1
|
Modaresinejad M, Yang X, Mohammad Nezhady MA, Zhu T, Bajon E, Hou X, Tahiri H, Hardy P, Rivera JC, Lachapelle P, Chemtob S. Endoplasmic Reticulum Stress Delays Choroid Development in the HCAR1 Knockout Mouse. THE AMERICAN JOURNAL OF PATHOLOGY 2024:S0002-9440(24)00354-7. [PMID: 39332673 DOI: 10.1016/j.ajpath.2024.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 08/20/2024] [Accepted: 09/03/2024] [Indexed: 09/29/2024]
Abstract
The subretina, composed of the choroid and the retinal pigment epithelium (RPE), bears a critical role in proper vision. In addition to phagocytosis of photoreceptor debris, the RPE shuttles oxygen and nutrients to the neuroretina. For their own energy production, RPE cells mainly rely on lactate, a major by-product of glycolysis. Lactate, in turn, is believed to convey most of its biological effects via the hydroxycarboxylic acid receptor 1 (HCAR1). Here, the lactate-specific receptor, HCAR1, is found to be exclusively expressed in the RPE cells within the subretina, and Hcar1-/- mice exhibit a substantially thinner choroidal vasculature during development. Notably, the angiogenic properties of lactate on the choroid are impacted by the absence of Hcar1. HCAR1-deficient mice exhibit elevated endoplasmic reticulum stress along with eukaryotic initiation factor 2α phosphorylation, a significant decrease in the global protein translation rate, and a lower proliferation rate of choroidal vasculature. Strikingly, inhibition of the integrated stress response using an inhibitor that reverses the effect of eukaryotic initiation factor 2α phosphorylation restores protein translation and rescues choroidal thinning. These results provide evidence that lactate signalling via HCAR1 is important for choroidal development/angiogenesis and highlight the importance of this receptor in establishing mature vision.
Collapse
Affiliation(s)
- Monir Modaresinejad
- Program in Biomedical Science, Faculty of Medicine, Université de Montréal, Montreal, Quebec; Department of Pediatrics, Ophthalmology and Pharmacology, Centre de Recherche du CHU Sainte-Justine, Montréal, Quebec
| | - Xiaojuan Yang
- School of Optometry, Université de Montréal, Montreal, Quebec; Department of Pediatrics, Ophthalmology and Pharmacology, Centre de Recherche du CHU Sainte-Justine, Montréal, Quebec; Departments of Ophthalmology and Neurology-Neurosurgery, Research Institute of the McGill University Health Centre-Montreal Children's Hospital, Montreal, Quebec, Canada
| | - Mohammad Ali Mohammad Nezhady
- Department of Pediatrics, Ophthalmology and Pharmacology, Centre de Recherche du CHU Sainte-Justine, Montréal, Quebec; Program in Molecular Biology, Faculty of Medicine, Université de Montréal, Montreal, Quebec
| | - Tang Zhu
- Department of Pediatrics, Ophthalmology and Pharmacology, Centre de Recherche du CHU Sainte-Justine, Montréal, Quebec
| | - Emmanuel Bajon
- Department of Pediatrics, Ophthalmology and Pharmacology, Centre de Recherche du CHU Sainte-Justine, Montréal, Quebec
| | - Xin Hou
- Department of Pediatrics, Ophthalmology and Pharmacology, Centre de Recherche du CHU Sainte-Justine, Montréal, Quebec
| | - Houda Tahiri
- Department of Pediatrics, Ophthalmology and Pharmacology, Centre de Recherche du CHU Sainte-Justine, Montréal, Quebec
| | - Pierre Hardy
- Department of Pediatrics, Ophthalmology and Pharmacology, Centre de Recherche du CHU Sainte-Justine, Montréal, Quebec
| | - José Carlos Rivera
- Department of Pediatrics, Ophthalmology and Pharmacology, Centre de Recherche du CHU Sainte-Justine, Montréal, Quebec
| | - Pierre Lachapelle
- Departments of Ophthalmology and Neurology-Neurosurgery, Research Institute of the McGill University Health Centre-Montreal Children's Hospital, Montreal, Quebec, Canada
| | - Sylvain Chemtob
- Program in Biomedical Science, Faculty of Medicine, Université de Montréal, Montreal, Quebec; School of Optometry, Université de Montréal, Montreal, Quebec; Department of Pediatrics, Ophthalmology and Pharmacology, Centre de Recherche du CHU Sainte-Justine, Montréal, Quebec.
