1
|
Abstract
Retinopathy of prematurity (ROP) is a complex disease involving development of the neural retina, ocular circulations, and other organ systems of the premature infant. The external stresses of the ex utero environment also influence the pathophysiology of ROP through interactions among retinal neural, vascular, and glial cells. There is variability among individual infants and presentations of the disease throughout the world, making ROP challenging to study. The methods used include representative animal models, cell culture, and clinical studies. This article describes the impact of maternal-fetal interactions; stresses that the preterm infant experiences; and biologic pathways of interest, including growth factor effects and cell-cell interactions, on the complex pathophysiology of ROP phenotypes in developed and emerging countries.
Collapse
|
2
|
Wang Z, Yemanyi F, Blomfield AK, Bora K, Huang S, Liu CH, Britton WR, Cho SS, Tomita Y, Fu Z, Ma JX, Li WH, Chen J. Amino acid transporter SLC38A5 regulates developmental and pathological retinal angiogenesis. eLife 2022; 11:e73105. [PMID: 36454214 PMCID: PMC9714971 DOI: 10.7554/elife.73105] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 11/17/2022] [Indexed: 12/03/2022] Open
Abstract
Amino acid (AA) metabolism in vascular endothelium is important for sprouting angiogenesis. SLC38A5 (solute carrier family 38 member 5), an AA transporter, shuttles neutral AAs across cell membrane, including glutamine, which may serve as metabolic fuel for proliferating endothelial cells (ECs) to promote angiogenesis. Here, we found that Slc38a5 is highly enriched in normal retinal vascular endothelium, and more specifically, in pathological sprouting neovessels. Slc38a5 is suppressed in retinal blood vessels from Lrp5-/- and Ndpy/- mice, both genetic models of defective retinal vascular development with Wnt signaling mutations. Additionally, Slc38a5 transcription is regulated by Wnt/β-catenin signaling. Genetic deficiency of Slc38a5 in mice substantially delays retinal vascular development and suppresses pathological neovascularization in oxygen-induced retinopathy modeling ischemic proliferative retinopathies. Inhibition of SLC38A5 in human retinal vascular ECs impairs EC proliferation and angiogenic function, suppresses glutamine uptake, and dampens vascular endothelial growth factor receptor 2. Together these findings suggest that SLC38A5 is a new metabolic regulator of retinal angiogenesis by controlling AA nutrient uptake and homeostasis in ECs.
Collapse
Affiliation(s)
- Zhongxiao Wang
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical SchoolBostonUnited States
| | - Felix Yemanyi
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical SchoolBostonUnited States
| | - Alexandra K Blomfield
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical SchoolBostonUnited States
| | - Kiran Bora
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical SchoolBostonUnited States
| | - Shuo Huang
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical SchoolBostonUnited States
| | - Chi-Hsiu Liu
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical SchoolBostonUnited States
| | - William R Britton
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical SchoolBostonUnited States
| | - Steve S Cho
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical SchoolBostonUnited States
| | - Yohei Tomita
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical SchoolBostonUnited States
| | - Zhongjie Fu
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical SchoolBostonUnited States
| | - Jian-xing Ma
- Department of Biochemistry, Wake Forest University School of MedicineWinston-SalemUnited States
| | - Wen-hong Li
- Departments of Cell Biology and of Biochemistry, University of Texas Southwestern Medical CenterDallasUnited States
| | - Jing Chen
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical SchoolBostonUnited States
| |
Collapse
|
3
|
Sarigul Sezenoz A, Akkoyun I, Helvacioglu F, Haberal N, Dagdeviren A, Bacanli D, Yilmaz G, Oto S. Antiproliferative and Mitochondrial Protective Effects of Apigenin in an Oxygen-Induced Retinopathy In Vivo Mouse Model. J Ocul Pharmacol Ther 2021; 37:580-590. [PMID: 34665015 DOI: 10.1089/jop.2021.0046] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Purpose: To investigate the effects of a common dietary flavonoid apigenin on retinal endothelial cell proliferation, retinal morphological structure, and apoptotic cell death in an oxygen-induced retinopathy (OIR) mouse model to evaluate the possibility of the use of apigenin in the treatment of ocular neovascular diseases (ONDs). Methods: Ninety-six newborn C57BL/6J mice were included. Eight groups were randomized, each including 12 mice. Two negative control groups were kept in room air: the first without any injection and the second received intravitreal (IV) dimethyl sulfoxide (DMSO), which is the solvent we used. The OIR groups were exposed to 75% ± 2% oxygen from postnatal days (PD) 7 to 12. On PD 12, the mice were randomly assigned to 6 groups: 2 OIR control groups (1 received no injection, 1 received IV-DMSO), 2 IV-apigenin groups (10 and 20 μg/mL), and 2 intraperitoneal (IP)-apigenin groups (10 and 20 mg/kg). We quantified retinal endothelial cell proliferation by counting neovascular tufts in cross-sections and examined histological and ultrastructural changes through light and electron microscopy. We evaluated apoptosis by terminal deoxynucleotidyl transferase-mediated nick end-labeling (TUNEL). Results: We detected a significant increase in endothelial cell proliferation in the OIR groups. Groups receiving apigenin, both IP and IV, had significant decreases in endothelial cells, atypical mitochondrion count, and apoptotic cells compared with the groups receiving no injections. None of the apigenin-injected groups revealed cystic degeneration or cell loss. Conclusions: Apigenin suppresses neovascularization, has antiapoptotic and antioxidative effects in an OIR mouse model, and can be considered a promising agent for treating OND. Clinical trial (Project number: DA15/19).
Collapse
Affiliation(s)
| | - Imren Akkoyun
- Department of Ophthalmology, Baskent University Faculty of Medicine, Ankara, Turkey
| | - Fatma Helvacioglu
- Department of Histology and Embryology, Baskent University Faculty of Medicine, Ankara, Turkey
| | - Nihan Haberal
- Department of Pathology, Baskent University Faculty of Medicine, Ankara, Turkey
| | - Attila Dagdeviren
- Department of Histology and Embryology, Baskent University Faculty of Medicine, Ankara, Turkey
| | - Didem Bacanli
- Baskent University Laboratory Animal Breeding and Research Center, Ankara, Turkey
| | - Gursel Yilmaz
- Department of Ophthalmology, Baskent University Faculty of Medicine, Ankara, Turkey
| | - Sibel Oto
- Department of Ophthalmology, Baskent University Faculty of Medicine, Ankara, Turkey
| |
Collapse
|
4
|
Sun Y, Ni Y, Kong N, Huang C. TLR2 signaling contributes to the angiogenesis of oxygen-induced retinopathy. Exp Eye Res 2021; 210:108716. [PMID: 34352266 DOI: 10.1016/j.exer.2021.108716] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 07/05/2021] [Accepted: 07/30/2021] [Indexed: 11/15/2022]
Abstract
PURPOSE To evaluate the role of Toll-like receptor 2 (TLR2) signaling in retinal neovascularization in a mouse model of oxygen-induced retinopathy (OIR). MATERIALS AND METHODS The OIR model was established in C57BL/6J wild type (WT) mice and TLR2-/- mice. Retinal neovascularization in the OIR model was measured by counting new vascular cell nuclei above the internal limiting membrane and analyzing flat-mounted retinas perfused with fluorescein dextran and immunostained with Griffonia Simplicifolia (GS) isolectin. The expression of TLR2 and VEGF in the retina was detected by immunofluorescence. Expression of TGF- β1, b-FGF, and IL-6 mRNA in the retina was measured by quantitative real-time PCR. RESULTS Compared to WT OIR mice, retinal neovascularization was attenuated in TLR2-/- OIR mice. The co-expressions of TLR2 and VEGF were remarkably and consistently increased in WT OIR mice; however, there was no expression of TLR2 and a significant decrease in VEGF expression in TLR2-/- OIR mice. These results suggest that TLR2 plays a central role in OIR model angiogenesis. Expression of TGF- β1, b-FGF, and IL-6 mRNA were reduced in the TLR2-/- OIR mice, suggesting that the inflammatory response induced by TLR2 relates to angiogenesis. CONCLUSION TLR2 signaling in the retina is associated with neovascularization in mice. Inflammation contributes to the activation of angiogenesis and is partially mediated through the TLR2-VEGF retinal signaling pathway.
Collapse
Affiliation(s)
- Yuying Sun
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong Province, China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong Province, China; Department of Cancer Prevention, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong Province, China
| | - Yao Ni
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, Guangdong Province, China
| | - Ning Kong
- Department of Ophthalmology, Panyu Central Hospital, Guangzhou, 510080, Guangdong Province, China.
| | - Chunyu Huang
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong Province, China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong Province, China; Department of Endoscopy, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong Province, China.
| |
Collapse
|
5
|
Dorrell MI, Kast-Woelbern HR, Botts RT, Bravo SA, Tremblay JR, Giles S, Wada JF, Alexander M, Garcia E, Villegas G, Booth CB, Purington KJ, Everett HM, Siles EN, Wheelock M, Silva JA, Fortin BM, Lowey CA, Hale AL, Kurz TL, Rusing JC, Goral DM, Thompson P, Johnson AM, Elson DJ, Tadros R, Gillette CE, Coopwood C, Rausch AL, Snowbarger JM. A novel method of screening combinations of angiostatics identifies bevacizumab and temsirolimus as synergistic inhibitors of glioma-induced angiogenesis. PLoS One 2021; 16:e0252233. [PMID: 34077449 PMCID: PMC8172048 DOI: 10.1371/journal.pone.0252233] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 05/11/2021] [Indexed: 12/15/2022] Open
Abstract
Tumor angiogenesis is critical for the growth and progression of cancer. As such, angiostasis is a treatment modality for cancer with potential utility for multiple types of cancer and fewer side effects. However, clinical success of angiostatic monotherapies has been moderate, at best, causing angiostatic treatments to lose their early luster. Previous studies demonstrated compensatory mechanisms that drive tumor vascularization despite the use of angiostatic monotherapies, as well as the potential for combination angiostatic therapies to overcome these compensatory mechanisms. We screened clinically approved angiostatics to identify specific combinations that confer potent inhibition of tumor-induced angiogenesis. We used a novel modification of the ex ovo chick chorioallantoic membrane (CAM) model that combined confocal and automated analyses to quantify tumor angiogenesis induced by glioblastoma tumor onplants. This model is advantageous due to its low cost and moderate throughput capabilities, while maintaining complex in vivo cellular interactions that are difficult to replicate in vitro. After screening multiple combinations, we determined that glioblastoma-induced angiogenesis was significantly reduced using a combination of bevacizumab (Avastin®) and temsirolimus (Torisel®) at doses below those where neither monotherapy demonstrated activity. These preliminary results were verified extensively, with this combination therapy effective even at concentrations further reduced 10-fold with a CI value of 2.42E-5, demonstrating high levels of synergy. Thus, combining bevacizumab and temsirolimus has great potential to increase the efficacy of angiostatic therapy and lower required dosing for improved clinical success and reduced side effects in glioblastoma patients.
