemThe privilege of age-related diseases is predicted to increase in the coming year and glaucoma patient number worldwide is expected reach 80 million by 2020, despite current advances in therapy[1].Glaucoma is characterized by the ongoing deterioration of the retinal ganglion layer and worsening of visual field defects, accompanied changes in the optic nerve head. Further support for communication between the ocular drainage systems came from cell culture experiments where co-cultured non-pigmented ciliary epithelium (NPCE) and TM cells induced significant increases in the activity of some phosphatases and MMPs (matrix metalloproteinases), MMP-2 and MMP-9,) in TM cells [9]. Still, it remains mechanistically unclear how active molecule proteins and enzymes known to be located in the intracellular moiety, could function in the AH which is found in the extracellular milieu. The involvement of Extracellular vesicles (EV) offers one possible solution

Objectives
Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call