| |
Collapse
|
2
|
Liang P, Ness J, Rapp J, Boneva S, Schwämmle M, Jung M, Schlunck G, Agostini H, Bucher F. Characterization of the angiomodulatory effects of Interleukin 11 cis- and trans-signaling in the retina. J Neuroinflammation 2024; 21:230. [PMID: 39294742 PMCID: PMC11412048 DOI: 10.1186/s12974-024-03223-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 09/03/2024] [Indexed: 09/21/2024] Open
Abstract
BACKGROUND The IL-6 cytokine family, with its crucial and pleiotropic intracellular signaling pathway STAT3, is a promising target for treating vasoproliferative retinal diseases. Previous research has shown that IL-6 cis-signaling (via membrane-bound receptors) and trans-signaling (via soluble receptors) can have distinct effects on target cells, leading to their application in various disease treatments. While IL-6 has been extensively studied, less is known about the angiogenic effects of IL-11, another member of the IL-6 family, in the retina. Therefore, the aim of this study was to characterize the effects of IL-11 on retinal angiogenesis. MAIN TEXT In vitreous samples from proliferative diabetic retinopathy (PDR) patients, elevated levels of IL-11Rα, but not IL-11, were detected. In vitro studies using vascular endothelial cells revealed distinct effects of cis- and trans-signaling: cis-signaling (IL-11 alone) had antiangiogenic effects, while trans-signaling (IL-11 + sIL-11Rα) had proangiogenic and pro-migratory effects. These differences can be attributed to their individual signaling responses and associated transcriptomic changes. Notably, no differences in cis- and trans-signaling were detected in primary mouse Müller cell cultures. STAT3 and STAT1 siRNA knockdown experiments revealed opposing effects on IL-11 signaling, with STAT3 functioning as an antiproliferative and proapoptotic player while STAT1 acts in opposition to STAT3. In vivo, both IL-11 and IL-11 + sIL-11Rα led to a reduction in retinal neovascularization. Immunohistochemical staining revealed Müller cell activation in response to treatment, suggesting that IL-11 affects multiple retinal cell types in vivo beyond vascular endothelial cells. CONCLUSIONS Cis- and trans-signaling by IL-11 have contrasting angiomodulatory effects on endothelial cells in vitro. In vivo, cis- and trans-signaling also influence Müller cells, ultimately determining the overall angiomodulatory impact on the retina, highlighting the intricate interplay between vascular and glial cells in the retina.
Collapse
Affiliation(s)
- Paula Liang
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Klinik für Augenheilkunde, Kilianstrasse 5, 79106, Freiburg im Breisgau, Germany
| | - Jan Ness
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Klinik für Augenheilkunde, Kilianstrasse 5, 79106, Freiburg im Breisgau, Germany
- Institute of Pharmaceutical Sciences, Faculty of Chemistry and Pharmacy, University of Freiburg, Freiburg, Germany
| | - Julian Rapp
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Klinik für Augenheilkunde, Kilianstrasse 5, 79106, Freiburg im Breisgau, Germany
- Department of Medicine I, Medical Center - Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Stefaniya Boneva
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Klinik für Augenheilkunde, Kilianstrasse 5, 79106, Freiburg im Breisgau, Germany
| | - Melanie Schwämmle
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Klinik für Augenheilkunde, Kilianstrasse 5, 79106, Freiburg im Breisgau, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Malte Jung
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Klinik für Augenheilkunde, Kilianstrasse 5, 79106, Freiburg im Breisgau, Germany
| | - Günther Schlunck
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Klinik für Augenheilkunde, Kilianstrasse 5, 79106, Freiburg im Breisgau, Germany
| | - Hansjürgen Agostini
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Klinik für Augenheilkunde, Kilianstrasse 5, 79106, Freiburg im Breisgau, Germany
| | - Felicitas Bucher
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Klinik für Augenheilkunde, Kilianstrasse 5, 79106, Freiburg im Breisgau, Germany.
| |
Collapse
|
3
|
Yagi H, Boeck M, Petrishka-Lozenska M, Lundgren P, Kasai T, Cagnone G, Neilsen K, Wang C, Lee J, Tomita Y, Singh SA, Joyal JS, Aikawa M, Negishi K, Fu Z, Hellström A, Smith LEH. Timed topical dexamethasone eye drops improve mitochondrial function to prevent severe retinopathy of prematurity. Angiogenesis 2024:10.1007/s10456-024-09948-2. [PMID: 39287727 DOI: 10.1007/s10456-024-09948-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 08/30/2024] [Indexed: 09/19/2024]
Abstract
Pathological neovascularization in retinopathy of prematurity (ROP) can cause visual impairment in preterm infants. Current ROP treatments which are not preventative and only address late neovascular ROP, are costly and can lead to severe complications. We showed that topical 0.1% dexamethasone eye drops administered prior to peak neovessel formation prevented neovascularization in five extremely preterm infants at high risk for ROP and suppressed neovascularization by 30% in mouse oxygen-induced retinopathy (OIR) modeling ROP. In contrast, in OIR, topical dexamethasone treatment before any neovessel formation had limited efficacy in preventing later neovascularization, while treatment after peak neovessel formation had a non-statistically significant trend to exacerbating disease. Optimally timed topical dexamethasone suppression of neovascularization in OIR was associated with increased retinal mitochondrial gene expression and decreased inflammatory marker expression, predominantly found in immune cells. Blocking mitochondrial ATP synthetase reversed the inhibitory effect of dexamethasone on neovascularization in OIR. This study provides new insights into topical steroid effects in retinal neovascularization and into mitochondrial function in phase II ROP, and suggests a simple clinical approach to prevent severe ROP.