Collapse
Affiliation(s)
- Michael I. Dorrell
- Department of Biology, Point Loma Nazarene University, San Diego, CA, United States of America
- * E-mail:
| | - Heidi R. Kast-Woelbern
- Department of Biology, Point Loma Nazarene University, San Diego, CA, United States of America
| | - Ryan T. Botts
- Department of Mathematical, Information, and Computer Sciences, Point Loma Nazarene University, San Diego, CA, United States of America
| | - Stephen A. Bravo
- Department of Biology, Dr. Michael Dorrell’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
| | - Jacob R. Tremblay
- Department of Biology, Dr. Michael Dorrell’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
| | - Sarah Giles
- Department of Biology, Dr. Michael Dorrell’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
| | - Jessica F. Wada
- Department of Biology, Dr. Michael Dorrell’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
| | - MaryAnn Alexander
- Department of Biology, Dr. Michael Dorrell’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
| | - Eric Garcia
- Department of Biology, Dr. Michael Dorrell’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
- Department of Biology, Dr. Heidi R. Kast-Woelbern’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
| | - Gabriel Villegas
- Department of Biology, Dr. Michael Dorrell’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
| | - Caylor B. Booth
- Department of Mathematical, Information, and Computer Sciences, Dr. Ryan Bott’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
| | - Kaitlyn J. Purington
- Department of Mathematical, Information, and Computer Sciences, Dr. Ryan Bott’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
| | - Haylie M. Everett
- Department of Mathematical, Information, and Computer Sciences, Dr. Ryan Bott’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
| | - Erik N. Siles
- Department of Mathematical, Information, and Computer Sciences, Dr. Ryan Bott’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
| | - Michael Wheelock
- Department of Mathematical, Information, and Computer Sciences, Dr. Ryan Bott’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
| | - Jordan A. Silva
- Department of Biology, Dr. Michael Dorrell’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
- Department of Biology, Dr. Heidi R. Kast-Woelbern’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
| | - Bridget M. Fortin
- Department of Biology, Dr. Michael Dorrell’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
- Department of Biology, Dr. Heidi R. Kast-Woelbern’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
| | - Connor A. Lowey
- Department of Biology, Dr. Michael Dorrell’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
- Department of Biology, Dr. Heidi R. Kast-Woelbern’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
| | - Allison L. Hale
- Department of Biology, Dr. Michael Dorrell’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
- Department of Biology, Dr. Heidi R. Kast-Woelbern’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
| | - Troy L. Kurz
- Department of Biology, Dr. Michael Dorrell’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
| | - Jack C. Rusing
- Department of Biology, Dr. Michael Dorrell’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
| | - Dawn M. Goral
- Department of Biology, Dr. Michael Dorrell’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
| | - Paul Thompson
- Department of Biology, Dr. Michael Dorrell’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
| | - Alec M. Johnson
- Department of Biology, Dr. Michael Dorrell’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
| | - Daniel J. Elson
- Department of Biology, Dr. Michael Dorrell’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
| | - Roujih Tadros
- Department of Biology, Dr. Michael Dorrell’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
- Department of Biology, Dr. Heidi R. Kast-Woelbern’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
| | - Charisa E. Gillette
- Department of Biology, Dr. Michael Dorrell’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
- Department of Biology, Dr. Heidi R. Kast-Woelbern’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
| | - Carley Coopwood
- Department of Biology, Dr. Michael Dorrell’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
- Department of Biology, Dr. Heidi R. Kast-Woelbern’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
| | - Amy L. Rausch
- Department of Biology, Dr. Michael Dorrell’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
- Department of Biology, Dr. Heidi R. Kast-Woelbern’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
| | - Jeffrey M. Snowbarger
- Department of Biology, Dr. Michael Dorrell’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
- Department of Biology, Dr. Heidi R. Kast-Woelbern’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
| |
Collapse
|
6
|
Ji L, Li L, Zhao Y, Liu S, Li J, Zhang J, Zhao Q, Wang S. Immunosubunit β5i Knockout Suppresses Neovascularization and Restores Autophagy in Retinal Neovascularization by Targeting ATG5 for Degradation. Invest Ophthalmol Vis Sci 2021; 61:30. [PMID: 33369639 PMCID: PMC7774061 DOI: 10.1167/iovs.61.14.30] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose To investigate the functional role of immunoproteasome subunit β5i in pathologic retinal neovascularization (RNV) and its ability to link the immunoproteasome and autophagy. Methods Oxygen-induced retinopathy (OIR) was induced in wild-type (WT) and β5i knockout (KO) mouse pups on a C57BL/6J background. Proteasome catalytic subunit expression and proteasome activity were evaluated by quantitative real-time PCR (qPCR) and proteasome activity. Retinal vascular anatomy and neovascularization were characterized and quantified by retinal vascular flat-mount staining, fluorescence angiography, platelet endothelial cell adhesion molecule (PECAM) immunostaining, and hematoxylin and eosin staining. Correlation factors, including VEGF and ICAM-1, were detected by qPCR. Autophagy was examined by transmission electron microscopy (TEM). Autophagy biomarkers, including LC3, P62, ATG5, and ATG7, were measured by immunostaining and immunoblotting. The protein interaction between β5i and ATG5 was detected by immunoprecipitation. Results We observed that β5i had the greatest effect in WT OIR mice. Fundus fluorescence angiography, retinal flat-mount staining, and PECAM staining revealed that pathologic RNV decreased in β5i KO OIR mice compared with WT OIR mice. Concurrently, TEM, immunostaining, and immunoblotting showed that autophagy was induced in β5i KO OIR mice compared to WT OIR mice through increases in autophagosome and LC3 expression and a decrease in P62. Mechanistically, β5i interacted with ATG5 and promoted its degradation, leading to autophagy inhibition and pathogenic RNV. Conclusions This study identifies a functional role for β5i in RNV regulation. β5i deletion ameliorates RNV and restores autophagy by stabilizing ATG5. These results demonstrate the potential of β5i to serve as a bridge linking the immunoproteasome and autophagy.
Collapse
Affiliation(s)
- Liyang Ji
- Department of Ophthalmology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Li Li
- Department of Ophthalmology, Zaozhuang Municipal Hospital, Zaozhuang, Shandong, China
| | - Ying Zhao
- Department of Ophthalmology, Jianping County Hospital, Chaoyang, China
| | - Shengqiang Liu
- Department of Ophthalmology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jingmin Li
- Department of Ophthalmology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jinsong Zhang
- Department of Ophthalmology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China.,Aier Excellence Eye Hospital, Shenyang, China
| | - Qi Zhao
- Department of Ophthalmology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Shuai Wang
- Department of Ophthalmology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
7
|
Bucher F, Aguilar E, Marra KV, Rapp J, Arnold J, Diaz-Aguilar S, Lange C, Agostini H, Schlunck G, Stahl A, Friedlander M. CNTF Prevents Development of Outer Retinal Neovascularization Through Upregulation of CxCl10. Invest Ophthalmol Vis Sci 2021; 61:20. [PMID: 32780864 PMCID: PMC7441336 DOI: 10.1167/iovs.61.10.20] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
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.
Collapse
Affiliation(s)
- Felicitas Bucher
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, United States.,Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Edith Aguilar
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, United States
| | - Kyle V Marra
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, United States.,Department of Bioengineering, University of California, San Diego, San Diego, California, United States
| | - Julian Rapp
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Jakob Arnold
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Sophia Diaz-Aguilar
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, United States.,Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Clemens Lange
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Hansjürgen Agostini
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Günther Schlunck
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Andreas Stahl
- Department of Ophthalmology, University Medical Center Greifswald, Greifswald, Germany
| | - Martin Friedlander
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, United States.,The Lowy Medical Research Institute, La Jolla, California, United States
| |
Collapse
|
8
|
Wang Z, Liu A, Zhang H, Wang M, Tang Q, Huang Y, Wang L. Inhibition of retinal neovascularization by VEGF siRNA delivered via bioreducible lipid-like nanoparticles. Graefes Arch Clin Exp Ophthalmol 2020; 258:2407-2418. [PMID: 32620992 DOI: 10.1007/s00417-020-04797-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 06/02/2020] [Accepted: 06/08/2020] [Indexed: 12/20/2022] Open
Abstract
PURPOSE Previously, we have demonstrated the use of lipidoid (lipid-like) nanoparticles (e.g., "1-O16B") for gene delivery to live cells, as an alternative to viral vectors. Here, we encapsulate VEGF siRNA (siVEGF) in bioreducible lipidoid nanoparticles and examine whether these nanocomplexes can reduce intravitreal neovascularization in a rodent model of oxygen-induced retinopathy (OIR). METHODS Firstly, we constructed siVEGF-nanoparticles (NPs) and transfected human umbilical vein endothelial cells, which caused significantly reduced expression of VEGF, compared to exposure to siVEGF in solution. Secondly, we compared the effect of intravitreal siVEGF-NPs and an anti-VEGF drug (ranibizumab) on retinal vascular development and VEGF mRNA/protein expression in the retinas of a rat model of OIR. RESULTS Compared to a non-functional lipid vehicle control group, the level of VEGF mRNA and protein was significantly lower in the siVEGF-NP group (p < 0.01), but the level of VEGF mRNA was not significantly lower in the ranibizumab group. Anatomically, the number of retinal neovascular endothelial nuclei that had protruded through the internal limiting membrane and the number of areas of non-perfusion of the retina were both significantly lower in the siVEGF-NP group and the ranibizumab group than in the OIR group (p < 0.01). CONCLUSION Our results demonstrate that bioreducible lipidoid nanoparticles conveying VEGF siRNA can effectively inhibit retinal neovascularization in a rodent model of OIR, and reduce the expression of VEGF mRNA and protein. This novel treatment modality could have profound implications for treating retinal angiogenic diseases.
Collapse
Affiliation(s)
- Zonghua Wang
- Department of Ophthalmology, Chinese People's Liberation Army General Hospital, Fuxing Road 28, Beijing, 100853, China.,Department of Ophthalmology, The Seventh Medical center of PLA General Hospital, Beijing, 100700, China
| | - Anqi Liu
- Department of Ophthalmology, Chinese People's Liberation Army General Hospital, Fuxing Road 28, Beijing, 100853, China.,The Chinese People's Liberation Army NO.32141, Beijing, China
| | - Han Zhang
- The 2nd Hospital of Shandong University, 247 Beiyuan Road, Jinan, 250031, China
| | - Ming Wang
- Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100049, China.,Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, the Chinese Academy of Sciences (CAS), Beijing, 100190, China
| | - Qiao Tang
- Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100049, China.,Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, the Chinese Academy of Sciences (CAS), Beijing, 100190, China
| | - Yifei Huang
- Department of Ophthalmology, Chinese People's Liberation Army General Hospital, Fuxing Road 28, Beijing, 100853, China.
| | - Liqiang Wang
- Department of Ophthalmology, Chinese People's Liberation Army General Hospital, Fuxing Road 28, Beijing, 100853, China.
| |
Collapse
|
9
|
Wang Y, Wang X, Ma Y, Wang YX, Di Y. Expression profiles of long noncoding RNAs in retinopathy of prematurity. Neural Regen Res 2020; 15:1962-1968. [PMID: 32246647 PMCID: PMC7513972 DOI: 10.4103/1673-5374.280328] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Long noncoding RNA (lncRNA) regulates the proliferation and migration of human retinal endothelial cells, as well as retinal neovascularization in diabetic retinopathy. Based on similarities between the pathogenesis of retinopathy of prematurity (ROP) and diabetic retinopathy, lncRNA may also play a role in ROP. Seven-day-old mice were administered 75 ± 2% oxygen for 5 days and normoxic air for another 5 days to establish a ROP model. Expression of lncRNA and mRNA in the retinal tissue of mice was detected by high-throughput sequencing technology, and biological functions of the resulted differentially expressed RNAs were evaluated by Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses. The results showed that compared with the control group, 57 lncRNAs were differentially expressed, including 43 upregulated and 14 downregulated, in the retinal tissue of ROP mice. Compared with control mice, 42 mRNAs were differentially expressed in the retinal tissue of ROP mice, including 24 upregulated and 18 downregulated mRNAs. Differentially expressed genes were involved in ocular development and related metabolic pathways. The differentially expressed lncRNAs may regulate ROP in mice via microRNAs and multiple signaling pathways. Our results revealed that these differentially expressed lncRNAs may be therapeutic targets for ROP treatment. This study was approved by the Medical Ethics Committee of Shengjing Hospital of China Medical University on February 25, 2016 (approval No. 2016PS074K).
Collapse
Affiliation(s)
- Yue Wang
- Department of Ophthalmology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Xue Wang
- Department of Ophthalmology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Yuan Ma
- Department of Ophthalmology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Yue-Xia Wang
- Department of Ophthalmology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Yu Di
- Department of Ophthalmology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| |
Collapse
|
10
|
Lu F, Liu Y, Guo Y, Gao Y, Piao Y, Tan S, Tang Y. Metabolomic changes of blood plasma associated with two phases of rat OIR. Exp Eye Res 2019; 190:107855. [PMID: 31669404 DOI: 10.1016/j.exer.2019.107855] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 10/18/2019] [Accepted: 10/21/2019] [Indexed: 02/05/2023]
Abstract
Although acute hyperoxia/hypoxia alternation can shift sharply physiological processes of vessel development, e.g. oxygen induced retinopathy (OIR), very little is known of metabolic products resulted from the neovascularization disorder. In this study, the influence of abnormal oxygen exposures on the plasma metabolomic profiles of rats with OIR was investigated by the gas chromatography mass spectrometry (GC-MS). Rat pups were divided into four groups, each with 12 individuals: (i) reared in room air and sampled at P12 (CT1); (ii) exposed to high oxygen for 5 days and sampled at P12 (HO1, simulating the vaso-obeliteration process (phase I)); (iii) reared in room air and sampled at P17 (CT2); (iv) exposed to high oxygen for 5 days then followed by room air for 5 days and sampled at P17 (HO2, simulating the neovasculization one (phase II)). Plasma samples were analyzed with GC-MS, resulted in 122 metabolite species. Distinct differences in the plasma metabolome were found between groups of CT1 vs. HO1, and HO1 vs. HO2, by using univariate and multivariate analyses. Alternating hyperoxia/hypoxia conditions induced significant changes of richness of proline, ornithine and glutamine, that were important components of 'arginine and proline metabolism' pathway. These metabolites contributed largely to plasma sample classification, determined with receiver operating characteristic curve analysis and were involved profoundly in the proline-dependent production of reactive oxygen species (ROS) related to the cellular redox reactions. Our results from the rat OIR model suggest proline and 'arginine and proline metabolism' pathway as the potential biomarkers for human retinopathy of prematurity (ROP) diagnosis.
Collapse
Affiliation(s)
- Fang Lu
- West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| | - Yang Liu
- Chengdu Institute of Biology, Chinese Academy of Sciences, Sichuan, 610041, China
| | - Yunlong Guo
- West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yunxia Gao
- West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yige Piao
- Chengdu Institute of Biology, Chinese Academy of Sciences, Sichuan, 610041, China
| | - Song Tan
- Chengdu Institute of Biology, Chinese Academy of Sciences, Sichuan, 610041, China
| | - Yezhong Tang
- Chengdu Institute of Biology, Chinese Academy of Sciences, Sichuan, 610041, China
| |
Collapse
|
11
|
Bucher F, Friedlander MS, Aguilar E, Kurihara T, Krohne TU, Usui Y, Friedlander M. The long dystrophin gene product Dp427 modulates retinal function and vascular morphology in response to age and retinal ischemia. Neurochem Int 2019; 129:104489. [DOI: 10.1016/j.neuint.2019.104489] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 06/08/2019] [Accepted: 06/10/2019] [Indexed: 01/07/2023]
|
12
|
Kim Y, Hong HK, Park JR, Choi W, Woo SJ, Park KH, Oh WY. Oxygen-Induced Retinopathy and Choroidopathy: In Vivo Longitudinal Observation of Vascular Changes Using OCTA. Invest Ophthalmol Vis Sci 2019; 59:3932-3942. [PMID: 30073364 DOI: 10.1167/iovs.18-24320] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose The purpose of this study was to assess the retinal and choroidal vasculatures of an oxygen-induced retinopathy (OIR) rat model using optical coherence tomography angiography (OCTA) as well as to verify the performance of OCTA for visualizing in vivo vascular alterations, longitudinally and quantitatively. Methods To induce OIR, Sprague Dawley rat pups were incubated in an 80% oxygen chamber from postnatal day 1 (P1) to P11 and returned to room air. OCTA imaging was performed in six eyes at P15, P18, P21, and P24. All eyes were imaged with ex vivo retinal flat mount immunofluorescence microscopy for comparison with OCTA. The areas of the neovascular tufts, retinal vessel tortuosities and diameters, and vessel densities of different retinal and choroidal layers were quantified. Results The neovascular tufts were observed in two OIR eyes. The tuft areas decreased spontaneously from P18 to P24. The increase in arterial tortuosity and venous dilation were observed in the OIR eyes at P15 and P18. The retardation of vascular developments was observed in the deep vascular plexus and the choroidal layer in the OIR group while the superficial vascular plexus did not show developmental delay. Conclusions This study demonstrates an application of OCTA for quantitative and longitudinal studies on in vivo vascular alterations, including neovascular tufts, increase in arterial tortuosity, venous dilation, and developmental delay in the OIR rat model.
