Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Cytokine ; 143: 155542, 2021 07.
Article in English | MEDLINE | ID: mdl-33926775

ABSTRACT

Interferon-γ (IFNG) is one of the key cytokines that regulates both innate and adaptive immune responses in the body. However, the role of IFNG in the regulation of vascularization, especially in the context of Vascular endothelial growth factor A (VEGFa)-induced angiogenesis is not clarified. Here, we report that IFNG shows potent anti-angiogenic potential against VEGFa-induced angiogenesis. IFNG significantly inhibited proliferation, migration, and tube formation of Human umbilical vein endothelial cells (HUVECs) both under basal and VEGFa-treated conditions. Intriguingly, Knockdown (KD) of STAT1 abolished the inhibitory effect of IFNG on VEGFa-induced angiogenic processes in HUVECs. Furthermore, IFNG exhibited potent anti-angiogenic efficacy in the mouse model of oxygen-induced retinopathy (OIR), an in vivo model for hypoxia-induced retinal neovascularization, without induction of functional side effects. Taken together, these results show that IFNG plays a crucial role in the regulation of VEGFa-dependent angiogenesis, suggesting its potential therapeutic applicability in neovascular diseases.


Subject(s)
Interferon-gamma/therapeutic use , Ischemia/complications , Retinal Neovascularization/complications , Retinal Neovascularization/drug therapy , Animals , Cell Movement/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Down-Regulation/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Hypoxia/complications , Interferon-gamma/administration & dosage , Interferon-gamma/pharmacology , Intravitreal Injections , Mice , Neovascularization, Physiologic/drug effects , Retina/drug effects , Retina/pathology , Retina/physiopathology , Retinal Neovascularization/physiopathology , STAT1 Transcription Factor/metabolism , Signal Transduction/drug effects
2.
Invest Ophthalmol Vis Sci ; 61(10): 20, 2020 08 03.
Article in English | MEDLINE | ID: mdl-32780864

ABSTRACT

Purpose: Ciliary neurotrophic factor (CNTF) is a well-characterized neurotrophic factor currently in clinical trials for the treatment of macular telangiectasia type II. Our previous work showed that CNTF-induced STAT3 signaling is a potent inhibitor of pathologic preretinal neovascular tuft formation in the mouse model of oxygen-induced retinopathy. In this study, we investigated the effect of CNTF on outer retinal and choroidal angiogenesis and the mechanisms that underpin the observed decrease in outer retinal neovascularization following CNTF treatment. Methods: In the Vldlr-/- and laser-CNV mouse models, mice received a one-time injection (on postnatal day [P] 12 in the Vldlr-/- model and 1 day after laser in the Choroidal Neovascularization (CNV) model) of recombinant CNTF or CxCl10, and the extent of neovascular lesions was assessed 6 days posttreatment. STAT3 downstream targets affected by CNTF treatment were identified using quantitative PCR analysis. A proteome array was used to compare media conditioned by CNTF-treated and control-treated primary Müller cells to screen for CNTF-induced changes in secreted angiogenic factors. Results: Intravitreal treatment with recombinant CNTF led to significant reduction in neovascularization in the Vldlr-/- and laser-CNV mouse models. Treatment effect in the Vldlr-/- was long-lasting but time sensitive, requiring intravitreal treatment before P19. Mechanistic workup in vitro as well as in vivo confirmed significant activation of the STAT3-signaling pathway in Müller cells in response to CNTF treatment and upregulation of CxCl10. Intravitreal injections of recombinant CxCl10 significantly reduced outer retinal neovascularization in vivo in both the Vldlr-/- and laser-CNV mouse models. Conclusions: CNTF treatment indirectly affects outer retinal and choroidal neovascularization by inducing CxCl10 secretion from retinal Müller cells.


Subject(s)
Chemokine CXCL10/metabolism , Ciliary Neurotrophic Factor/therapeutic use , Retinal Neovascularization/prevention & control , Animals , Blotting, Western , Cells, Cultured , Choroidal Neovascularization/metabolism , Choroidal Neovascularization/pathology , Choroidal Neovascularization/prevention & control , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Ependymoglial Cells , Immunohistochemistry , Laser Coagulation , Mice , Mice, Inbred C57BL , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Retinal Neovascularization/metabolism , Retinal Neovascularization/pathology , STAT3 Transcription Factor/metabolism , Up-Regulation
3.
JCI Insight ; 2(24)2017 12 21.
Article in English | MEDLINE | ID: mdl-29263301

ABSTRACT

Oxygen-induced retinopathy (OIR) is a widely used model to study ischemia-driven neovascularization (NV) in the retina and to serve in proof-of-concept studies in evaluating antiangiogenic drugs for ocular, as well as nonocular, diseases. The primary parameters that are analyzed in this mouse model include the percentage of retina with vaso-obliteration (VO) and NV areas. However, quantification of these two key variables comes with a great challenge due to the requirement of human experts to read the images. Human readers are costly, time-consuming, and subject to bias. Using recent advances in machine learning and computer vision, we trained deep learning neural networks using over a thousand segmentations to fully automate segmentation in OIR images. While determining the percentage area of VO, our algorithm achieved a similar range of correlation coefficients to that of expert inter-human correlation coefficients. In addition, our algorithm achieved a higher range of correlation coefficients compared with inter-expert correlation coefficients for quantification of the percentage area of neovascular tufts. In summary, we have created an open-source, fully automated pipeline for the quantification of key values of OIR images using deep learning neural networks.