Collapse
Affiliation(s)
- Hitomi Yagi
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, 3 Blackfan Circle, CLS 18, Boston, MA, 02115, USA
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Myriam Boeck
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, 3 Blackfan Circle, CLS 18, Boston, MA, 02115, USA
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, 79106, Germany
| | - Mariya Petrishka-Lozenska
- The Sahlgrenska Centre for Pediatric Ophthalmology Research, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, 40530, Sweden
| | - Pia Lundgren
- The Sahlgrenska Centre for Pediatric Ophthalmology Research, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, 40530, Sweden
| | - Taku Kasai
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Gael Cagnone
- CHU Sainte-Justine Research Center, Montreal, QC, CA, H3T 1C5, Canada
- Department of Pediatrics, Ophthalmology, and Pharmacology, CHU Sainte-Justine, Université de Montréal, Montreal, QC, CA, H3T 1C5, Canada
| | - Katherine Neilsen
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, 3 Blackfan Circle, CLS 18, Boston, MA, 02115, USA
| | - Chaomei Wang
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, 3 Blackfan Circle, CLS 18, Boston, MA, 02115, USA
| | - Jeff Lee
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, 3 Blackfan Circle, CLS 18, Boston, MA, 02115, USA
| | - Yohei Tomita
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, 3 Blackfan Circle, CLS 18, Boston, MA, 02115, USA
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Sasha A Singh
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Jean-Sébastien Joyal
- CHU Sainte-Justine Research Center, Montreal, QC, CA, H3T 1C5, Canada
- Department of Pediatrics, Ophthalmology, and Pharmacology, CHU Sainte-Justine, Université de Montréal, Montreal, QC, CA, H3T 1C5, Canada
- Department of Ophthalmology, Université de Montréal, Montreal, QC, CA, H3T 1J4, Canada
| | - Masanori Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Kazuno Negishi
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Zhongjie Fu
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, 3 Blackfan Circle, CLS 18, Boston, MA, 02115, USA
| | - Ann Hellström
- The Sahlgrenska Centre for Pediatric Ophthalmology Research, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, 40530, Sweden.
| | - Lois E H Smith
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, 3 Blackfan Circle, CLS 18, Boston, MA, 02115, USA.
| |
Collapse
|
4
|
Maurya M, Liu CH, Bora K, Kushwah N, Pavlovich MC, Wang Z, Chen J. Animal Models of Retinopathy of Prematurity: Advances and Metabolic Regulators. Biomedicines 2024; 12:1937. [PMID: 39335451 PMCID: PMC11428941 DOI: 10.3390/biomedicines12091937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 08/08/2024] [Accepted: 08/15/2024] [Indexed: 09/30/2024] Open
Abstract
Retinopathy of prematurity (ROP) is a primary cause of visual impairment and blindness in premature newborns, characterized by vascular abnormalities in the developing retina, with microvascular alteration, neovascularization, and in the most severe cases retinal detachment. To elucidate the pathophysiology and develop therapeutics for ROP, several pre-clinical experimental models of ROP were developed in different species. Among them, the oxygen-induced retinopathy (OIR) mouse model has gained the most popularity and critically contributed to our current understanding of pathological retinal angiogenesis and the discovery of potential anti-angiogenic therapies. A deeper comprehension of molecular regulators of OIR such as hypoxia-inducible growth factors including vascular endothelial growth factors as primary perpetrators and other new metabolic modulators such as lipids and amino acids influencing pathological retinal angiogenesis is also emerging, indicating possible targets for treatment strategies. This review delves into the historical progressions that gave rise to the modern OIR models with a focus on the mouse model. It also reviews the fundamental principles of OIR, recent advances in its automated assessment, and a selected summary of metabolic investigation enabled by OIR models including amino acid transport and metabolism.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jing Chen
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| |
Collapse
|
5
|
Wang X, Wang T, Kaneko S, Kriukov E, Lam E, Szczepan M, Chen J, Gregg A, Wang X, Fernandez-Gonzalez A, Mitsialis SA, Kourembanas S, Baranov P, Sun Y. Photoreceptors inhibit pathological retinal angiogenesis through transcriptional regulation of Adam17 via c-Fos. Angiogenesis 2024; 27:379-395. [PMID: 38483712 PMCID: PMC11303108 DOI: 10.1007/s10456-024-09912-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 03/03/2024] [Indexed: 04/11/2024]
Abstract
Pathological retinal angiogenesis profoundly impacts visual function in vascular eye diseases, such as retinopathy of prematurity (ROP) in preterm infants and age-related macular degeneration in the elderly. While the involvement of photoreceptors in these diseases is recognized, the underlying mechanisms remain unclear. This study delved into the pivotal role of photoreceptors in regulating abnormal retinal blood vessel growth using an oxygen-induced retinopathy (OIR) mouse model through the c-Fos/A disintegrin and metalloprotease 17 (Adam17) axis. Our findings revealed a significant induction of c-Fos expression in rod photoreceptors, and c-Fos depletion in these cells inhibited pathological neovascularization and reduced blood vessel leakage in the OIR mouse model. Mechanistically, c-Fos directly regulated the transcription of Adam17 a shedding protease responsible for the production of bioactive molecules involved in inflammation, angiogenesis, and cell adhesion and migration. Furthermore, we demonstrated the therapeutic potential by using an adeno-associated virus carrying a rod photoreceptor-specific short hairpin RNA against c-fos which effectively mitigated abnormal retinal blood vessel overgrowth, restored retinal thickness, and improved electroretinographic (ERG) responses. In conclusion, this study highlights the significance of photoreceptor c-Fos in ROP pathology, offering a novel perspective for the treatment of this disease.