Collapse
Affiliation(s)
- Yongjoo Kim
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology (KAIST), Yuseong-gu, Daejeon, Republic of Korea.,KI for Health Science and Technology, Korea Advanced Institute of Science and Technology (KAIST), Yuseong-gu, Daejeon, Republic of Korea
| | - Hye Kyoung Hong
- Department of Ophthalmology, Seoul National University College of Medicine, Seoul National University Bundang Hospital (SNUBH), Seongnam, Gyeongi-do, Republic of Korea
| | - Jang Ryul Park
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology (KAIST), Yuseong-gu, Daejeon, Republic of Korea.,KI for Health Science and Technology, Korea Advanced Institute of Science and Technology (KAIST), Yuseong-gu, Daejeon, Republic of Korea
| | - WooJhon Choi
- KI for Health Science and Technology, Korea Advanced Institute of Science and Technology (KAIST), Yuseong-gu, Daejeon, Republic of Korea
| | - Se Joon Woo
- Department of Ophthalmology, Seoul National University College of Medicine, Seoul National University Bundang Hospital (SNUBH), Seongnam, Gyeongi-do, Republic of Korea
| | - Kyu Hyung Park
- Department of Ophthalmology, Seoul National University College of Medicine, Seoul National University Bundang Hospital (SNUBH), Seongnam, Gyeongi-do, Republic of Korea
| | - Wang-Yuhl Oh
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology (KAIST), Yuseong-gu, Daejeon, Republic of Korea.,KI for Health Science and Technology, Korea Advanced Institute of Science and Technology (KAIST), Yuseong-gu, Daejeon, Republic of Korea
| |
Collapse
|
13
|
Küçüködük A, Helvacioglu F, Haberal N, Dagdeviren A, Bacanli D, Yilmaz G, Akkoyun I. Antiproliferative and anti-apoptotic effect of astaxanthin in an oxygen-induced retinopathy mouse model. CANADIAN JOURNAL OF OPHTHALMOLOGY 2019; 54:65-74. [PMID: 30851776 DOI: 10.1016/j.jcjo.2018.02.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 02/19/2018] [Accepted: 02/27/2018] [Indexed: 02/07/2023]
Abstract
OBJECTIVE To evaluate the impact of intravitreal (IV) and intraperitoneal (IP) astaxanthin (AST) injections on neovascular development (ND), retinal morphology, and apoptotic activity in a C57BL/6J mouse model with hyperoxia-induced retinopathy (HIR). DESIGN C57BL/6J mouse model. METHODS Two negative control groups (n = 6 each; one of which received IV sterile dimethyl sulfoxide [DMSO]) of C57BL/6J-type mice were exposed to room air. The HIR groups included 36 C57BL/6J-type mice exposed to 75% ± 2% oxygen from postnatal day (PD) 7 to PD 12. On PD 12, these mice were randomized into 6 groups (n = 6 each): 2 HIR control groups (one of which received IV-DMSO), 2 IV-AST groups (10 and 100 µg/mL), and 2 IP-AST groups (0.5 and 5 mg/kg). We measured ND by counting neovascular tufts in cross sections and examined histological, ultrastructural changes via light and electron microscopy. Apoptosis was detected using terminal deoxynucleotidyl transferase-mediated nick end-labeling. RESULTS No ND was detected in the negative control groups. ND levels were not significantly different between high- and low-dose AST for either means of administration. However, ND levels were significantly lower in the AST groups, regardless of delivery, compared to the control groups. The means of delivery (IP versus IV) also yielded significant differences in ND. The incidence of mitochondrial dysmorphology and apoptosis were lower in groups receiving AST. CONCLUSIONS AST seems to suppress ND and has anti-apoptotic activity in the HIR mouse model.
Collapse
Affiliation(s)
- Ali Küçüködük
- Department of Ophthalmology, Faculty of Medicine, Baskent University, Ankara, Turkey
| | - Fatma Helvacioglu
- Department of Histology and Embryology, Faculty of Medicine, Baskent University, Ankara, Turkey
| | - Nihan Haberal
- Department of Pathology, Faculty of Medicine, Baskent University, Ankara, Turkey
| | - Attila Dagdeviren
- Department of Histology and Embryology, Faculty of Medicine, Baskent University, Ankara, Turkey
| | - Didem Bacanli
- Breeding Center, Faculty of Medicine, Baskent University, Ankara, Turkey
| | - Gursel Yilmaz
- Department of Ophthalmology, Faculty of Medicine, Baskent University, Ankara, Turkey
| | - Imren Akkoyun
- Department of Ophthalmology, Faculty of Medicine, Baskent University, Ankara, Turkey.
| |
Collapse
|
14
|
Elshaer SL, El-Remessy AB. Deletion of p75 NTR prevents vaso-obliteration and retinal neovascularization via activation of Trk- A receptor in ischemic retinopathy model. Sci Rep 2018; 8:12490. [PMID: 30131506 PMCID: PMC6104090 DOI: 10.1038/s41598-018-30029-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 07/23/2018] [Indexed: 02/07/2023] Open
Abstract
Ischemic retinopathy is characterized by ischemia followed by retinal neovascularization (RNV) resulting in visual impairment. Given the role of neuron-secreted growth factors in regulating angiogenesis, we examined how genetic deletion of the neurotrophin receptor; p75NTR can overcome retinal ischemia using oxygen-induced retinopathy (OIR) mouse model. Wildtype (WT) or p75NTR-/- mice pups were subjected to hyperoxia (70% O2, p7-p12) then returned to normal air (relative hypoxia, p12-p17). Vascular alterations were assessed at p12 and p17 time-points. Deletion of p75NTR prevented hyperoxia-associated central vascular cell death (p12) and hypoxia-associated RNV and enhanced central vascular repair (p17). Decreased expression of apoptotic markers; preserved Akt survival signal decreased proNGF were also observed at p12. During hypoxia, deletion of p75NTR maintained VEGF and VEGFR2 activation and restored NGF/proNGF and BDNF/proBDNF levels. Deletion of p75NTR coincided with significant increases in expression and activation of NGF survival receptor, TrkA at basal and hyperoxic condition. Pharmacological inhibition of TrkA using compound K-252a (0.5 μg 1 μl-1/eye) resulted in 2-fold increase in pathological RNV and 1.34-fold increase in central vascular cell death in p75NTR-/- pups. In conclusion, deletion of p75NTR protected against retinal ischemia and prevented RNV, in part, through restoring neurotrophic support and activating TrkA receptor.
Collapse
Affiliation(s)
- Sally L Elshaer
- Augusta Biomedical Research Corporation, Augusta, GA, 30912, USA
- Charlie Norwood VA Medical Center, Augusta, GA, 30912, USA
- Ophthalmology Department, Hamilton Eye Institute, University of Tennessee Health Sciences Center, Memphis, TN, 38163, USA
| | - Azza B El-Remessy
- Augusta Biomedical Research Corporation, Augusta, GA, 30912, USA.
- Charlie Norwood VA Medical Center, Augusta, GA, 30912, USA.
| |
Collapse
|
15
|
Xiao S, Bucher F, Wu Y, Rokem A, Lee CS, Marra KV, Fallon R, Diaz-Aguilar S, Aguilar E, Friedlander M, Lee AY. Fully automated, deep learning segmentation of oxygen-induced retinopathy images. JCI Insight 2017; 2:97585. [PMID: 29263301 PMCID: PMC5752269 DOI: 10.1172/jci.insight.97585] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 11/15/2017] [Indexed: 12/29/2022] Open
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.
Collapse
Affiliation(s)
- Sa Xiao
- Department of Ophthalmology, University of Washington, Seattle, Washington, USA
| | - Felicitas Bucher
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
- Eye Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Yue Wu
- Department of Ophthalmology, University of Washington, Seattle, Washington, USA
| | - Ariel Rokem
- eScience Institute, University of Washington, Seattle, Washington, USA
| | - Cecilia S. Lee
- Department of Ophthalmology, University of Washington, Seattle, Washington, USA
| | - Kyle V. Marra
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
- Department of Bioengineering, University of California, San Diego, San Diego, California, USA
| | - Regis Fallon
- Lowy Medical Research Institute, La Jolla, California, USA
| | - Sophia Diaz-Aguilar
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Edith Aguilar
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Martin Friedlander
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
- Lowy Medical Research Institute, La Jolla, California, USA
| | - Aaron Y. Lee
- Department of Ophthalmology, University of Washington, Seattle, Washington, USA
- eScience Institute, University of Washington, Seattle, Washington, USA
- Department of Ophthalmology, Puget Sound Veteran Affairs, Seattle, Washington, USA
| |
Collapse
|
16
|
Wang JD, An Y, Zhang JS, Wan XH, Jonas JB, Xu L, Zhang W. Human bone marrow mesenchymal stem cells for retinal vascular injury. Acta Ophthalmol 2017; 95:e453-e461. [PMID: 27807930 DOI: 10.1111/aos.13154] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 05/10/2016] [Indexed: 11/30/2022]
Abstract
PURPOSE To examine the potential of intravitreally implanted human bone marrow-derived mesenchymal stem cells (BMSCs) to affect vascular repair and the blood-retina barrier in mice and rats with oxygen-induced retinopathy, diabetic retinopathy or retinal ischaemia-reperfusion damage. METHODS Three study groups (oxygen-induced retinopathy group: 18 C57BL/6J mice; diabetic retinopathy group: 15 rats; retinal ischaemia-reperfusion model: 18 rats) received BMSCs injected intravitreally. Control groups (oxygen-induced retinopathy group: 12 C57BL/6J mice; diabetic retinopathy group: 15 rats; retinal ischaemia-reperfusion model: 18 rats) received an intravitreal injection of phosphate-buffered saline. We applied immunohistological techniques to measure retinal vascularization, spectroscopic measurements of intraretinally extravasated fluorescein-conjugated dextran to quantify the blood-retina barrier breakdown, and histomorphometry to assess retinal thickness and retinal ganglion cell count. RESULTS In the oxygen-induced retinopathy model, the study group with intravitreally injected BMSCs as compared with the control group showed a significantly (p = 0.001) smaller area of retinal neovascularization. In the diabetic retinopathy model, study group and control group did not differ significantly in the amount of intraretinally extravasated dextran. In the retinal ischaemia-reperfusion model, on the 7th day after retina injury, the retina was significantly thicker in the study group than in the control group (p = 0.02), with no significant difference in the retinal ganglion cell count (p = 0.36). CONCLUSIONS Intravitreally implanted human BMSCs were associated with a reduced retinal neovascularization in the oxygen-induced retinopathy model and with a potentially cell preserving effect in the retinal ischaemia-reperfusion model. Intravitreal BMSCs may be of potential interest for the therapy of retinal vascular disorders.
Collapse
Affiliation(s)
- Jin-Da Wang
- Beijing Institute of Ophthalmology; Beijing Tongren Eye Center; Beijing Tongren Hospital of Capital Medical University; Beijing Key Laboratory of Ophthalmology and Visual Sciences; Beijing China
| | - Ying An
- Beijing Institute of Ophthalmology; Beijing Tongren Eye Center; Beijing Tongren Hospital of Capital Medical University; Beijing Key Laboratory of Ophthalmology and Visual Sciences; Beijing China
| | - Jing-Shang Zhang
- Beijing Institute of Ophthalmology; Beijing Tongren Eye Center; Beijing Tongren Hospital of Capital Medical University; Beijing Key Laboratory of Ophthalmology and Visual Sciences; Beijing China
| | - Xiu-Hua Wan
- Beijing Institute of Ophthalmology; Beijing Tongren Eye Center; Beijing Tongren Hospital of Capital Medical University; Beijing Key Laboratory of Ophthalmology and Visual Sciences; Beijing China
| | - Jost B. Jonas
- Beijing Institute of Ophthalmology; Beijing Tongren Eye Center; Beijing Tongren Hospital of Capital Medical University; Beijing Key Laboratory of Ophthalmology and Visual Sciences; Beijing China
- Department of Ophthalmology; Medical Faculty Mannheim of the Ruprecht-Karls-University Heidelberg; Seegartenklinik; Heidelberg Germany
| | - Liang Xu
- Beijing Institute of Ophthalmology; Beijing Tongren Eye Center; Beijing Tongren Hospital of Capital Medical University; Beijing Key Laboratory of Ophthalmology and Visual Sciences; Beijing China
| | - Wei Zhang
- Beijing Institute of Ophthalmology; Beijing Tongren Eye Center; Beijing Tongren Hospital of Capital Medical University; Beijing Key Laboratory of Ophthalmology and Visual Sciences; Beijing China
| |
Collapse
|
17
|
Wang JD, An Y, Zhang JS, Wan XH, Zhang W, Lanza R, Lu SJ, Jonas JB, Xu L. Retinal vascular injuries and intravitreal human embryonic stem cell-derived haemangioblasts. Acta Ophthalmol 2017. [PMID: 28636206 DOI: 10.1111/aos.13477] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
OBJECTIVE To investigate whether intravitreally applied haemangioblasts (HB) derived from human embryonic stem cells (hESCs) are helpful for the repair of vascular damage caused in animals by an oxygen-induced retinopathy (OIR), by an induced diabetic retinopathy (DR) or by an induced retinal ischaemia with subsequent reperfusion. METHODS Human embryonic stem cell-derived HBs were transplanted intravitreally into C57BL/6J mice (OIR model), into male Wistar rats with an induced DR and into male Wistar rats undergoing induced retinal ischaemia with subsequent reperfusion. Control groups of animals received an intravitreal injection of endothelial cells (ECs) or phosphate-buffered saline (PBS). We examined the vasculature integrity in the mice with OIR, the blood-retina barrier in the rats with induced DR, and retinal thickness and retinal ganglion cell density in retina flat mounts of the rats with the retinal ischaemic-reperfusion retinopathy. RESULTS In the OIR model, the study group versus control groups showed a significantly (p < 0.001) smaller retinal avascular area [5.1 ± 2.7%;n = 18 animals versus 12.2 ± 2.8% (PBS group; n = 10 animals) and versus 11.8 ± 3.7% (EC group; n = 8 animals)] and less retinal neovascularization [6.3 ± 2.5%;n = 18 versus 15.2 ± 6.3% (n = 10; PBS group) and versus 15.8 ± 3.3% (n = 8; EC group)]. On retinal flat mounts, hESC-HBs were integrated into damaged retinal vessels and stained positive for PECAM (CD31) as EC marker. In the DR model, the study group versus the EC control group showed a significantly (p = 0.001) better blood-retina barrier function as measured at 2 days after the intravitreal injections [study group: 20.2 ± 12.8 μl/(g × hr); n = 6; versus EC control group: 52.9 ± 9.9 μl/(g × hr; n = 6)]. In the retinal ischaemia-reperfusion model, the groups did not differ significantly in retinal thickness and retinal ganglion cell density at 2, 5 and 7 days after baseline. CONCLUSION By integrating into damaged retinal vessels and differentiating into ECs, intravitreally administered hESC-HBs may have partially repaired a retinal vascular injury caused by OIR model and DR.
