In understanding the mechanisms of pathological retinal angiogenesis, we have discovered that besides VEGFA, VEGFB, VEGFC, DLL4 and cleaved NOTCH1 induction, hypoxia activates CREB and STAT3 in the mouse retina in a robust and sustained manner. Interestingly enough, blockade of either CREB or STAT3 inhibits VEGFC and FGF2-induced expression of DLL4 and cleaved NOTCH1 in HRMVECs. A large number of studies suggest that NOTCH signaling by modulating the tip versus stalk cell differentiation plays a pivotal role in developmental and postnatal angiogenesis. CREB belongs to a basic/leucine zipper (bZIP) family of transcriptional factors and possess the ability to bind o the consensus sequence TGAC/GTCA in the promoter regions of genes enhancing their expression in response to a wide variety of stimulants, including cytokines, growth factors and hormones and is involved in the regulation of neuronal plasticity, long-term memory and cellular metabolism. Similarly, STAT3, which belongs to a family of transcriptional factors and whose activity is regulated by Tyr/Ser phosphorylation and acetylation, possess the ability to bind to th consensus sequence TTCCGGGAA in the promoter regions of genes inducing their expression in response to a large spectrum of stimulants, including cytokines, growth factors and hormones and is involved in the modulation of cellular growth and migration. While a large body of data demonstrates the importance of these transcriptional factors in the regulation of many cellular processes, including cell proliferation, migration and differentiation, literally nothing is known n regard to their role in the regulation of pathological retinal angiogenesis, particularly in concer with NOTCH signaling. Based on our novel preliminary observations, we hypothesize that CREB and STAT3 via enhancing the expression of DLL4 and thereby NOTCH1 signaling mediates pathological retinal neovascularization. To address this hypothesis, we will test the following three specific aims:
Aim 1. To test the hypothesis that CREB mediates hypoxia-induced pathological retinal neovascularization.
Aim 2. To test the hypothesis that STAT3 mediates hypoxia-induced pathological retinal neovascularization.
Aim 3. To test the hypothesis that hypoxia induces DLL4-NOTCH1 signaling via CREB and STAT3 leading to leaky and non- patterning pathological retinal neovascularization. The execution of the experiments proposed in this grant application should provide us novel information on the upstream mechanisms of NOTCH signaling activation in mediating pathological retinal neovascularization and such knowledge would be useful in the development of therapeutic drugs against this debilitating ocular disease.

Public Health Relevance

Retinopathies/proliferative retinopathies and age-related macular degeneration are the leading causes of vision loss. The central factor associated with these diseases is increased vascular permeability and formation of new blood vessels that are more vulnerable to hemorrhage. Therefore, understanding the mechanisms by which these new blood vessels develop may provide useful information for developing effective anti-angiogenesis based therapeutics against these ocular diseases.

Agency
National Institute of Health (NIH)
Institute
National Eye Institute (NEI)
Type
Research Project (R01)
Project #
2R01EY014856-07
Application #
8453956
Study Section
Atherosclerosis and Inflammation of the Cardiovascular System Study Section (AICS)
Program Officer
Shen, Grace L
Project Start
2003-07-01
Project End
2016-11-30
Budget Start
2012-12-01
Budget End
2013-11-30
Support Year
7
Fiscal Year
2013
Total Cost
$375,000
Indirect Cost
$125,000
Name
University of Tennessee Health Science Center
Department
Physiology
Type
Schools of Medicine
DUNS #
941884009
City
Memphis
State
TN
Country
United States
Zip Code
38163
Singh, Nikhlesh K; Rao, Gadiparthi N (2018) Emerging role of 12/15-Lipoxygenase (ALOX15) in human pathologies. Prog Lipid Res 73:28-45
Kumar, Raj; Singh, Nikhlesh K; Rao, Gadiparthi N (2016) Proline-rich tyrosine kinase 2 via enhancing signal transducer and activator of transcription 3-dependent cJun expression mediates retinal neovascularization. Sci Rep 6:26480
Kumar, Raj; Janjanam, Jagadeesh; Singh, Nikhlesh K et al. (2016) A new role for cofilin in retinal neovascularization. J Cell Sci 129:1234-49
Singh, Nikhlesh K; Kotla, Sivareddy; Kumar, Raj et al. (2015) Cyclic AMP Response Element Binding Protein Mediates Pathological Retinal Neovascularization via Modulating DLL4-NOTCH1 Signaling. EBioMedicine 2:1767-84
Singh, Nikhlesh K; Hansen 3rd, Dale E; Kundumani-Sridharan, Venkatesh et al. (2013) Both Kdr and Flt1 play a vital role in hypoxia-induced Src-PLD1-PKC?-cPLA(2) activation and retinal neovascularization. Blood 121:1911-23
Keilani, Serene; Chandwani, Samira; Dolios, Georgia et al. (2012) Egr-1 induces DARPP-32 expression in striatal medium spiny neurons via a conserved intragenic element. J Neurosci 32:6808-18
Zhang, Qiuhua; Wang, Dong; Singh, Nikhlesh K et al. (2011) Activation of cytosolic phospholipase A2 downstream of the Src-phospholipase D1 (PLD1)-protein kinase C ? (PKC?) signaling axis is required for hypoxia-induced pathological retinal angiogenesis. J Biol Chem 286:22489-98
Zhang, Qiuhua; Wang, Dong; Kundumani-Sridharan, Venkatesh et al. (2010) PLD1-dependent PKCgamma activation downstream to Src is essential for the development of pathologic retinal neovascularization. Blood 116:1377-85
Zhao, Tieqiang; Wang, Dong; Cheranov, Sergey Y et al. (2009) A novel role for activating transcription factor-2 in 15(S)-hydroxyeicosatetraenoic acid-induced angiogenesis. J Lipid Res 50:521-33
Cheranov, Sergey Y; Wang, Dong; Kundumani-Sridharan, Venkatesh et al. (2009) The 15(S)-hydroxyeicosatetraenoic acid-induced angiogenesis requires Janus kinase 2-signal transducer and activator of transcription-5B-dependent expression of interleukin-8. Blood 113:6023-33

Showing the most recent 10 out of 11 publications