Collapse
Affiliation(s)
- Xudong Wang
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Tianxi Wang
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Satoshi Kaneko
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Emil Kriukov
- Department of Ophthalmology, The Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Enton Lam
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Manon Szczepan
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jasmine Chen
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Austin Gregg
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Xingyan Wang
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Angeles Fernandez-Gonzalez
- Division of Newborn Medicine, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - S Alex Mitsialis
- Division of Newborn Medicine, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Stella Kourembanas
- Division of Newborn Medicine, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Petr Baranov
- Department of Ophthalmology, The Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Ye Sun
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
6
|
Gregg AT, Wang T, Szczepan M, Lam E, Yagi H, Neilsen K, Wang X, Smith LEH, Sun Y. Botulinum neurotoxin serotype A inhibited ocular angiogenesis through modulating glial activation via SOCS3. Angiogenesis 2024:10.1007/s10456-024-09935-7. [PMID: 38922557 DOI: 10.1007/s10456-024-09935-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 06/18/2024] [Indexed: 06/27/2024]
Abstract
BACKGROUND Pathological angiogenesis causes significant vision loss in neovascular age-related macular degeneration and other retinopathies with neovascularization (NV). Neuronal/glial-vascular interactions influence the release of angiogenic and neurotrophic factors. We hypothesized that botulinum neurotoxin serotype A (BoNT/A) modulates pathological endothelial cell proliferation through glial cell activation and growth factor release. METHODS A laser-induced choroidal NV (CNV) was employed to investigate the anti-angiogenic effects of BoNT/A. Fundus fluorescence angiography, immunohistochemistry, and real-time PCR were used to assess BoNT/A efficacy in inhibiting CNV and the molecular mechanisms underlying this inhibition. Neuronal and glial suppressor of cytokine signaling 3 (SOCS3) deficient mice were used to investigate the molecular mechanisms of BoNT/A in inhibiting CNV via SOCS3. FINDINGS In laser-induced CNV mice with intravitreal BoNT/A treatment, CNV lesions decreased > 30%; vascular leakage and retinal glial activation were suppressed; and Socs3 mRNA expression was induced while vascular endothelial growth factor A (Vegfa) mRNA expression was suppressed. The protective effects of BoNT/A on CNV development were diminished in mice lacking neuronal/glial SOCS3. CONCLUSION BoNT/A suppressed laser-induced CNV and glial cell activation, in part through SOCS3 induction in neuronal/glial cells. BoNT/A treatment led to a decrease of pro-angiogenic factors, including VEGFA, highlighting the potential of BoNT/A as a therapeutic intervention for pathological angiogenesis in retinopathies.
Collapse
Affiliation(s)
- Austin T Gregg
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Tianxi Wang
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Manon Szczepan
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Enton Lam
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Hitomi Yagi
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Katherine Neilsen
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Xingyan Wang
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Lois E H Smith
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
| | - Ye Sun
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
7
|
Yagi H, Boeck M, Petrishka-Lozenska M, Lundgren P, Kasai T, Cagnone G, Wang C, Lee J, Tomita Y, Singh SA, Joyal JS, Aikawa M, Negishi K, Fu Z, Hellström A, Smith LEH. Timed topical dexamethasone eye drops improve mitochondrial function to prevent severe retinopathy of prematurity. RESEARCH SQUARE 2024:rs.3.rs-4619093. [PMID: 38978601 PMCID: PMC11230485 DOI: 10.21203/rs.3.rs-4619093/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Pathological neovascularization in retinopathy of prematurity (ROP) can cause visual impairment in preterm infants. Current ROP treatments which are not preventative and only address late neovascular ROP, are costly and can lead to severe complications. We showed that topical 0.1% dexamethasone eye drops administered prior to peak neovessel formation prevented neovascularization in five extremely preterm infants at high risk for ROP and suppressed neovascularization by 30% in mouse oxygen-induced retinopathy (OIR) modeling ROP. In contrast, in OIR, topical dexamethasone treatment before any neovessel formation had limited efficacy in preventing later neovascularization, while treatment after peak neovessel formation had a non-statistically significant trend to exacerbating disease. Optimally timed topical dexamethasone suppression of neovascularization in OIR was associated with increased retinal mitochondrial gene expression and decreased inflammatory marker expression, predominantly found in immune cells. Blocking mitochondrial ATP synthetase reversed the inhibitory effect of dexamethasone on neovascularization in OIR. This study provides new insights into topical steroid effects in retinal neovascularization and into mitochondrial function in phase II ROP, and suggests a simple clinical approach to prevent severe ROP.