Collapse
Affiliation(s)
- Jin-Da Wang
- Beijing Institute of Ophthalmology; Beijing Tongren Eye Center; Beijing Key Laboratory of Ophthalmology and Visual Sciences; Beijing Tongren Hospital of Capital Medical University; Beijing China
| | - Ying An
- Beijing Institute of Ophthalmology; Beijing Tongren Eye Center; Beijing Key Laboratory of Ophthalmology and Visual Sciences; Beijing Tongren Hospital of Capital Medical University; Beijing China
| | - Jing-Shang Zhang
- Beijing Institute of Ophthalmology; Beijing Tongren Eye Center; Beijing Key Laboratory of Ophthalmology and Visual Sciences; Beijing Tongren Hospital of Capital Medical University; Beijing China
| | - Xiu-Hua Wan
- Beijing Institute of Ophthalmology; Beijing Tongren Eye Center; Beijing Key Laboratory of Ophthalmology and Visual Sciences; Beijing Tongren Hospital of Capital Medical University; Beijing China
| | - Wei Zhang
- University of North Texas Health Science Center; Fort Worth Texas USA
| | - Robert Lanza
- Ocata Therapeutics; Marlborough Massachusetts USA
| | - Shi-Jiang Lu
- Ocata Therapeutics; Marlborough Massachusetts USA
| | - Jost B. Jonas
- Department of Ophthalmology; Medical Faculty Mannheim of the Ruprecht-Karls-University Heidelberg; Mannheim Germany
| | - Liang Xu
- Beijing Institute of Ophthalmology; Beijing Tongren Eye Center; Beijing Key Laboratory of Ophthalmology and Visual Sciences; Beijing Tongren Hospital of Capital Medical University; Beijing China
| |
Collapse
|
18
|
Zhang P, Wang H, Cao H, Xu X, Sun T. Insulin-Like Growth Factor Binding Protein-Related Protein 1 Inhibit Retinal Neovascularization in the Mouse Model of Oxygen-Induced Retinopathy. J Ocul Pharmacol Ther 2017; 33:459-465. [PMID: 28402720 DOI: 10.1089/jop.2016.0171] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
PURPOSE To explore the inhibitory effect of insulin-like growth factor binding protein-related protein 1 (IGFBP-rP1) on retinal angiogenesis and its underlying molecular mechanisms in the mouse model of oxygen-induced retinopathy (OIR). METHODS C57BL/6J mice were classified into three groups as control group, OIR nonintervention group, and OIR intervention group. Postnatal day 12 (P12) mice in OIR intervention group were received recombinant mouse IGFBP-rP1 (50, 100, and 200 ng/mL) intravitreal injection. Five days later, the proliferative neovascular responses were estimated by quantifying the new vessel areas in flattening retinal tissues stained by high molecular fluorescein isothiocyanate-dextran and counting the numbers of neovascular cell nuclei breaking through the internal limiting membrane in cross sections. Expressions of phospho-extracellular signal-regulated kinase 1/2 (p-ERK1/2), ERK1/2, and vascular endothelial growth factor (VEGF) proteins in retinal tissues were assessed by western blot analysis. RESULTS Irregular neovascularization, nonperfusion region, and fluorescence leakage were observed in OIR models. The expression of retinal p-ERK1/2 and VEGF proteins were significantly upregulated in OIR nonintervention group compared with control group. The area ratio of retinal new vessels and the number of neovascular cell nuclei in OIR intervention group both decreased significantly, following the downregulation of retinal p-ERK1/2 protein expression and VEGF protein expression in a dose-dependent manner. Moreover, there was no significant difference in retinal ERK1/2 protein expression. CONCLUSIONS IGFBP-rP1 inhibits retinal angiogenesis by blocking ERK signaling pathway and downregulating VEGF expression in the mouse model of OIR. It highlights the potential importance of IGFBP-rP1 serving as a target of gene therapy for retinal neovascularization in the future.
Collapse
Affiliation(s)
- Ping Zhang
- Department of Ophthalmology, Shanghai General Hospital Affiliated to Shanghai Jiao Tong University School of Medicine , Shanghai, China
| | - Hong Wang
- Department of Ophthalmology, Shanghai General Hospital Affiliated to Shanghai Jiao Tong University School of Medicine , Shanghai, China
| | - Hui Cao
- Department of Ophthalmology, Shanghai General Hospital Affiliated to Shanghai Jiao Tong University School of Medicine , Shanghai, China
| | - Xun Xu
- Department of Ophthalmology, Shanghai General Hospital Affiliated to Shanghai Jiao Tong University School of Medicine , Shanghai, China
| | - Tao Sun
- Department of Ophthalmology, Shanghai General Hospital Affiliated to Shanghai Jiao Tong University School of Medicine , Shanghai, China
| |
Collapse
|
19
|
Lee YM, Lee YR, Kim CS, Jo K, Sohn E, Kim JS, Kim J. Cnidium officinale extract and butylidenephthalide inhibits retinal neovascularization in vitro and in vivo. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2016; 16:231. [PMID: 27435599 PMCID: PMC4949763 DOI: 10.1186/s12906-016-1216-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2015] [Accepted: 07/14/2016] [Indexed: 12/20/2022]
Abstract
BACKGROUND Retinal neovascularization, which is the pathological growth of new blood vessels, is associated with retinopathy of prematurity, neovascular age-related macular degeneration, diabetic retinopathy and retinal vein occlusion. In this study, we evaluated the effect of an extract of Cnidium officinale Makino (COE) and its bioactive compound, butylidenephthalide (BP), on the migration and tube formation of human umbilical vein endothelial cells (HUVECs), and on retinal pathogenic neovascularization in the oxygen-induced retinopathy (OIR) mouse model. METHOD The HUVECs were incubated with COE and BP (0.1-10 μg/ml). The mice were exposed to 75 % oxygen for 5 days starting on the 7(th) postnatal day (P7-P12). Then, the mice were returned to room air and intraperitoneally injected with COE (100 mg/kg) and BP (5 mg/kg) once per day for 5 days (P12-P16). On P17, we measured retinal neovascularization and analyzed the angiogenesis-related proteins expression using protein arrays. RESULTS COE and BP inhibit the HUVECs migration and the tube formation in a dose-dependent manner. In addition, COE significantly decreased retinal neovascularization in the OIR mice. COE reduced the expression levels of AREG, ANG, DLL4, Endostatin, IGFBP-2 and VEGF. Additionally, BP also inhibited the retinal neovascularization and down-regulated the expression of AREG, ANG, DLL4 and VEGF. CONCLUSION These results suggest that COE and BP exerts antiangiogenic effects on retinal neovascularization by inhibiting the expression of AREG, ANG, DLL4 and VEGF, indicating that antiangiogenic activities of COE may be in part due to its bioactive compound, BP.
Collapse
|
20
|
Abstract
Abnormal blood vessel growth in the retina is a hallmark of many retinal diseases, such as retinopathy of prematurity (ROP), proliferative diabetic retinopathy, and the wet form of age-related macular degeneration. In particular, ROP has been an important health concern for physicians since the advent of routine supplemental oxygen therapy for premature neonates more than 70 years ago. Since then, researchers have explored several animal models to better understand ROP and retinal vascular development. Of these models, the mouse model of oxygen-induced retinopathy (OIR) has become the most widely used, and has played a pivotal role in our understanding of retinal angiogenesis and ocular immunology, as well as in the development of groundbreaking therapeutics such as anti-vascular endothelial growth factor injections for wet age-related macular degeneration. Numerous refinements to the model have been made since its inception in the 1950s, and technological advancements have expanded the use of the model across multiple scientific fields. In this review, we explore the historical developments that have led to the mouse OIR model utilized today, essential concepts of OIR, limitations of the model, and a representative selection of key findings from OIR, with particular emphasis on current research progress.
Collapse
Affiliation(s)
- Clifford B Kim
- Angiogenesis Laboratory, Massachusetts Eye and Ear; Department of Ophthalmology, Harvard Medical School
| | - Patricia A D'Amore
- Department of Ophthalmology, Harvard Medical School; Schepens Eye Research Institute, Massachusetts Eye and Ear; Department of Pathology, Harvard Medical School, Boston, MA, USA
| | - Kip M Connor
- Angiogenesis Laboratory, Massachusetts Eye and Ear; Department of Ophthalmology, Harvard Medical School
| |
Collapse
|
21
|
El-Hadidy AR, El-Mohandes EM, Asker SA, Ghonaim FM. A histological and immunohistochemical study of the effects of N-acetyl cysteine on retinopathy of prematurity by modifying insulin-like growth factor-1. Biotech Histochem 2016; 91:401-11. [PMID: 27149563 DOI: 10.1080/10520295.2016.1180428] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Retinopathy of prematurity (ROP) is a vasoproliferative disorder that occurs in premature infants and may lead to permanent visual impairment. We investigated both the possible protective role of N-acetyl cysteine (NAC) for preventing ROP and the role of IGF-1 in the disorder. Forty-five newborn rats were divided into three groups. Group 1 was raised in room air as controls. Group 2 was exposed to 60% oxygen for 14 days after birth, then transferred to room air. Group 3 was exposed to the same conditions as group 2, but received intraperitoneal injections of NAC on postnatal days 7-17. After 35 days, both eyes of all rats were processed for histology. Some sections were stained with hematoxylin and eosin to assess structural changes and other sections were immunostained to determine the location of IGF-1. Frozen sections also were prepared and stained for adenosine triphosphatase to detect retinal blood vessels. Compared to the controls, more blood vessels, many of which were abnormal, and increased IGF-1 expression were observed in group 2. In group 3, abnormal blood vessels and IGF-1 expression were less evident. NAC appeared to be an effective vascular-protective agent for ROP by decreasing IGF-1 expression.
Collapse
Affiliation(s)
- A R El-Hadidy
- a Histology and Cell Biology Department, Faculty of Medicine , Mansoura University , Mansoura , Egypt
| | - E M El-Mohandes
- a Histology and Cell Biology Department, Faculty of Medicine , Mansoura University , Mansoura , Egypt
| | - S A Asker
- a Histology and Cell Biology Department, Faculty of Medicine , Mansoura University , Mansoura , Egypt
| | - F M Ghonaim
- a Histology and Cell Biology Department, Faculty of Medicine , Mansoura University , Mansoura , Egypt
| |
Collapse
|
22
|
Gao X, Wang YS, Li XQ, Hou HY, Su JB, Yao LB, Zhang J. Macrophages promote vasculogenesis of retinal neovascularization in an oxygen-induced retinopathy model in mice. Cell Tissue Res 2016; 364:599-610. [DOI: 10.1007/s00441-015-2353-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 12/16/2015] [Indexed: 01/06/2023]
|
23
|
Michael IP, Westenskow PD, Hacibekiroglu S, Greenwald AC, Ballios BG, Kurihara T, Li Z, Warren CM, Zhang P, Aguilar E, Donaldson L, Marchetti V, Baba T, Hussein SM, Sung HK, Iruela-Arispe ML, Rini JM, van der Kooy D, Friedlander M, Nagy A. Local acting Sticky-trap inhibits vascular endothelial growth factor dependent pathological angiogenesis in the eye. EMBO Mol Med 2014; 6:604-23. [PMID: 24705878 PMCID: PMC4023884 DOI: 10.1002/emmm.201303708] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Current therapeutic antiangiogenic biologics used for the treatment of pathological ocular angiogenesis could have serious side effects due to their interference with normal blood vessel physiology. Here, we report the generation of novel antivascular endothelial growth factor-A (VEGF) biologics, termed VEGF “Sticky-traps,” with unique properties that allow for local inhibition of angiogenesis without detectable systemic side effects. Using genetic and pharmacological approaches, we demonstrated that Sticky-traps could locally inhibit angiogenesis to at least the same extent as the original VEGF-trap that also gains whole-body access. Sticky-traps did not cause systemic effects, as shown by uncompromised wound healing and normal tracheal vessel density. Moreover, if injected intravitreally, recombinant Sticky-trap remained localized to various regions of the eye, such as the inner-limiting membrane and ciliary body, for prolonged time periods, without gaining access either to the photoreceptors/choriocapillaris area or the circulation. These unique pharmacological characteristics of Sticky-trap could allow for safe treatment of pathological angiogenesis in patients with diabetic retinopathy and retinopathy of pre-maturity.
