A Combination Therapy Targeting Endoglin and VEGF-A Prevents Subretinal Fibro-Neovascularization Caused by Induced Müller Cell Disruption

Invest Ophthalmol Vis Sci. 2018 Dec 3;59(15):6075-6088. doi: 10.1167/iovs.18-25628.

Abstract

Purpose: Subretinal fibroneovascularization is one of the most common causes of vision loss in neovascular AMD (nAMD). Anti-VEGF therapy effectively inhibits vascular leak and neovascularization but has little effect on fibrosis. This study aimed to identify a combination therapy to concurrently inhibit subretinal neovascularization and prevent fibrosis.

Methods: We generated transgenic mice in which induced disruption of Müller cells leads to subretinal neovascularization, which is reliably accompanied by subretinal fibrosis. We conducted Western blots and immunohistochemistry to study changes in transforming growth factor-β (TGFβ) signaling including endoglin, a coreceptor essential for TGFβ signaling, and then tested the effects of monthly intravitreal injection of anti-VEGF-A and anti-endoglin, either alone or in combination, on the development of subretinal fibroneovascularization in our transgenic mice.

Results: Müller cell disruption increased expression of TGFβ1, TGFβ type 1 receptor, and phosphorylated-Smad3. Endoglin was strongly expressed in subretinal fibroneovascular tissue. Fluorescein angiography and measurements of retinal vascular permeability indicated that intravitreal anti-VEGF-A in combination with anti-endoglin treatment more efficiently inhibited vascular leak compared with either monotherapy. Immunostaining of retinal wholemounts with antibodies against glial fibrillary acidic protein and ionized calcium binding adaptor molecule 1 indicated that the combination therapy also effectively prevented subretinal fibrosis and inhibited microglial activation. Luminex cytokine assays indicated that intravitreal anti-VEGF-A and anti-endoglin treatment, either alone or in combination, reduced the production of IL33 and macrophage inflammatory protein-3α.

Conclusions: Our findings offer a potentially novel combination approach to concurrently managing subretinal neovascularization and fibrosis in nAMD.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Antibodies, Monoclonal / pharmacology*
  • Blotting, Western
  • Calcium-Binding Proteins / metabolism
  • Capillary Permeability / drug effects
  • Drug Combinations
  • Endoglin / immunology*
  • Ependymoglial Cells / metabolism
  • Ependymoglial Cells / pathology*
  • Fibrosis / metabolism
  • Fibrosis / prevention & control
  • Fluorescein Angiography
  • Glial Fibrillary Acidic Protein / metabolism
  • Immunohistochemistry
  • Intravitreal Injections
  • Mice
  • Mice, Transgenic
  • Microfilament Proteins / metabolism
  • Phosphorylation
  • Receptor, Transforming Growth Factor-beta Type I / metabolism
  • Retina / metabolism
  • Retina / pathology*
  • Retinal Neovascularization / etiology
  • Retinal Neovascularization / metabolism
  • Retinal Neovascularization / prevention & control*
  • Retinal Vessels
  • Smad3 Protein / metabolism
  • Transforming Growth Factor beta1 / metabolism
  • Vascular Endothelial Growth Factor A / immunology*

Substances

  • Aif1 protein, mouse
  • Antibodies, Monoclonal
  • Calcium-Binding Proteins
  • Drug Combinations
  • Endoglin
  • Eng protein, mouse
  • Glial Fibrillary Acidic Protein
  • Microfilament Proteins
  • Smad3 Protein
  • Smad3 protein, mouse
  • Transforming Growth Factor beta1
  • Vascular Endothelial Growth Factor A
  • glial fibrillary astrocytic protein, mouse
  • vascular endothelial growth factor A, mouse
  • Receptor, Transforming Growth Factor-beta Type I