Collapse
Affiliation(s)
| | | | | | | | | | - Gael Cagnone
- CHU Sainte-Justine, Université de Montréal
- Boston Children's Hospital
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Wang T, Kaneko S, Kriukov E, Alvarez D, Lam E, Wang Y, La Manna S, Marasco D, Fernandez-Gonzalez A, Mitsialis SA, Kourembanas S, Stahl A, Chen M, Xu H, Baranov P, Cai G, von Andrian UH, Sun Y. SOCS3 regulates pathological retinal angiogenesis through modulating SPP1 expression in microglia and macrophages. Mol Ther 2024; 32:1425-1444. [PMID: 38504518 PMCID: PMC11081920 DOI: 10.1016/j.ymthe.2024.03.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 02/18/2024] [Accepted: 03/15/2024] [Indexed: 03/21/2024] Open
Abstract
Pathological ocular angiogenesis has long been associated with myeloid cell activation. However, the precise cellular and molecular mechanisms governing the intricate crosstalk between the immune system and vascular changes during ocular neovascularization formation remain elusive. In this study, we demonstrated that the absence of the suppressor of cytokine signaling 3 (SOCS3) in myeloid cells led to a substantial accumulation of microglia and macrophage subsets during the neovascularization process. Our single-cell RNA sequencing data analysis revealed a remarkable increase in the expression of the secreted phosphoprotein 1 (Spp1) gene within these microglia and macrophages, identifying subsets of Spp1-expressing microglia and macrophages during neovascularization formation in angiogenesis mouse models. Notably, the number of Spp1-expressing microglia and macrophages exhibited further elevation during neovascularization in mice lacking myeloid SOCS3. Moreover, our investigation unveiled the Spp1 gene as a direct transcriptional target gene of signal transducer and activator of transcription 3. Importantly, pharmaceutical activation of SOCS3 or blocking of SPP1 resulted in a significant reduction in pathological neovascularization. In conclusion, our study highlights the pivotal role of the SOCS3/STAT3/SPP1 axis in the regulation of pathological retinal angiogenesis.
Collapse
Affiliation(s)
- Tianxi Wang
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Satoshi Kaneko
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Emil Kriukov
- Department of Ophthalmology, The Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - David Alvarez
- Department of Immunology and HMS Center for Immune Imaging, Harvard Medical School, Boston, MA 02115, USA
| | - Enton Lam
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Yidi Wang
- Department of Immunology and HMS Center for Immune Imaging, Harvard Medical School, Boston, MA 02115, USA
| | - Sara La Manna
- Department of Pharmacy, University of Naples "Federico II", 80138 Naples, Italy
| | - Daniela Marasco
- Department of Pharmacy, University of Naples "Federico II", 80138 Naples, Italy
| | - Angeles Fernandez-Gonzalez
- Division of Newborn Medicine, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - S Alex Mitsialis
- Division of Newborn Medicine, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Stella Kourembanas
- Division of Newborn Medicine, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Andreas Stahl
- Department of Ophthalmology, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Mei Chen
- Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, UK
| | - Heping Xu
- Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, UK
| | - Petr Baranov
- Department of Ophthalmology, The Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Guoshuai Cai
- Department of Surgery, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Ulrich H von Andrian
- Department of Immunology and HMS Center for Immune Imaging, Harvard Medical School, Boston, MA 02115, USA; The Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Ye Sun
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
9
|
Wu J, Jo DH, Fruttiger M, Kim JH. Cone cell dysfunction attenuates retinal neovascularization in oxygen-induced retinopathy mouse model. J Neurosci Res 2024; 102:e25316. [PMID: 38415926 DOI: 10.1002/jnr.25316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 01/21/2024] [Accepted: 02/19/2024] [Indexed: 02/29/2024]
Abstract
Aberrant neovascularization is the most common feature in retinopathy of prematurity (ROP), which leads to the retinal detachment and visual defects in neonates with a low gestational age eventually. Understanding the regulation of inappropriate angiogenic signaling benefits individuals at-risk. Recently, neural activity originating from the specific neural activity has been considered to contribute to retinal angiogenesis. Here, we explored the impact of cone cell dysfunction on oxygen-induced retinopathy (OIR), a mouse model commonly employed to understand retinal diseases associated with abnormal blood vessel growth, using the Gnat2cpfl3 (cone photoreceptor function loss-3) strain of mice (regardless of the sex), which is known for its inherent cone cell dysfunction. We found that the retinal avascular area, hypoxic area, and neovascular area were significantly attenuated in Gnat2cpfl3 OIR mice compared to those in C57BL/6 OIR mice. Moreover, the HIF-1α/VEGF axis was also reduced in Gnat2cpfl3 OIR mice. Collectively, our results indicated that cone cell dysfunction, as observed in Gnat2cpfl3 OIR mice, leads to attenuated retinal neovascularization. This finding suggests that retinal neural activity may precede and potentially influence the onset of pathological neovascularization.