Collapse
Affiliation(s)
- Iacovos P Michael
- Lunenfeld-Tanenbaum Research Institute Mount Sinai Hospital, Toronto, ON, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Li J, Liu CH, Sun Y, Gong Y, Fu Z, Evans LP, Tian KT, Juan AM, Hurst CG, Mammoto A, Chen J. Endothelial TWIST1 promotes pathological ocular angiogenesis. Invest Ophthalmol Vis Sci 2014; 55:8267-77. [PMID: 25414194 DOI: 10.1167/iovs.14-15623] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
PURPOSE Pathological neovessel formation impacts many blinding vascular eye diseases. Identification of molecular signatures distinguishing pathological neovascularization from normal quiescent vessels is critical for developing new interventions. Twist-related protein 1 (TWIST1) is a transcription factor important in tumor and pulmonary angiogenesis. This study investigated the potential role of TWIST1 in modulating pathological ocular angiogenesis in mice. METHODS Twist1 expression and localization were analyzed in a mouse model of oxygen-induced retinopathy (OIR). Pathological ocular angiogenesis in Tie2-driven conditional Twist1 knockout mice were evaluated in both OIR and laser-induced choroidal neovascularization models. In addition, the effects of TWIST1 on angiogenesis and endothelial cell function were analyzed in sprouting assays of aortic rings and choroidal explants isolated from Twist1 knockout mice, and in human retinal microvascular endothelial cells treated with TWIST1 small interfering RNA (siRNA). RESULTS TWIST1 is highly enriched in pathological neovessels in OIR retinas. Conditional Tie2-driven depletion of Twist1 significantly suppressed pathological neovessels in OIR without impacting developmental retinal angiogenesis. In a laser-induced choroidal neovascularization model, Twist1 deficiency also resulted in significantly smaller lesions with decreased vascular leakage. In addition, loss of Twist1 significantly decreased vascular sprouting in both aortic ring and choroid explants. Knockdown of TWIST1 in endothelial cells led to dampened expression of vascular endothelial growth factor receptor 2 (VEGFR2) and decreased endothelial cell proliferation. CONCLUSIONS Our study suggests that TWIST1 is a novel regulator of pathologic ocular angiogenesis and may represent a new molecular target for developing potential therapeutic treatments to suppress pathological neovascularization in vascular eye diseases.
Collapse
Affiliation(s)
- Jie Li
- Department of Ophthalmology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, United States Department of Ophthalmology, West China Hospital/West China School of Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Chi-Hsiu Liu
- Department of Ophthalmology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, United States
| | - Ye Sun
- Department of Ophthalmology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, United States
| | - Yan Gong
- Department of Ophthalmology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, United States
| | - Zhongjie Fu
- Department of Ophthalmology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, United States
| | - Lucy P Evans
- Department of Ophthalmology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, United States
| | - Katherine T Tian
- Department of Ophthalmology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, United States
| | - Aimee M Juan
- Department of Ophthalmology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, United States
| | - Christian G Hurst
- Department of Ophthalmology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, United States
| | - Akiko Mammoto
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, United States
| | - Jing Chen
- Department of Ophthalmology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, United States
| |
Collapse
|
25
|
Biswal MR, Prentice HM, Dorey CK, Blanks JC. A hypoxia-responsive glial cell-specific gene therapy vector for targeting retinal neovascularization. Invest Ophthalmol Vis Sci 2014; 55:8044-53. [PMID: 25377223 DOI: 10.1167/iovs.14-13932] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
PURPOSE Müller cells, the major glial cell in the retina, play a significant role in retinal neovascularization in response to tissue hypoxia. We previously designed and tested a vector using a hypoxia-responsive domain and a glial fibrillary acidic protein (GFAP) promoter to drive green fluorescent protein (GFP) expression in Müller cells in the murine model of oxygen-induced retinopathy (OIR). This study compares the efficacy of regulated and unregulated Müller cell delivery of endostatin in preventing neovascularization in the OIR model. METHODS Endostatin cDNA was cloned into plasmids with hypoxia-regulated GFAP or unregulated GFAP promoters, and packaged into self-complementary adeno-associated virus serotype 2 vectors (scAAV2). Before placement in hyperoxia on postnatal day (P)7, mice were given intravitreal injections of regulated or unregulated scAAV2, capsid, or PBS. Five days after return to room air, on P17, neovascular and avascular areas, as well as expression of the transgene and vascular endothelial growth factor (VEGF), were compared in OIR animals treated with a vector, capsid, or PBS. RESULTS The hypoxia-regulated, glial-specific, vector-expressing endostatin reduced neovascularization by 93% and reduced the central vaso-obliteration area by 90%, matching the results with the unregulated GFAP-Endo vector. Retinas treated with the regulated endostatin vector expressed substantial amounts of endostatin protein, and significantly reduced VEGF protein. Endostatin production from the regulated vector was undetectable in retinas with undamaged vasculature. CONCLUSIONS These findings suggest that the hypoxia-regulated, glial cell-specific vector expressing endostatin may be useful for treatment of neovascularization in proliferative diabetic retinopathy.
Collapse
Affiliation(s)
- Manas R Biswal
- Integrative Biology PhD Program, Florida Atlantic University, Boca Raton, Florida, United States
| | - Howard M Prentice
- Center for Complex Systems and Brain Sciences, Charles E. Schmidt College of Science, Florida Atlantic University, Boca Raton, Florida, United States Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, Florida, United States
| | - C Kathleen Dorey
- Virginia Tech Carilion School of Medicine, Roanoke, Virginia, United States
| | - Janet C Blanks
- Center for Complex Systems and Brain Sciences, Charles E. Schmidt College of Science, Florida Atlantic University, Boca Raton, Florida, United States
| |
Collapse
|
26
|
Shen W, Chung SH, Irhimeh MR, Li S, Lee SR, Gillies MC. Systemic administration of erythropoietin inhibits retinopathy in RCS rats. PLoS One 2014; 9:e104759. [PMID: 25119659 PMCID: PMC4132022 DOI: 10.1371/journal.pone.0104759] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 07/12/2014] [Indexed: 12/17/2022] Open
Abstract
Objective Royal College of Surgeons (RCS) rats develop vasculopathy as photoreceptors degenerate. The aim of this study was to examine the effect of erythropoietin (EPO) on retinopathy in RCS rats. Methods Fluorescein angiography was used to monitor retinal vascular changes over time. Changes in retinal glia and vasculature were studied by immunostaining. To study the effects of EPO on retinal pathology, EPO (5000 IU/kg) was injected intraperitoneally in 14 week old normal and RCS rats twice a week for 4 weeks. Changes in the retinal vasculature, glia and microglia, photoreceptor apoptosis, differential expression of p75 neurotrophin receptor (p75NTR), pro-neurotrophin 3 (pro-NT3), tumour necrosis factor-α (TNFα), pigment epithelium derived factor (PEDF) and vascular endothelial growth factor-A (VEGF-A), the production of CD34+ cells and mobilization of CD34+/VEGF-R2+ cells as well as recruitment of CD34+ cells into the retina were examined after EPO treatment. Results RCS rats developed progressive capillary dropout and subretinal neovascularization which were accompanied by retinal gliosis. Systemic administration of EPO stabilized the retinal vasculature and inhibited the development of focal vascular lesions. Further studies showed that EPO modulated retinal gliosis, attenuated photoreceptor apoptosis and p75NTR and pro-NT3 upregulation, promoted the infiltration of ramified microglia and stimulated VEGF-A expression but had little effect on TNFα and PEDF expression. EPO stimulated the production of red and white blood cells and CD34+ cells along with effective mobilization of CD34+/VEGF-R2+ cells. Immunofluorescence study demonstrated that EPO enhanced the recruitment of CD34+ cells into the retina. Conclusions Our results suggest that EPO has therapeutic potentials in treatment of neuronal and vascular pathology in retinal disease. The protective effects of EPO on photoreceptors and the retinal vasculature may involve multiple mechanisms including regulation of retinal glia and microglia, inhibition of p75NTR-pro-NT3 signaling together with stimulation of production and mobilization of bone marrow derived cells.
Collapse
Affiliation(s)
- Weiyong Shen
- Save Sight Institute, the University of Sydney, Sydney, Australia
| | - Sook H Chung
- Save Sight Institute, the University of Sydney, Sydney, Australia
| | | | - Shiying Li
- Save Sight Institute, the University of Sydney, Sydney, Australia; Southwest Hospital/Southwest Eye Hospital, Third Military Medical University, Chongqing, China
| | - So-Ra Lee
- Save Sight Institute, the University of Sydney, Sydney, Australia
| | - Mark C Gillies
- Save Sight Institute, the University of Sydney, Sydney, Australia
| |
Collapse
|
27
|
Miloudi K, Dejda A, Binet F, Lapalme E, Cerani A, Sapieha P. Assessment of vascular regeneration in the CNS using the mouse retina. J Vis Exp 2014:e51351. [PMID: 24998265 DOI: 10.3791/51351] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The rodent retina is perhaps the most accessible mammalian system in which to investigate neurovascular interplay within the central nervous system (CNS). It is increasingly being recognized that several neurodegenerative diseases such as Alzheimer's, multiple sclerosis, and amyotrophic lateral sclerosis present elements of vascular compromise. In addition, the most prominent causes of blindness in pediatric and working age populations (retinopathy of prematurity and diabetic retinopathy, respectively) are characterized by vascular degeneration and failure of physiological vascular regrowth. The aim of this technical paper is to provide a detailed protocol to study CNS vascular regeneration in the retina. The method can be employed to elucidate molecular mechanisms that lead to failure of vascular growth after ischemic injury. In addition, potential therapeutic modalities to accelerate and restore healthy vascular plexuses can be explored. Findings obtained using the described approach may provide therapeutic avenues for ischemic retinopathies such as that of diabetes or prematurity and possibly benefit other vascular disorders of the CNS.
Collapse
Affiliation(s)
| | - Agnieszka Dejda
- Department of Biochemistry, Maisonneuve-Rosemont Hospital Research Centre, University of Montréal
| | - François Binet
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, University of Montréal
| | - Eric Lapalme
- Department of Biochemistry, Maisonneuve-Rosemont Hospital Research Centre, University of Montréal
| | - Agustin Cerani
- Department of Biochemistry, Maisonneuve-Rosemont Hospital Research Centre, University of Montréal
| | - Przemyslaw Sapieha
- Department of Neurology-Neurosurgery, McGill University; Department of Biochemistry, Maisonneuve-Rosemont Hospital Research Centre, University of Montréal; Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, University of Montréal;
| |
Collapse
|
28
|
Zhou JJ, Wang F, Xu Z, Lo WS, Lau CF, Chiang KP, Nangle LA, Ashlock MA, Mendlein JD, Yang XL, Zhang M, Schimmel P. Secreted histidyl-tRNA synthetase splice variants elaborate major epitopes for autoantibodies in inflammatory myositis. J Biol Chem 2014; 289:19269-75. [PMID: 24898250 DOI: 10.1074/jbc.c114.571026] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Inflammatory and debilitating myositis and interstitial lung disease are commonly associated with autoantibodies (anti-Jo-1 antibodies) to cytoplasmic histidyl-tRNA synthetase (HisRS). Anti-Jo-1 antibodies from different disease-afflicted patients react mostly with spatially separated epitopes in the three-dimensional structure of human HisRS. We noted that two HisRS splice variants (SVs) include these spatially separated regions, but each SV lacks the HisRS catalytic domain. Despite the large deletions, the two SVs cross-react with a substantial population of anti-Jo-l antibodies from myositis patients. Moreover, expression of at least one of the SVs is up-regulated in dermatomyositis patients, and cell-based experiments show that both SVs and HisRS can be secreted. We suggest that, in patients with inflammatory myositis, anti-Jo-1 antibodies may have extracellular activity.
