Diabetic retinopathy (DR) and retinopathy of prematurity (ROP) are characterized by vessel loss followed by hyper-vascularization at the non-vascularized border. These vessels fail to grow into and rescue hypoxic retina and are misdirected towards the vitreous with formation of contractile bands and retinal detachment. If hypoxic retina could revascularize properly, destructive neovascularization (NV) would not occur. The misdirected NV is thought to result from high levels of pro-angiogenic factors released from hypoxic retina into the vitreous. Yet levels are even higher in hypoxic retina. We hypothesize that vaso-repulsive factors from stressed neurons prevent revascularization. Neuronal guidance cues, Semaphorins (Sema) and their receptor Neuropilin (Nrp) might be shared with vessels to direct this misguided growth. Nrp-1 binds opposing factors Sema3A and VEGF. Sema3A provokes EC apoptosis, inhibits VEGF dependant chemotaxis and could mediate cross talk between vessels and neurons. Hypothesis: Stressed ganglion cells (RGC) produce Sema3A which promotes vessel loss via EC apoptosis and repels growing neovessels from hypoxic retina towards vitreous. Suppressing Sema3A promotes revascularization and prevents retinopathy. Preliminarily we find that Sema3A is induced in stressed RGCs by hyperglycemia as well as in oxygen- induced retinopathy (OIR). Sema3A is elevated >25X in vitreous with proliferative DR. Blocking Sema3A in RGCs in OIR suppresses vascular loss (VO), improves revascularization of hypoxic retina and decreases pathological NV, suggesting a fundamentally new approach to treat retinopathy. We will test this hypothesis in diabetes models, in OIR, in vitro (RGCs and ECs) and ex vivo with aortic explants with O2 and glucose stress simulating phases I, II of retinopathy. We will:
AIM 1 temporally quantify and localize retinal Sema3A, Nrp1, VEGF in DR, OIR.
AIM 2 in DR, OIR determine if Sema3A suppression in retina or in RGCs suppresses retinopathy in mice with mutant Nrp-1 binding VEGF but not Sema3A and in WT mice with lentivirus (Lv)-driven shRNA targeting Sema3A in RGCs.
AIM 3 in vitro determine competition between Sema3A and VEGF on vessel guidance and EC apoptosis and determine molecular signaling mechanisms. The proposed studies are pioneering as they would establish neuronal influence on vaso-degeneration and the importance of vascular repulsive cues in retinopathy. Our results suggest that the Sema3A-Nrp axis is likely to be an attractive therapeutic target to promote revascularization in retinopathy and other pathologies such as cancer and cerebral- vascular infarcts where vascular re-growth is a key determinant of area of injury.

Public Health Relevance

Diabetic retinopathy and retinopathy of prematurity are leading causes of blindness and severely reduce the quality of life. Current laser treatment destroys hypoxic neural retina to prevent release of vascular growth factors. We need to more fundamentally address the pathology of retinopathy to promote normal vascular growth. This proposed work will address manipulation of neuronal guidance cues to promote revascularization of the retina.

Agency
National Institute of Health (NIH)
Institute
National Eye Institute (NEI)
Type
Research Project (R01)
Project #
5R01EY022275-03
Application #
8656349
Study Section
Biology and Diseases of the Posterior Eye Study Section (BDPE)
Program Officer
Shen, Grace L
Project Start
2012-05-01
Project End
2015-04-30
Budget Start
2014-05-01
Budget End
2015-04-30
Support Year
3
Fiscal Year
2014
Total Cost
$427,052
Indirect Cost
$181,620
Name
Children's Hospital Boston
Department
Type
DUNS #
076593722
City
Boston
State
MA
Country
United States
Zip Code
02115
Fu, Zhongjie; Gong, Yan; Liegl, Raffael et al. (2017) FGF21 Administration Suppresses Retinal and Choroidal Neovascularization in Mice. Cell Rep 18:1606-1613
Lundgren, Pia; Lundberg, Linnea; Hellgren, Gunnel et al. (2017) Aggressive Posterior Retinopathy of Prematurity Is Associated with Multiple Infectious Episodes and Thrombocytopenia. Neonatology 111:79-85
Fu, Zhongjie; Gong, Yan; Löfqvist, Chatarina et al. (2016) Review: adiponectin in retinopathy. Biochim Biophys Acta 1862:1392-400
Klevebro, S; Lundgren, P; Hammar, U et al. (2016) Cohort study of growth patterns by gestational age in preterm infants developing morbidity. BMJ Open 6:e012872
Joyal, Jean-Sébastien; Sun, Ye; Gantner, Marin L et al. (2016) Retinal lipid and glucose metabolism dictates angiogenesis through the lipid sensor Ffar1. Nat Med 22:439-45
Gong, Yan; Fu, Zhongjie; Edin, Matthew L et al. (2016) Cytochrome P450 Oxidase 2C Inhibition Adds to ?-3 Long-Chain Polyunsaturated Fatty Acids Protection Against Retinal and Choroidal Neovascularization. Arterioscler Thromb Vasc Biol 36:1919-27
Gong, Yan; Li, Jie; Sun, Ye et al. (2015) Optimization of an Image-Guided Laser-Induced Choroidal Neovascularization Model in Mice. PLoS One 10:e0132643
Lundgren, Pia; Stoltz Sjöström, Elisabeth; Domellöf, Magnus et al. (2015) The Specificity of the WINROP Algorithm Can Be Significantly Increased by Reassessment of the WINROP Alarm. Neonatology 108:152-6
Sun, Ye; Ju, Meihua; Lin, Zhiqiang et al. (2015) SOCS3 in retinal neurons and glial cells suppresses VEGF signaling to prevent pathological neovascular growth. Sci Signal 8:ra94
Hellgren, Gunnel; Engström, Eva; Smith, Lois E et al. (2015) Effect of Preterm Birth on Postnatal Apolipoprotein and Adipocytokine Profiles. Neonatology 108:16-22

Showing the most recent 10 out of 39 publications