Collapse
Affiliation(s)
- Jun Wu
- Fight Against Angiogenesis-Related Blindness (FARB) Laboratory, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Dong Hyun Jo
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Marcus Fruttiger
- UCL Institute of Ophthalmology, University College London, London, UK
| | - Jeong Hun Kim
- Fight Against Angiogenesis-Related Blindness (FARB) Laboratory, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Ophthalmology, Seoul National University College of Medicine, Seoul, Republic of Korea
- Global Excellence Center for Gene & Cell Therapy (GEC-GCT), Seoul National University Hospital, Seoul, Republic of Korea
- Institute of Reproductive Medicine and Population, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
10
|
Rapp J, Hospach A, Liang P, Schwämmle M, Renz L, Agostini H, Schlunck G, Bucher F. Oncostatin M Reduces Pathological Neovascularization in the Retina Through Müller Cell Activation. Invest Ophthalmol Vis Sci 2024; 65:22. [PMID: 38190125 PMCID: PMC10777876 DOI: 10.1167/iovs.65.1.22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 12/01/2023] [Indexed: 01/09/2024] Open
Abstract
Purpose Continuous vision loss due to vasoproliferative eye disease still represents an unsolved issue despite anti-vascular endothelial growth factor (VEGF) therapy. The impact of signal transducer and activator of transcription 3 (STAT3) signaling on retinal angiogenesis and its potential use as a therapeutic target remain controversial. In vitro, oncostatin M (OSM), as a strong STAT3 activator, possesses robust proangiogenic activity. This study investigated to what extent the proangiogenic effects of OSM translate to the in vivo setting of vasoproliferative eye disease. Methods The in vitro effect of OSM on endothelial cells was investigated in the spheroid sprouting assay and through RNA sequencing. The mouse model for oxygen-induced retinopathy (OIR) was used to evaluate the impact of OSM in vivo. Signaling patterns were measured by western blot and retinal cryosections. Primary Müller cell cultures were used to evaluate the effect of OSM on the Müller cell secretome. Murine retinal vascular endothelial cells were isolated from OIR retinas using fluorescence-activated cell sorting (FACS) and were used for RNA sequencing. Results Although OSM induced pro-angiogenic responses in vitro, in the OIR model intravitreal injection of OSM reduced retinal neovascularization by 65.2% and vaso-obliteration by 45.5% in Müller cells. Injecting OSM into the vitreous activated the STAT3 signaling pathway in multiple retinal cell types, including Müller cells. In vitro, OSM treatment increased CXCL10 secretion. RNA sequencing of sorted vascular endothelial cells at OIR P17 following OSM treatment indicated downregulation of angiogenesis- and mitosis-associated genes. Conclusions In vivo, OSM reveals a beneficial angiomodulatory effect by activating Müller cells and changing their secretome. The data highlight contradictions between cytokine-induced effects in vitro and in vivo depending on the cell types mediating the effect.
Collapse
Affiliation(s)
- Julian Rapp
- Eye Center, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Alban Hospach
- Eye Center, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Paula Liang
- Eye Center, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Melanie Schwämmle
- Eye Center, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Lisa Renz
- Eye Center, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Hansjürgen Agostini
- Eye Center, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Günther Schlunck
- Eye Center, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Felicitas Bucher
- Eye Center, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
11
|
Pauleikhoff L, Boneva S, Boeck M, Schlecht A, Schlunck G, Agostini H, Lange C, Wolf J. Transcriptional Comparison of Human and Murine Retinal Neovascularization. Invest Ophthalmol Vis Sci 2023; 64:46. [PMID: 38153746 PMCID: PMC10756240 DOI: 10.1167/iovs.64.15.46] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 11/30/2023] [Indexed: 12/29/2023] Open
Abstract
Purpose Retinal neovascularization (RNV) is the leading cause of vision loss in diseases like proliferative diabetic retinopathy (PDR). A significant failure rate of current treatments indicates the need for novel treatment targets. Animal models are crucial in this process, but current diabetic retinopathy models do not develop RNV. Although the nondiabetic oxygen-induced retinopathy (OIR) mouse model is used to study RNV development, it is largely unknown how closely it resembles human PDR. Methods We therefore performed RNA sequencing on murine (C57BL/6J) OIR retinas (n = 14) and human PDR RNV membranes (n = 7) extracted during vitrectomy, each with reference to control tissue (n=13/10). Differentially expressed genes (DEG) and associated biological processes were analyzed and compared between human and murine RNV to assess molecular overlap and identify phylogenetically conserved factors. Results In total, 213 murine- and 1223 human-specific factors were upregulated with a small overlap of 94 DEG (7% of human DEG), although similar biological processes such as angiogenesis, regulation of immune response, and extracellular matrix organization were activated in both species. Phylogenetically conserved mediators included ANGPT2, S100A8, MCAM, EDNRA, and CCR7. Conclusions Even though few individual genes were upregulated simultaneously in both species, similar biological processes appeared to be activated. These findings demonstrate the potential and limitations of the OIR model to study human PDR and identify phylogenetically conserved potential treatment targets for PDR.
Collapse
Affiliation(s)
- Laurenz Pauleikhoff
- Eye Center, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Department of Ophthalmology, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Stefaniya Boneva
- Eye Center, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Myriam Boeck
- Eye Center, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Anja Schlecht
- Institute of Anatomy and Cell Biology, University of Würzburg, Würzburg, Germany
| | - Günther Schlunck
- Eye Center, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Hansjürgen Agostini
- Eye Center, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Clemens Lange
- Eye Center at St. Franziskus Hospital, Münster, Germany
| | - Julian Wolf
- Eye Center, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Molecular Surgery Laboratory, Department of Ophthalmology, Byers Eye Institute, Stanford University, Palo Alto, California, United States
| |
Collapse
|
12
|
Harman JC, Pivodic A, Nilsson AK, Boeck M, Yagi H, Neilsen K, Ko M, Yang J, Kinter M, Hellström A, Fu Z. Postnatal hyperglycemia alters amino acid profile in retinas (model of Phase I ROP). iScience 2023; 26:108021. [PMID: 37841591 PMCID: PMC10568433 DOI: 10.1016/j.isci.2023.108021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 05/03/2023] [Accepted: 09/19/2023] [Indexed: 10/17/2023] Open
Abstract
Nutritional deprivation occurring in most preterm infants postnatally can induce hyperglycemia, a significant and independent risk factor for suppressing physiological retinal vascularization (Phase I retinopathy of prematurity (ROP)), leading to compensatory but pathological neovascularization. Amino acid supplementation reduces retinal neovascularization in mice. Little is known about amino acid contribution to Phase I ROP. In mice modeling hyperglycemia-associated Phase I ROP, we found significant changes in retinal amino acids (including most decreased L-leucine, L-isoleucine, and L-valine). Parenteral L-isoleucine suppressed physiological retinal vascularization. In premature infants, severe ROP was associated with a higher mean intake of parenteral versus enteral amino acids in the first two weeks of life after adjustment for treatment group, gestational age at birth, birth weight, and sex. The number of days with parenteral amino acids support independently predicted severe ROP. Further understanding and modulating amino acids may help improve nutritional intervention and prevent Phase I ROP.