Collapse
Affiliation(s)
- Jie J Zhou
- From the IAS HKUST-Scripps R&D Laboratory, Institute for Advanced Study, and Pangu BioPharma, Hong Kong, China
| | - Feng Wang
- From the IAS HKUST-Scripps R&D Laboratory, Institute for Advanced Study, and Pangu BioPharma, Hong Kong, China
| | - Zhiwen Xu
- From the IAS HKUST-Scripps R&D Laboratory, Institute for Advanced Study, and Pangu BioPharma, Hong Kong, China
| | - Wing-Sze Lo
- From the IAS HKUST-Scripps R&D Laboratory, Institute for Advanced Study, and Pangu BioPharma, Hong Kong, China
| | - Ching-Fun Lau
- From the IAS HKUST-Scripps R&D Laboratory, Institute for Advanced Study, and Pangu BioPharma, Hong Kong, China
| | | | | | | | | | - Xiang-Lei Yang
- From the IAS HKUST-Scripps R&D Laboratory, Institute for Advanced Study, and The Scripps Research Institute, La Jolla, California 92037, and
| | - Mingjie Zhang
- From the IAS HKUST-Scripps R&D Laboratory, Institute for Advanced Study, and Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Paul Schimmel
- From the IAS HKUST-Scripps R&D Laboratory, Institute for Advanced Study, and The Scripps Research Institute, La Jolla, California 92037, and Scripps Florida, Jupiter, Florida 33458
| |
Collapse
|
29
|
Tian LL, Ren B, Gao XW, Luo Y, Cai Y, Zhou K, Du AJ, Zhao Y. Inhibition of retinopathy of prematurity in rat by intravitreal injection of sorafenib. Int J Ophthalmol 2014; 7:198-204. [PMID: 24790858 DOI: 10.3980/j.issn.2222-3959.2014.02.03] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 12/11/2013] [Indexed: 12/23/2022] Open
Abstract
AIM To investigate the effect of intravitreal injection administered sorafenib, a multikinase inhibitor, in a rat model of oxygen-induced retinopathy (OIR). METHODS Seven-day-old Sprague-Dawley rats (n=144) were randomly assigned to six groups. Group A received normal partial oxygen pressure and groups B, C, D, E and F were exposed to hyperoxia (75±2)% from postnatal 7d (P7) to P12 to induce retinopathy of prematurity. The rats in groups C, D, E and F were received intravitreal injections of either vehicle (DMSO) or sorafenib at P12 (5, 20 and 80 µg, respectively). Then they returned to normoxia after P12. The retinas were whole-mounted and imaged with a confocal microscopy. The vascular branching points were counted to quantify neovascularization at P17. Cross-sections of the retina were stained with hematoxylin and eosin (HE). The nuclei of new vessels breaking the internal limiting membrane were counted to quantify the proliferative neovascular response. RESULTS The retinal vessel in groups B and C turned into tortuosity and a great deal of neovascularization were observed. Sorafenib-treated rats had significantly less neovascularization as compared with vehicle-treated and control rats in a dose dependent manner (P<0.05). The number of vascular branching points in A, B, C, D, E and F were 16.50±3.90, 37.44±6.47, 37.08±5.10, 30.80±6.85, 26.08±5.08 and 19.83±3.51, respectively. The number of the nuclei of retinal new vessel in A, B, C, D, E and F were 0.22±0.42, 35.66±4.70, 35.30±4.54, 27.30±4.28, 21.41±3.53, and 7.41±2.87, respectively. There were significant difference between each group (P<0.05) except groups B and C. CONCLUSION In the rat OIR model, sorafenib could inhibit retinal neovascularization in a dose dependent manner.
Collapse
Affiliation(s)
- Li-Li Tian
- Medical College of Shihezi University, Shihezi 832000, Xinjiang Uygur Autonomous Region, China ; Ophthalmic Center, No.474 Hospital of Chinese PLA, Urumqi 830013, Xinjiang Uygur Autonomous Region, China
| | - Bing Ren
- Ophthalmic Center, No.474 Hospital of Chinese PLA, Urumqi 830013, Xinjiang Uygur Autonomous Region, China
| | - Xiao-Wei Gao
- Ophthalmic Center, No.474 Hospital of Chinese PLA, Urumqi 830013, Xinjiang Uygur Autonomous Region, China
| | - Ying Luo
- Ophthalmic Center, No.474 Hospital of Chinese PLA, Urumqi 830013, Xinjiang Uygur Autonomous Region, China
| | - Yan Cai
- Ophthalmic Center, No.474 Hospital of Chinese PLA, Urumqi 830013, Xinjiang Uygur Autonomous Region, China
| | - Kun Zhou
- Medical College of Shihezi University, Shihezi 832000, Xinjiang Uygur Autonomous Region, China ; Ophthalmic Center, No.474 Hospital of Chinese PLA, Urumqi 830013, Xinjiang Uygur Autonomous Region, China
| | - An-Jie Du
- Department of Ophthalmology, Yuncheng Central Hospital, Yuncheng 044000, Shanxi Province, China
| | - Yong Zhao
- Ophthalmic Center, No.474 Hospital of Chinese PLA, Urumqi 830013, Xinjiang Uygur Autonomous Region, China
| |
Collapse
|
30
|
Zhang L, Jiang J, Xia X. Suppression of retinal neovascularization by small interfering RNA targeting PGC-1α. Int J Mol Med 2014; 33:1523-30. [PMID: 24682380 DOI: 10.3892/ijmm.2014.1717] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Accepted: 03/13/2014] [Indexed: 11/05/2022] Open
Abstract
Peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α) is a key coordinator of gene programs in metabolism and energy homeostasis in mammals. The aim of this study was to determine whether PGC-1α is involved in the transcriptional regulation of retinal neovascularization in oxygen-induced retinopathy (OIR). The expression of PGC-1α in the retina of mice with OIR was detected by real-time polymerase chain reaction (PCR) and western blot analysis. Mice with OIR were administered small interfering RNA (siRNA) targeting PGC-1α by intravitreal injection, and the effects of PGC-1α siRNA were confirmed by fluorescein angiography and quantification of pre-retinal neovascular nuclei in the retinal sections. PGC-1α was upregulated at both the mRNA and protein level under hypoxic conditions. Retinal neovascularization was inhibited by PGC-1α siRNA. Furthermore, PGC-1α mRNA and protein levels were also reduced by PGC-1α siRNA, which were detected by real-time PCR and western blot analysis. The downregulation of PGC-1α expression resulted in the reduction of vascular endothelial growth factor (VEGF) expression in the mice. In conclusion, siRNA targeting PGC-1α inhibits retinal neovascularization by downregulating the expression of PGC-1α and VEGF in the murine retina. Therefore, PGC-1α represents a potential therapeutic target for ischemia-induced retinal diseases and other ocular neovascular diseases.
Collapse
Affiliation(s)
- Lixin Zhang
- Department of Ophthalmology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Jian Jiang
- Department of Ophthalmology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Xiaobo Xia
- Department of Ophthalmology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| |
Collapse
|
31
|
Westenskow PD, Kurihara T, Aguilar E, Scheppke EL, Moreno SK, Wittgrove C, Marchetti V, Michael IP, Anand S, Nagy A, Cheresh D, Friedlander M. Ras pathway inhibition prevents neovascularization by repressing endothelial cell sprouting. J Clin Invest 2014; 123:4900-8. [PMID: 24084735 DOI: 10.1172/jci70230] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Accepted: 08/01/2013] [Indexed: 12/27/2022] Open
Abstract
Vascular networks develop from a growing vascular front that responds to VEGF and other guidance cues. Angiogenesis is required for normal tissue function, but, under conditions of stress, inappropriate vascularization can lead to disease. Therefore, inhibition of angiogenic sprouting may prevent neovascularization in patients with blinding neovascular eye diseases, including macular degeneration. VEGF antagonists have therapeutic benefits but also can elicit off-target effects. Here, we found that the Ras pathway, which functions downstream of a wide range of cytokines including VEGF, is active in the growing vascular front of developing and pathological vascular networks. The endogenous Ras inhibitor p120RasGAP was expressed predominately in quiescent VEGF-insensitive endothelial cells and was ectopically downregulated in multiple neovascular models. MicroRNA-132 negatively regulated p120RasGAP expression. Experimental delivery of α-miR-132 to developing mouse eyes disrupted tip cell Ras activity and prevented angiogenic sprouting. This strategy prevented ocular neovascularization in multiple rodent models even more potently than the VEGF antagonist, VEGF-trap. Targeting microRNA-132 as a therapeutic strategy may prove useful for treating multiple neovascular diseases of the eye and for preventing vision loss regardless of the neovascular stimulus.
Collapse
|
32
|
Liu X, Zhang B, McBride JD, Zhou K, Lee K, Zhou Y, Liu Z, Ma JX. Antiangiogenic and antineuroinflammatory effects of kallistatin through interactions with the canonical Wnt pathway. Diabetes 2013; 62:4228-38. [PMID: 23884893 PMCID: PMC3837048 DOI: 10.2337/db12-1710] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Kallistatin is a member of the serine proteinase inhibitor superfamily. Kallistatin levels have been shown to be decreased in the vitreous while increased in the circulation of patients with diabetic retinopathy (DR). Overactivation of the Wnt pathway is known to play pathogenic roles in DR. To investigate the role of kallistatin in DR and in Wnt pathway activation, we generated kallistatin transgenic (kallistatin-TG) mice overexpressing kallistatin in multiple tissues including the retina. In the oxygen-induced retinopathy (OIR) model, kallistatin overexpression attenuated ischemia-induced retinal neovascularization. In diabetic kallistatin-TG mice, kallistatin overexpression ameliorated retinal vascular leakage, leukostasis, and overexpression of vascular endothelial growth factor and intracellular adhesion molecule. Furthermore, kallistatin overexpression also suppressed Wnt pathway activation in the retinas of the OIR and diabetic models. In diabetic Wnt reporter (BAT-gal) mice, kallistatin overexpression suppressed retinal Wnt reporter activity. In cultured retinal cells, kallistatin blocked Wnt pathway activation induced by high glucose and by Wnt ligand. Coprecipitation and ligand-binding assays both showed that kallistatin binds to a Wnt coreceptor LRP6 with high affinity (Kd = 4.5 nmol/L). These observations suggest that kallistatin is an endogenous antagonist of LRP6 and inhibitor of Wnt signaling. The blockade of Wnt signaling may represent a mechanism for its antiangiogenic and antineuroinflammatory effects.
Collapse
Affiliation(s)
- Xiaochen Liu
- Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Xiamen, China
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Bin Zhang
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Jeffrey D. McBride
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Kevin Zhou
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Kyungwon Lee
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Yueping Zhou
- Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Xiamen, China
| | - Zuguo Liu
- Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Xiamen, China
- Corresponding authors: Zuguo Liu, , and Jian-xing Ma,
| | - Jian-xing Ma
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
- Corresponding authors: Zuguo Liu, , and Jian-xing Ma,
| |
Collapse
|
33
|
Neuronal sirtuin1 mediates retinal vascular regeneration in oxygen-induced ischemic retinopathy. Angiogenesis 2013; 16:985-92. [PMID: 23912262 DOI: 10.1007/s10456-013-9374-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Accepted: 07/24/2013] [Indexed: 10/26/2022]
Abstract
Regeneration of blood vessels in ischemic neuronal tissue is critical to reduce tissue damage in diseases. In proliferative retinopathy, initial vessel loss leads to retinal ischemia, which can induce either regrowth of vessels to restore normal metabolism and minimize damage, or progress to hypoxia-induced sight-threatening pathologic vaso-proliferation. It is not well understood how retinal neurons mediate regeneration of vascular growth in response to ischemic insults. In this study we aim to investigate the potential role of Sirtuin 1 (Sirt1), a metabolically-regulated protein deacetylase, in mediating the response of ischemic neurons to regulate vascular regrowth in a mouse model of oxygen-induced ischemic retinopathy (OIR). We found that Sirt1 is highly induced in the avascular ischemic retina in OIR. Conditional depletion of neuronal Sirt1 leads to significantly decreased retinal vascular regeneration into the avascular zone and increased hypoxia-induced pathologic vascular growth. This effect is likely independent of PGC-1α, a known Sirt1 target, as absence of PGC-1α in knockout mice does not impact vascular growth in retinopathy. We found that neuronal Sirt1 controls vascular regrowth in part through modulating deacetylation and stability of hypoxia-induced factor 1α and 2α, and thereby modulating expression of angiogenic factors. These results indicate that ischemic neurons induce Sirt1 to promote revascularization into ischemic neuronal areas, suggesting a novel role of neuronal Sirt1 in mediating vascular regeneration in ischemic conditions, with potential implications beyond retinopathy.
Collapse
|
34
|
Guo M, Schimmel P. Essential nontranslational functions of tRNA synthetases. Nat Chem Biol 2013; 9:145-53. [PMID: 23416400 DOI: 10.1038/nchembio.1158] [Citation(s) in RCA: 289] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Accepted: 11/28/2012] [Indexed: 12/25/2022]
Abstract
Nontranslational functions of vertebrate aminoacyl tRNA synthetases (aaRSs), which catalyze the production of aminoacyl-tRNAs for protein synthesis, have recently been discovered. Although these new functions were thought to be 'moonlighting activities', many are as critical for cellular homeostasis as their activity in translation. New roles have been associated with their cytoplasmic forms as well as with nuclear and secreted extracellular forms that affect pathways for cardiovascular development and the immune response and mTOR, IFN-γ and p53 signaling. The associations of aaRSs with autoimmune disorders, cancers and neurological disorders further highlight nontranslational functions of these proteins. New architecture elaborations of the aaRSs accompany their functional expansion in higher organisms and have been associated with the nontranslational functions for several aaRSs. Although a general understanding of how these functions developed is limited, the expropriation of aaRSs for essential nontranslational functions may have been initiated by co-opting the amino acid-binding site for another purpose.
Collapse
Affiliation(s)
- Min Guo
- Department of Cancer Biology, The Scripps Research Institute, Jupiter, Florida, USA
| | | |
Collapse
|
35
|
Kasai A, Ishimaru Y, Higashino K, Kobayashi K, Yamamuro A, Yoshioka Y, Maeda S. Inhibition of apelin expression switches endothelial cells from proliferative to mature state in pathological retinal angiogenesis. Angiogenesis 2013; 16:723-34. [PMID: 23640575 PMCID: PMC3682100 DOI: 10.1007/s10456-013-9349-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Accepted: 04/24/2013] [Indexed: 11/25/2022]
Abstract
The recruitment of mural cells such as pericytes to patent vessels with an endothelial lumen is a key factor for the maturation of blood vessels and the prevention of hemorrhage in pathological angiogenesis. To date, our understanding of the specific trigger underlying the transition from cell growth to the maturation phase remains incomplete. Since rapid endothelial cell growth causes pericyte loss, we hypothesized that suppression of endothelial growth factors would both promote pericyte recruitment, in addition to inhibiting pathological angiogenesis. Here, we demonstrate that targeted knockdown of apelin in endothelial cells using siRNA induced the expression of monocyte chemoattractant protein-1 (MCP-1) through activation of Smad3, via suppression of the PI3K/Akt pathway. The conditioned medium of endothelial cells treated with apelin siRNA enhanced the migration of vascular smooth muscle cells, through MCP-1 and its receptor pathway. Moreover, in vivo delivery of siRNA targeting apelin, which causes exuberant endothelial cell proliferation and pathological angiogenesis through its receptor APJ, led to increased pericyte coverage and suppressed pathological angiogenesis in an oxygen-induced retinopathy model. These data demonstrate that apelin is not only a potent endothelial growth factor, but also restricts pericyte recruitment, establishing a new connection between endothelial cell proliferation signaling and a trigger of mural recruitment.
