4.7 Article

In vitrostem cell modelling demonstrates a proof-of-concept for excess functional mutantTIMP3as the cause ofSorsbyfundusdystrophy

Journal

JOURNAL OF PATHOLOGY
Volume 252, Issue 2, Pages 138-150

Publisher

WILEY
DOI: 10.1002/path.5506

Keywords

Sorsby fundus dystrophy; human induced pluripotent stem cell; retinal pigment epithelial cell; retinal degeneration; metalloproteinase inhibitor 3

Funding

  1. Academy of Finland [315085]
  2. Finnish Cultural Foundation
  3. Mary and Georg C. Ehrnrooth's Foundation
  4. Sokeain Ystavatry
  5. Retina UK [GR590]
  6. Alzheimer's Research UK South Coast Network
  7. Gift of Sight Appeal
  8. Academy of Finland (AKA) [315085, 315085] Funding Source: Academy of Finland (AKA)

Ask authors/readers for more resources

Sorsby fundus dystrophy (SFD) is a rare autosomal dominant disease of the macula that leads to bilateral loss of central vision and is caused by mutations in theTIMP3gene. However, the mechanisms by whichTIMP3mutations cause SFD are poorly understood. Here, we generated human induced pluripotent stem cell-derived retinal pigmented epithelial (hiPSC-RPE) cells from three SFD patients carrying TIMP3 p.(Ser204Cys) and three non-affected controls to study disease-related structural and functional differences in the RPE. SFD-hiPSC-RPE exhibited characteristic RPE structure and physiology but showed significantly reduced transepithelial electrical resistance associated with enriched expression of cytoskeletal remodelling proteins. SFD-hiPSC-RPE exhibited basolateral accumulation of TIMP3 monomers, despite no change inTIMP3gene expression. TIMP3 dimers were observed in both SFD and control hiPSC-RPE, suggesting that mutant TIMP3 dimerisation does not drive SFD pathology. Furthermore, mutant TIMP3 retained matrix metalloproteinase activity. Proteomic profiling showed increased expression of ECM proteins, endothelial cell interactions and angiogenesis-related pathways in SFD-hiPSC-RPE. By contrast, there were no changes in VEGF secretion. However, SFD-hiPSC-RPE secreted higher levels of monocyte chemoattractant protein 1, PDGF and angiogenin. Our findings provide a proof-of-concept that SFD patient-derived hiPSC-RPE mimic mature RPE cells and support the hypothesis that excess accumulation of mutant TIMP3, rather than an absence or deficiency of functional TIMP3, drives ECM and angiogenesis-related changes in SFD. (c) 2020 The Authors. TheJournal of Pathologypublished by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available