Collapse
Affiliation(s)
- Jarrod C. Harman
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Aldina Pivodic
- The Sahlgrenska Centre for Pediatric Ophthalmology Research, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Anders K. Nilsson
- The Sahlgrenska Centre for Pediatric Ophthalmology Research, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Myriam Boeck
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Hitomi Yagi
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Ophthalmology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Katherine Neilsen
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Minji Ko
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jay Yang
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Michael Kinter
- Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Ann Hellström
- The Sahlgrenska Centre for Pediatric Ophthalmology Research, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Zhongjie Fu
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
13
|
Čolić A, Vukojević N, Anić Jurica S. ASSESSMENT OF NEONATAL CARE STANDARD BY THE PREDICTIVE MODEL FOR RETINOPATHY OF PREMATURITY BASED ON WEIGHT GAIN. Acta Clin Croat 2023; 62:175-183. [PMID: 38304365 PMCID: PMC10829965 DOI: 10.20471/acc.2023.62.01.20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Accepted: 02/26/2023] [Indexed: 02/03/2024] Open
Abstract
Care of extremely premature infants is in constant need for evaluation and progress. WINROP, a predictive model based on weight gain, has been developed to reduce the number of stressful examinations for retinopathy for prematurity. Validation studies of WINROP emphasize the difference of applicability in neonatal units of various practice. The aim of the study was to assess the standard of neonatal care by WINROP. Data on extremely premature infants were collected from medical records and entered in WINROP. High- and low-risk WINROP distribution and retinopathy of prematurity outcomes were analyzed. Fifty-four infants, gestational age ≤28 weeks, were included in the study after exclusion of weight related comorbidities. High risk was noted in 74% (n=40) of infants with 24% (n=13) developing retinopathy of prematurity requiring treatment. In low alarm group, there were 3 cases with severe disease. In conclusion, WINROP is not just a provider of predictive information on the severity of retinopathy of prematurity. High-risk alarm indicates the need of adjustment of nutritional strategies. Infants without pathological growth morbidities who develop severe retinopathy of prematurity in low-risk group point to other risk factors for retinopathy of prematurity to be evaluated and changed in future practice.
Collapse
Affiliation(s)
- Ana Čolić
- Division of Neonatology, Department of Gynecology and Obstetrics, Zagreb University Hospital Center, Zagreb, Croatia
| | - Nenad Vukojević
- School of Medicine, University of Zagreb, Department of Ophthalmology, Zagreb University Hospital Center, Zagreb, Croatia
| | - Sonja Anić Jurica
- Division of Neonatology, Department of Gynecology and Obstetrics, Zagreb University Hospital Center, Zagreb, Croatia
| |
Collapse
|
14
|
Liu J, Tsang JKW, Fung FKC, Chung SK, Fu Z, Lo ACY. Retinal microglia protect against vascular damage in a mouse model of retinopathy of prematurity. Front Pharmacol 2022; 13:945130. [PMID: 36059936 PMCID: PMC9431881 DOI: 10.3389/fphar.2022.945130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 07/11/2022] [Indexed: 11/25/2022] Open
Abstract
Retinopathy of prematurity (ROP) is a common cause of blindness in preterm babies. As a hypoxia-induced eye disease characterized by neovascularization, its association with retinal microglia has been noted but not well documented. We performed a comprehensive analysis of retinal microglia and retinal vessels in mouse oxygen-induced retinopathy (OIR), an animal model of ROP. In combination with a pharmacological inhibitory strategy, the role of retinal microglia in vascular network maintenance was investigated. Postnatal day (P) 7 C57BL/6J mouse pups with their nursing mother were exposed to 75% oxygen for 5 days to induce OIR. Age-matched room air-treated pups served as controls. On P12, P17, P21, P25, and P30, retinal microglia and vessels were visualized and quantified based on their location and activation status. Their relationship with retinal vessels was also analyzed. On P5 or P12, retinal microglia inhibition was achieved by intravitreal injection of liposomes containing clodronate (CLD); retinal vasculature and microglia were examined in P12 and P17 OIR retinae. The number of retinal microglia was increased in the superficial areas of OIR retinae on P12, P17, P21, P25, and P30, and most of them displayed an amoeboid (activated) morphology. The increased retinal microglia were associated with increased superficial retinal vessels in OIR retinae. The number of retinal microglia in deep retinal areas of OIR retinae also increased from P17 to P30 with a ramified morphology, which was not associated with reduced retinal vessels. Intravitreal injection of liposomes-CLD caused a significant reduction in retinal microglia. Loss of retinal microglia before hyperoxia treatment resulted in increased vessel obliteration on P12 and subsequent neovascularization on P17 in OIR retinae. Meanwhile, loss of retinal microglia immediately after hyperoxia treatment on P12 also led to more neovascularization in P17 OIR retinae. Our data showed that activated microglia were strongly associated with vascular abnormalities upon OIR. Retinal microglial activation continued throughout OIR and lasted until after retinal vessel recovery. Pharmacological inhibition of retinal microglia in either hyperoxic or hypoxic stage of OIR exacerbated retinal vascular consequences. These results suggested that retinal microglia may play a protective role in retinal vasculature maintenance in the OIR process.