Collapse
Affiliation(s)
- Atsushi Kasai
- Interdisciplinary Program for Biomedical Sciences, Institute for Academic Initiatives, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan.
| | | | | | | | | | | | | |
Collapse
|
36
|
He C, Sun Y, Ren X, Lin Q, Hu X, Huang X, Su SB, Liu Y, Liu X. Angiogenesis Mediated by Toll-Like Receptor 4 in Ischemic Neural Tissue. Arterioscler Thromb Vasc Biol 2013; 33:330-8. [DOI: 10.1161/atvbaha.112.300679] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Chang He
- From the State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center (C.H., Y.S., X.R., Q.L., X. Hu., S. -B.S., Y.L., X.L.) and Department of Immunology, Zhongshan School of Medicine, Institute of Human Virology (X. Huang), Sun Yat-sen University, Guangzhou, China
| | - Yuying Sun
- From the State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center (C.H., Y.S., X.R., Q.L., X. Hu., S. -B.S., Y.L., X.L.) and Department of Immunology, Zhongshan School of Medicine, Institute of Human Virology (X. Huang), Sun Yat-sen University, Guangzhou, China
| | - Xiangrong Ren
- From the State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center (C.H., Y.S., X.R., Q.L., X. Hu., S. -B.S., Y.L., X.L.) and Department of Immunology, Zhongshan School of Medicine, Institute of Human Virology (X. Huang), Sun Yat-sen University, Guangzhou, China
| | - Qing Lin
- From the State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center (C.H., Y.S., X.R., Q.L., X. Hu., S. -B.S., Y.L., X.L.) and Department of Immunology, Zhongshan School of Medicine, Institute of Human Virology (X. Huang), Sun Yat-sen University, Guangzhou, China
| | - Xiao Hu
- From the State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center (C.H., Y.S., X.R., Q.L., X. Hu., S. -B.S., Y.L., X.L.) and Department of Immunology, Zhongshan School of Medicine, Institute of Human Virology (X. Huang), Sun Yat-sen University, Guangzhou, China
| | - Xi Huang
- From the State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center (C.H., Y.S., X.R., Q.L., X. Hu., S. -B.S., Y.L., X.L.) and Department of Immunology, Zhongshan School of Medicine, Institute of Human Virology (X. Huang), Sun Yat-sen University, Guangzhou, China
| | - Shao-Bo Su
- From the State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center (C.H., Y.S., X.R., Q.L., X. Hu., S. -B.S., Y.L., X.L.) and Department of Immunology, Zhongshan School of Medicine, Institute of Human Virology (X. Huang), Sun Yat-sen University, Guangzhou, China
| | - Yizhi Liu
- From the State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center (C.H., Y.S., X.R., Q.L., X. Hu., S. -B.S., Y.L., X.L.) and Department of Immunology, Zhongshan School of Medicine, Institute of Human Virology (X. Huang), Sun Yat-sen University, Guangzhou, China
| | - Xialin Liu
- From the State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center (C.H., Y.S., X.R., Q.L., X. Hu., S. -B.S., Y.L., X.L.) and Department of Immunology, Zhongshan School of Medicine, Institute of Human Virology (X. Huang), Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
37
|
Nakamura S, Tsuruma K, Shimazawa M, Hara H. Candesartan, an angiotensin II type 1 receptor antagonist, inhibits pathological retinal neovascularization by downregulating VEGF receptor-2 expression. Eur J Pharmacol 2012; 685:8-14. [PMID: 22543084 DOI: 10.1016/j.ejphar.2012.04.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2011] [Revised: 03/30/2012] [Accepted: 04/05/2012] [Indexed: 01/23/2023]
Abstract
Several studies have examined the anti-angiogenic effects of angiotensin II type 1 (AT(1)) receptor antagonists; however, the mechanisms underlying these effects are currently unclear. In the present study, we examined the efficacy and the mechanism of candesartan, an AT(1) receptor antagonist, in suppressing pathological retinal neovascularization. We used an in vivo murine oxygen-induced retinopathy (OIR) model and also studied the in vitro proliferation and migration of human retinal microvascular endothelial cells (HRMECs) induced by vascular endothelial growth factor (VEGF)-A. The regulation of angiogenesis-associated genes such as hypoxia-inducible factor (HIF-1α), VEGF-A, VEGF receptor-1, and VEGF receptor-2 was evaluated with real-time RT-PCR in the OIR model. In the OIR model, candesartan suppressed the pathological neovascularization in a dose-dependent manner, but did not prevent the physiological angiogenesis. However, candesartan did not inhibit VEGF-A-induced proliferation or migration in HRMECs in the in vitro study. When administered interperitoneally in the OIR model, candesartan reduced the upregulation of VEGF receptor-2 in the retina, but had no effects in the other angiogenesis-related genes, such as HIF-1α, VEGF-A, and VEGF receptor-1. These findings indicate that candesartan inhibited the retinal pathological neovascularization, at least in part, by suppressing the expression of VEGF receptor-2, independent of VEGF signaling cascade. Therefore, candesartan may be a useful therapeutic target for the inhibition of retinal neovascularization that has a low risk of serious side effects.
Collapse
Affiliation(s)
- Shinsuke Nakamura
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | | | | | | |
Collapse
|
38
|
Shang B, Cao Z, Zhou Q. Progress in tumor vascular normalization for anticancer therapy: challenges and perspectives. Front Med 2012; 6:67-78. [PMID: 22460450 DOI: 10.1007/s11684-012-0176-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2011] [Accepted: 11/16/2011] [Indexed: 02/07/2023]
|
39
|
Mowat FM, Gonzalez F, Luhmann UFO, Lange CA, Duran Y, Smith AJ, Maxwell PH, Ali RR, Bainbridge JWB. Endogenous erythropoietin protects neuroretinal function in ischemic retinopathy. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 180:1726-39. [PMID: 22342523 DOI: 10.1016/j.ajpath.2011.12.033] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2011] [Revised: 11/14/2011] [Accepted: 12/08/2011] [Indexed: 01/12/2023]
Abstract
Because retinal ischemia is a common cause of vision loss, we sought to determine the effects of ischemia on neuroretinal function and survival in murine oxygen-induced retinopathy (OIR) and to define the role of endogenous erythropoietin (EPO) in this model. OIR is a reproducible model of ischemia-induced retinal neovascularization; it is used commonly to develop antiangiogenic strategies. We investigated the effects of ischemia in murine OIR on retinal function and neurodegeneration by electroretinography and detailed morphology. OIR was associated with significant neuroretinal dysfunction, with reduced photopic and scotopic ERG responses and reduced b-wave/a-wave ratios consistent with specific inner-retinal dysfunction. OIR resulted in significantly increased apoptosis and atrophy of the inner retina in areas of ischemia. EPO deficiency in heterozygous Epo-Tag transgenic mice was associated with more profound retinal dysfunction after OIR, indicated by a significantly greater suppression of ERG amplitudes, but had no measurable effect on the extent of retinal ischemia, preretinal neovascularization, or neuroretinal degeneration in OIR. Systemic administration of recombinant EPO protected EPO-deficient mice against this additional suppression, but EPO supplementation in wild-type animals with OIR did not rescue neuroretinal dysfunction or degeneration. Murine OIR offers a valuable model of ischemic neuroretinal dysfunction and degeneration in which to investigate adaptive tissue responses and evaluate novel therapeutic approaches. Endogenous EPO can protect neuroretinal function in ischemic retinopathy.
Collapse
Affiliation(s)
- Freya M Mowat
- Department of Genetics, University College London Institute of Ophthalmology, London, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Nakamura S, Imai S, Ogishima H, Tsuruma K, Shimazawa M, Hara H. Morphological and functional changes in the retina after chronic oxygen-induced retinopathy. PLoS One 2012; 7:e32167. [PMID: 22348151 PMCID: PMC3279421 DOI: 10.1371/journal.pone.0032167] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2011] [Accepted: 01/20/2012] [Indexed: 11/23/2022] Open
Abstract
The mouse model of oxygen-induced retinopathy (OIR) has been widely used for studies of retinopathy of prematurity (ROP). This disorder, characterized by abnormal vascularization of the retina, tends to occur in low birth weight neonates after exposure to high supplemental oxygen. Currently, the incidence of ROP is increasing because of increased survival of these infants due to medical progress. However, little is known about changes in the chronic phase after ROP. Therefore, in this study, we examined morphological and functional changes in the retina using a chronic OIR model. Both the a- and b-waves in the OIR model recovered in a time-dependent manner at 4 weeks (w), 6 w, and 8 w, but the oscillatory potential (OP) amplitudes remained depressed following a return to normoxic conditions. Furthermore, decrease in the thicknesses of the inner plexiform layer (IPL) and inner nuclear layer (INL) at postnatal day (P) 17, 4 w, and 8 w and hyperpermeability of blood vessels were observed in conjunction with the decrease in the expression of claudin-5 and occludin at 8 w. The chronic OIR model revealed the following: (1) a decrease in OP amplitudes, (2) morphological abnormalities in the retinal cells (limited to the IPL and INL) and blood vessels, and (3) an increase in retinal vascular permeability via the impairment of the tight junction proteins. These findings suggest that the experimental animal model used in this study is suitable for elucidating the pathogenesis of ROP and may lead to the development of potential therapeutic agents for ROP treatment.
Collapse
Affiliation(s)
- Shinsuke Nakamura
- Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | | | | | | | | | | |
Collapse
|
41
|
Yanai R, Thanos A, Connor KM. Complement involvement in neovascular ocular diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 946:161-83. [PMID: 21948368 DOI: 10.1007/978-1-4614-0106-3_10] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Pathological neovascularization (NV) is a hallmark of late stage neovascular age-related macular degeneration (AMD), diabetic retinopathy (DR), and retinopathy of prematurity (ROP). There is accumulating evidence that alterations in inflammatory and immune system pathways that arise from genetic differences, injury, and disease can predispose individuals to retinal neovascular eye diseases. Yet the mechanism of disease progression with respect to the complement system in these maladies is not fully understood. Recent studies have implicated the complement system as an emerging player in the etiology of several retinal diseases. We will summarize herein several of the complement system pathways known to be involved in ocular neovascular pathologies. Current treatment for many neovascular eye diseases focuses on suppression of NV with laser ablation, photodynamic therapy, or anti-VEGF angiogenic inhibitors. However, these treatments do not address the underlying cause of many of these diseases. A clear understanding of the cellular and molecular mechanisms could bring a major shift in our approach to disease treatment and prevention.
Collapse
Affiliation(s)
- Ryoji Yanai
- Angiogenesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 02114, USA.
| | | | | |
Collapse
|
42
|
Martini D, Monte MD, Ristori C, Cupisti E, Mei S, Fiorini P, Filippi L, Bagnoli P. Antiangiogenic effects of β2 -adrenergic receptor blockade in a mouse model of oxygen-induced retinopathy. J Neurochem 2011; 119:1317-29. [PMID: 21988318 DOI: 10.1111/j.1471-4159.2011.07530.x] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Oxygen-induced retinopathy (OIR) is a model for human retinopathy of prematurity. In mice with OIR, beta-adrenergic receptor (β-AR) blockade with propranolol has been shown to ameliorate different aspects of retinal dysfunction in response to hypoxia. In the present study, we used the OIR model to investigate the role of distinct β-ARs on retinal proangiogenic factors, pathogenic neovascularization and electroretinographic responses. Our results demonstrate that β(2) -AR blockade with ICI 118,551 decreases retinal levels of proangiogenic factors and reduces pathogenic neovascularization, whereas β(1) - and β(3) -AR antagonists do not. Determination of retinal protein kinase A activity is indicative of the fact that β-AR blockers are indeed effective at the receptor level. In addition, the specificity of ICI 118,551 on retinal angiogenesis has been demonstrated by the finding that in mouse retinal explants, β(2) -AR silencing prevents ICI 118,551 effects on hypoxia-induced vascular endothelial growth factor accumulation. In OIR mice, ICI 118,551 is effective in increasing electroretinographic responses suggesting that activation of β(2) -ARs constitutes an important part of the retinal response to hypoxia. Lastly, immunohistochemical studies demonstrate that β(2) -ARs are localized to several retinal cells, particularly to Müller cells suggesting the possibility that β(2) -ARs play a role in regulating vascular endothelial growth factor production by these cells. The present results suggest that pathogenic angiogenesis, a key change in many hypoxic/ischemic vision-threatening retinal diseases, depends at least in part on β(2) -AR activity and indicate that β(2) -AR blockade can be effective against retinal angiogenesis.
Collapse
Affiliation(s)
- Davide Martini
- Department of Biology, University of Pisa, Via San Zeno, Pisa, Italy
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Nakagawa Y, Masuda H, Ito R, Kobori M, Wada M, Shizuno T, Sato A, Suzuki T, Kawai K, Asahara T. Aberrant kinetics of bone marrow-derived endothelial progenitor cells in the murine oxygen-induced retinopathy model. Invest Ophthalmol Vis Sci 2011; 52:7835-41. [PMID: 21896844 DOI: 10.1167/iovs.10-5880] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
PURPOSE Retinopathy of prematurity (ROP) causes serious blindness because of the vasculopathy that results from the abnormal oxygen dynamics. However, the systemic kinetics of bone marrow-derived endothelial progenitor cells (BM-derived EPCs) during the "postnatal vasculogenesis " of ROP has yet to be elucidated. Thus, the authors investigated the kinetics of BM-derived EPCs using a murine oxygen-induced retinopathy (OIR) model. METHODS OIR was induced in C57BL/6J mice by continual aeration with 75% oxygen from postnatal day (P) 7 to P12 that afterward returned to normal room air. RESULTS The frequency of circulating EPCs (Sca-1(+)/c-Kit(+) cells in blood) in an OIR model estimated by FACS decreased immediately after the hyperoxic phase (P12) and then increased at the hypoxic phase (P17) compared with control. Further, EPC colony-forming assay of BM-Lin(-)/Sca-1(+) (BM-LS) cells exhibited a conversion from the predominant primitive EPC colony production at P12 to the definitive EPC colony at P17. In the OIR retinas of BM-transplanted mice with BM-LS cells of EGFP transgenic mice, there was less incorporation of GFP(+) cells into vascular structures at P12, whereas there was a drastic recruitment into the "tufts " and for the intact vasculature at P17. Moreover, the definitive EPC colony cells intravitreally injected into OIR significantly abrogated pathologic versus primitive vascular growth. CONCLUSIONS Taken together, these findings propose that the deviation of functional bioactivities of BM-derived EPCs contributing to intact vascular development under the abnormal oxygen dynamics may provide important mechanistic insight into pathologic vascular development in ROP.