Subject(s)
Deep Learning , Retinal Neovascularization/diagnosis , Algorithms , Animals , Disease Models, Animal , Female , Image Processing, Computer-Assisted/methods , Male , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Confocal/methods , Observer Variation , Oxygen , Retinal Neovascularization/etiology , Retinal Neovascularization/pathology
4.
Circulation ; 136(2): 180-195, 2017 Jul 11.
Article in English | MEDLINE | ID: mdl-28356444

ABSTRACT

BACKGROUND: Anti-angiogenic biologicals represent an important concept for the treatment of vasoproliferative diseases. However, the need for continued treatment, the presence of nonresponders, and the risk of long-term side effects limit the success of existing therapeutic agents. Although Tspan12 has been shown to regulate retinal vascular development, nothing is known about its involvement in neovascular disease and its potential as a novel therapeutic target for the treatment of vasoproliferative diseases. METHODS: Rodent models of retinal neovascular disease, including the mouse model of oxygen-induced retinopathy and the very low density lipoprotein receptor knockout mouse model were analyzed for Tspan/ß-catenin regulation. Screening of a phage display of a human combinatorial antibody (Ab) library was used for the development of a high-affinity Ab against Tspan12. Therapeutic effects of the newly developed Ab on vascular endothelial cells were tested in vitro and in vivo in the oxygen-induced retinopathy and very low density lipoprotein receptor knockout mouse model. RESULTS: The newly developed anti-Tspan12 Ab exhibited potent inhibitory effects on endothelial cell migration and tube formation. Mechanistic studies confirmed that the Ab inhibited the interaction between Tspan12 and Frizzled-4 and effectively modulates ß-catenin levels and target genes in vascular endothelial cells. Tspan12/ß-catenin signaling was activated in response to acute and chronic stress in the oxygen-induced retinopathy and very low density lipoprotein receptor mouse model of proliferative retinopathy. Intravitreal application of the Ab showed significant therapeutic effects in both models without inducing negative side effects on retina function. Moreover, combined intravitreal injection of the Ab with a known vascular endothelial growth factor inhibitor, Aflibercept, resulted in significant enhancement of the therapeutic efficacy of each monotherapy. Combination therapy with the Tspan12 blocking antibody can be used to reduce anti-vascular endothelial growth factor doses, thus decreasing the risk of long-term off-target effects. CONCLUSIONS: Tspan12/ß-catenin signaling is critical for the progression of vasoproliferative disease. The newly developed anti-Tspan12 antibody has therapeutic effects in vasoproliferative retinopathy and can enhance the potency of existing anti- vascular endothelial growth factor agents.


Subject(s)
Retinal Neovascularization/metabolism , Signal Transduction/physiology , Tetraspanins/antagonists & inhibitors , Tetraspanins/metabolism , beta Catenin/antagonists & inhibitors , beta Catenin/metabolism , Amino Acid Sequence , Animals , Antibodies/genetics , Antibodies/pharmacology , Antibodies/therapeutic use , HEK293 Cells , Human Umbilical Vein Endothelial Cells , Humans , Intravitreal Injections , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Retinal Neovascularization/drug therapy , Signal Transduction/drug effects
5.
Invest Ophthalmol Vis Sci ; 57(10): 4017-26, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27494343

ABSTRACT

PURPOSE: Retinal vascular disease represents a major cause for vision loss in the Western world. Recent research has shown that neuronal and vascular damage are closely related in retinal disease. Ciliary neurotrophic factor (CNTF) is a well-studied neurotrophic factor that is currently being tested in clinical trials for the treatment of retinal degenerative diseases and macular telangiectasia. However, little is known about its effect on retinal vasculature. In this study, we investigate the effects of CNTF in retinal neovascular disease using the mouse model of oxygen-induced retinopathy (OIR). METHODS: Newborn pups were exposed to 75% oxygen from postnatal day (P)7 to P12 and subsequently returned to room air. Ciliary neurotrophic factor was injected intravitreally at OIR P12 and the vaso-obliterated and neovascular areas were quantified at OIR P17. Immunohistochemistry, RNA, and protein analysis were used to identify CNTF-responsive cells. In vitro experiments were performed to analyze the effect of CNTF on endothelial and astroglial cells. RESULTS: In the OIR model, CNTF facilitated capillary regrowth and attenuated preretinal neovascularization in a dose-dependent manner. The protective effect of CNTF was mediated via activation of the JAK/STAT3/SOCS3 signaling pathway. Immunohistochemical studies identified endothelial cells among others as CNTF-responsive cells in the retina. In vitro studies confirmed the anti-angiogenic effect of CNTF on endothelial cell sprouting. CONCLUSIONS: This study provides evidence for a therapeutic potential of CNTF beyond degenerative retinal disease. Vasoproliferative retinopathies may benefit from a CNTF-dependent and SOCS3-mediated angiomodulatory effect.


Subject(s)
Ciliary Neurotrophic Factor/pharmacology , Gene Expression Regulation , Retinal Degeneration/drug therapy , Retinal Neovascularization/drug therapy , Retinal Vessels/pathology , Suppressor of Cytokine Signaling 3 Protein/genetics , Up-Regulation , Animals , Animals, Newborn , Cells, Cultured , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Humans , Immunohistochemistry , Mice, Inbred C57BL , Oxygen/toxicity , Polymerase Chain Reaction , RNA/genetics , Recombinant Proteins/pharmacology , Retinal Degeneration/chemically induced , Retinal Degeneration/diagnosis , Retinal Neovascularization/genetics , Retinal Neovascularization/metabolism , Retinal Vessels/drug effects , Signal Transduction , Suppressor of Cytokine Signaling 3 Protein/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...