Collapse
Affiliation(s)
- Yoshihiro Nakagawa
- Departments of Regenerative Medicine Science, Tokai University School of Medicine, Isehara, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Differential macrophage polarization promotes tissue remodeling and repair in a model of ischemic retinopathy. Sci Rep 2011; 1:76. [PMID: 22355595 PMCID: PMC3216563 DOI: 10.1038/srep00076] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2011] [Accepted: 08/08/2011] [Indexed: 11/24/2022] Open
Abstract
Diabetic retinopathy is the leading cause of visual loss in individuals under the age of 55. Umbilical cord blood (UCB)–derived myeloid progenitor cells have been shown to decrease neuronal damage associated with ischemia in the central nervous system. In this study we show that UCB-derived CD14+ progenitor cells provide rescue effects in a mouse model of ischemic retinopathy by promoting physiological angiogenesis and reducing associated inflammation. We use confocal microscopy to trace the fate of injected human UCB-derived CD14+ cells and PCR with species-specific probes to investigate their gene expression profile before and after injection. Metabolomic analysis measures changes induced by CD14+ cells. Our results demonstrate that human cells differentiate in vivo into M2 macrophages and induce the polarization of resident M2 macrophages. This leads to stabilization of the ischemia-injured retinal vasculature by modulating the inflammatory response, reducing oxidative stress and apoptosis and promoting tissue repair.
Collapse
|
45
|
Zhang W, Yokota H, Xu Z, Narayanan SP, Yancey L, Yoshida A, Marcus DM, Caldwell RW, Caldwell RB, Brooks SE. Hyperoxia therapy of pre-proliferative ischemic retinopathy in a mouse model. Invest Ophthalmol Vis Sci 2011; 52:6384-95. [PMID: 21705685 DOI: 10.1167/iovs.11-7666] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
PURPOSE To investigate the therapeutic use and mechanisms of action of normobaric hyperoxia to promote revascularization and to prevent neovascularization in a mouse model of oxygen-induced ischemic retinopathy. METHODS Hyperoxia treatment (HT, 40%-75% oxygen) was initiated on postnatal day (P) 14 during the pre-proliferative phase of ischemic retinopathy. Immunohistochemistry, ELISA, and quantitative PCR were used to assess effects on retinal vascular repair and pathologic angiogenesis in relation to glial cell injury, VEGF protein, and mRNA levels of key mediators of pathologic angiogenesis. Effects of intravitreal injections of VEGF and the VEGF inhibitor VEGFR1/Fc fusion protein were also studied. RESULTS Administration of HT during the ischemic pre-proliferative phase of retinopathy effectively accelerated the process of revascularization while preventing the development of vitreous neovascularization. HT enhanced the formation of specialized endothelial tip cells at the edges of the repairing capillary networks and blocked the overexpression of several molecular mediators of angiogenesis, inflammation, and extracellular proteolysis. HT markedly reduced the reactive expression of GFAP in Müller cells and improved the morphology of astrocytes in the avascular region of the retina. Exogenous VEGF administered into the vitreous on P14 was not sufficient to cause vitreous neovascularization in the HT mice. Injection of the VEGF antagonist VEGFR1/Fc blocked both pathologic and physiological angiogenesis and did not rescue astrocytes. CONCLUSIONS HT may be clinically useful to facilitate vascular repair while blocking neovascularization in the pre-proliferative stage of ischemic retinopathy by correcting a broad range of biochemical and cellular abnormalities.
Collapse
Affiliation(s)
- Wenbo Zhang
- Vascular Biology Center, Georgia Health Sciences University, Augusta, Georgia 30912-2500, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Zaniolo K, Sapieha P, Shao Z, Stahl A, Zhu T, Tremblay S, Picard E, Madaan A, Blais M, Lachapelle P, Mancini J, Hardy P, Smith LEH, Ong H, Chemtob S. Ghrelin modulates physiologic and pathologic retinal angiogenesis through GHSR-1a. Invest Ophthalmol Vis Sci 2011; 52:5376-86. [PMID: 21642627 DOI: 10.1167/iovs.10-7152] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE Vascular degeneration and the ensuing abnormal vascular proliferation are central to proliferative retinopathies. Given the metabolic discordance associated with these diseases, the authors explored the role of ghrelin and its growth hormone secretagogue receptor 1a (GHSR-1a) in proliferative retinopathy. METHODS In a rat model of oxygen-induced retinopathy (OIR), the contribution of ghrelin and GHSR-1a was investigated using the stable ghrelin analogs [Dap3]-ghrelin and GHRP6 and the GSHR-1a antagonists JMV-2959 and [D-Lys3]-GHRP-6. Plasma and retinal levels of ghrelin were analyzed by ELISA, whereas retinal expression and localization of GHSR-1a were examined by immunohistochemistry and Western blot analysis. The angiogenic and vasoprotective properties of ghrelin and its receptor were further confirmed in aortic explants and in models of vaso-obliteration. RESULTS Ghrelin is produced locally in the retina, whereas GHSR-1a is abundantly expressed in retinal endothelial cells. Ghrelin levels decrease during the vaso-obliterative phase and rise during the proliferative phase of OIR. Intravitreal delivery of [Dap3]-ghrelin during OIR significantly reduces retinal vessel loss when administered during the hyperoxic phase. Conversely, during the neovascular phase, ghrelin promotes pathologic angiogenesis through the activation of GHSR-1a. These angiogenic effects were confirmed ex vivo in aortic explants. CONCLUSIONS New roles were disclosed for the ghrelin-GHSR-1a pathway in the preservation of retinal vasculature during the vaso-obliterative phase of OIR and during the angiogenic phase of OIR. These findings suggest that the ghrelin-GHSR-1a pathway can exert opposing effects on retinal vasculature, depending on the phase of retinopathy, and thus holds therapeutic potential for proliferative retinopathies.
Collapse
Affiliation(s)
- Karine Zaniolo
- Department of Pediatrics, Hôpital Sainte-Justine Research Center, Montreal, Quebec, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Weidemann A, Krohne TU, Aguilar E, Kurihara T, Takeda N, Dorrell MI, Simon MC, Haase VH, Friedlander M, Johnson RS. Astrocyte hypoxic response is essential for pathological but not developmental angiogenesis of the retina. Glia 2010; 58:1177-85. [PMID: 20544853 DOI: 10.1002/glia.20997] [Citation(s) in RCA: 132] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Vascular/parenchymal crosstalk is increasingly recognized as important in the development and maintenance of healthy vascularized tissues. The retina is an excellent model in which to study the role of cell type-specific contributions to the process of blood vessel and neuronal growth. During retinal vascular development, glial cells such as astrocytes provide the template over which endothelial cells migrate to form the retinal vascular network, and hypoxia-regulated vascular endothelial growth factor (VEGF) has been demonstrated to play a critical role in this process as well as pathological neovascularization. To investigate the nature of cell-specific contributions to this process, we deleted VEGF and its upstream regulators, the hypoxia-inducible transcription factors HIF-1 alpha and HIF-2 alpha, and the negative regulator of HIF alpha, von Hippel-Lindau protein (VHL), in astrocytes. We found that loss of hypoxic response and VEGF production in astrocytes does not impair normal development of retinal vasculature, indicating that astrocyte-derived VEGF is not essential for this process. In contrast, using a model of oxygen-induced ischemic retinopathy, we show that astrocyte-derived VEGF is essential for hypoxia-induced neovascularization. Thus, we demonstrate that astrocytes in the retina have highly divergent roles during developmental, physiological angiogenesis, and ischemia-driven, pathological neovascularization.
Collapse
Affiliation(s)
- Alexander Weidemann
- Division of Biology, University of California, San Diego, La Jolla, California 92093, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Stahl A, Connor KM, Sapieha P, Chen J, Dennison RJ, Krah NM, Seaward MR, Willett KL, Aderman CM, Guerin KI, Hua J, Löfqvist C, Hellström A, Smith LEH. The mouse retina as an angiogenesis model. Invest Ophthalmol Vis Sci 2010; 51:2813-26. [PMID: 20484600 DOI: 10.1167/iovs.10-5176] [Citation(s) in RCA: 493] [Impact Index Per Article: 32.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The mouse retina has been used extensively over the past decades to study both physiologic and pathologic angiogenesis. Over time, various mouse retina models have evolved into well-characterized and robust tools for in vivo angiogenesis research. This article is a review of the angiogenic development of the mouse retina and a discussion of some of the most widely used vascular disease models. From the multitude of studies performed in the mouse retina, a selection of representative works is discussed in more detail regarding their role in advancing the understanding of both the ocular and general mechanisms of angiogenesis.
Collapse
Affiliation(s)
- Andreas Stahl
- Department of Ophthalmology, Harvard Medical School, Children's Hospital Boston, Boston, Massachusetts 02115, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Bai Y, Ma JX, Guo J, Wang J, Zhu M, Chen Y, Le YZ. Müller cell-derived VEGF is a significant contributor to retinal neovascularization. J Pathol 2010; 219:446-54. [PMID: 19768732 DOI: 10.1002/path.2611] [Citation(s) in RCA: 162] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Vascular endothelial growth factor (VEGF-A) is a major pathogenic factor and a therapeutic target for age-related macular degeneration, diabetic retinopathy, and retinopathy of prematurity. Despite intensive effort in the field, the cellular mechanisms of VEGF action remain virtually uninvestigated. This situation makes it difficult to design cellular target-based therapeutics for these diseases. In light of the recent finding that VEGF is a potential neurotrophic factor, revealing the cellular mechanisms of VEGF action becomes necessary to preserve its beneficial effect and inhibit its pathological function in long-term anti-VEGF therapeutics for ocular vascular diseases. We therefore generated conditional VEGF knockout mice with an inducible Cre/lox system and determined the significance of Müller cell-derived VEGF in retinal development and maintenance and ischaemia-induced neovascularizartion and vascular leakage. Retinal development in the conditional VEGF knockout mice was analysed by examining retinal and choroidal vasculatures and retinal morphology and function. Ischaemia-induced retinal neovascularization and vascular leakage in the conditional VEGF knockout mice were analysed with fluorescein angiography, quantification of proliferative neovascular cells, immunohistochemistry, and immunoblotting using an oxygen-induced retinopathy model. Our results demonstrated that disruption of Müller cell-derived VEGF resulted in no apparent defects in retinal and choroidal vasculatures and retinal morphology and function, significant inhibition of the ischaemia-induced retinal neovascularization and vascular leakage, and attenuation of the ischaemia-induced breakdown of the blood-retina barrier. These results suggest that the retinal Müller cell-derived VEGF is a major contributor to ischaemia-induced retinal vascular leakage and pre-retinal and intra-retinal neovascularization. The observation that a significant, but not complete, reduction of VEGF in the retina does not cause detectable retinal degeneration suggests that appropriate doses of anti-VEGF agents may be important to the safe treatment of retinal vascular diseases.
Collapse
Affiliation(s)
- Yanyan Bai
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | | | | | | | | | | | | |
Collapse
|
50
|
Dorrell MI, Aguilar E, Jacobson R, Trauger SA, Friedlander J, Siuzdak G, Friedlander M. Maintaining retinal astrocytes normalizes revascularization and prevents vascular pathology associated with oxygen-induced retinopathy. Glia 2010; 58:43-54. [PMID: 19544395 DOI: 10.1002/glia.20900] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Astrocytes are well known modulators of normal developmental retinal vascularization. However, relatively little is known about the role of glial cells during pathological retinal neovascularization (NV), a leading contributor to vision loss in industrialized nations. We demonstrate that the loss of astrocytes and microglia directly correlates with the development of pathological NV in a mouse model of oxygen-induced retinopathy (OIR). These two distinct glial cell populations were found to have cooperative survival effects in vitro and in vivo. The intravitreal injection of myeloid progenitor cells, astrocytes, or astrocyte-conditioned media rescued endogenous astrocytes from degeneration that normally occurs within the hypoxic, vaso-obliterated retina following return to normoxia. Protection of the retinal astrocytes and microglia was directly correlated with accelerated revascularization of the normal retinal plexuses and reduction of pathological intravitreal NV normally associated with OIR. Using astrocyte-conditioned media, several factors were identified that may contribute to the observed astrocytic protection and subsequent normalization of the retinal vasculature, including vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF). Injection of VEGF or bFGF at specific doses rescued the retinas from developing OIR-associated pathology, an effect that was also preceded by protection of endogenous glia from hypoxia-induced degeneration. Together, these data suggest that vascular-associated glia are also required for normalized revascularization of the hypoxic retina. Methods developed to target and protect glial cells may provide a novel strategy by which normalized revascularization can be promoted and the consequences of abnormal NV in retinal vascular diseases can be prevented.
Collapse
Affiliation(s)
- Michael I Dorrell
- Department of Cell Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | | | | | | | | | | |
Collapse
|