Collapse
Affiliation(s)
- Jin Liu
- Department of Ophthalmology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Jessica Kwan Wun Tsang
- Department of Ophthalmology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Frederic Khe Cheong Fung
- Department of Ophthalmology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Sookja Kim Chung
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Zhongjie Fu
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
- *Correspondence: Zhongjie Fu, ; Amy Cheuk Yin Lo,
| | - Amy Cheuk Yin Lo
- Department of Ophthalmology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- *Correspondence: Zhongjie Fu, ; Amy Cheuk Yin Lo,
| |
Collapse
|
15
|
Huang CW, Yeh PT, Tsao PN, Chou HC, Chen CY, Yen TA, Huang HC, Lai TT. Validation of the Postnatal Growth and Retinopathy of Prematurity Screening Criteria in a Taiwanese Cohort. Am J Ophthalmol 2022; 237:22-31. [PMID: 34780799 DOI: 10.1016/j.ajo.2021.11.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 11/04/2021] [Accepted: 11/05/2021] [Indexed: 11/29/2022]
Abstract
PURPOSE To validate the performance of Postnatal Growth and Retinopathy of Prematurity (G-ROP) screening criteria in a Taiwanese cohort. DESIGN Screening evaluation with retrospective data. METHOD Premature infants who underwent retinopathy of prematurity (ROP) screening between January 2015 and April 2019 at a tertiary hospital were examined. Infants with known final ROP results and complete longitudinal weight records were included. G-ROP screening criteria, both original and simplified (G-ROP 180 g), were applied as the prediction model for type 1 ROP; sensitivity and specificity were analyzed. The reduction in the number of infants requiring ROP screening and the number of funduscopic examinations were calculated. RESULT A total of 303 infants with documented ROP outcomes and complete weight gain records were examined. Of these, 103 infants developed ROP, of whom 29 developed type 1 ROP, whereas the other 200 did not develop ROP. For the detection of type 1 ROP, the sensitivity and specificity of the original G-ROP screening criteria were 96.6% and 42.3%, and 100% and 31%, for the simplified G-ROP 180 g model, respectively. The reduction in the number of infants requiring screening and funduscopic examinations was 32.6% and 33.5% for the original G-ROP criteria, and 28.1% and 23.2% for the G-ROP 180 g model, respectively. CONCLUSION Both the original G-ROP and G-ROP 180 g criteria attained high sensitivities in detecting type 1 ROP in the current Taiwanese cohort, with the G-ROP 180-g model outperforming the original one. Validation and modification may be required before applying G-ROP screening criteria to different populations.
Collapse
Affiliation(s)
- Ching-Wen Huang
- From the Department of Ophthalmology (C.-W.H., P.-T.Y., T.-T.L.), National Taiwan University Hospital, Taipei, Taiwan
| | - Po-Ting Yeh
- From the Department of Ophthalmology (C.-W.H., P.-T.Y., T.-T.L.), National Taiwan University Hospital, Taipei, Taiwan
| | - Po-Nien Tsao
- Department of Pediatrics (P.-N.T., H.-C.C., C.-Y.C., T.-A.Y., H.-C.H.), National Taiwan University Hospital, Taipei, Taiwan; Research Center for Developmental Biology & Regenerative Medicine (P.-N.T.), National Taiwan University, Taipei, Taiwan
| | - Hung-Chieh Chou
- Department of Pediatrics (P.-N.T., H.-C.C., C.-Y.C., T.-A.Y., H.-C.H.), National Taiwan University Hospital, Taipei, Taiwan
| | - Chien-Yi Chen
- Department of Pediatrics (P.-N.T., H.-C.C., C.-Y.C., T.-A.Y., H.-C.H.), National Taiwan University Hospital, Taipei, Taiwan
| | - Ting-An Yen
- Department of Pediatrics (P.-N.T., H.-C.C., C.-Y.C., T.-A.Y., H.-C.H.), National Taiwan University Hospital, Taipei, Taiwan
| | - Hsin-Chung Huang
- Department of Pediatrics (P.-N.T., H.-C.C., C.-Y.C., T.-A.Y., H.-C.H.), National Taiwan University Hospital, Taipei, Taiwan
| | - Tso-Ting Lai
- From the Department of Ophthalmology (C.-W.H., P.-T.Y., T.-T.L.), National Taiwan University Hospital, Taipei, Taiwan; Graduate Institute of Clinical Medicine (T.-T.L.), College of Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
16
|
Dai C, Waduge P, Ji L, Huang C, He Y, Tian H, Zuniga-Sanchez E, Bhatt A, Pang IH, Su G, Webster KA, Li W. Secretogranin III stringently regulates pathological but not physiological angiogenesis in oxygen-induced retinopathy. Cell Mol Life Sci 2022; 79:63. [PMID: 35006382 PMCID: PMC9007175 DOI: 10.1007/s00018-021-04111-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 11/29/2021] [Accepted: 12/17/2021] [Indexed: 01/12/2023]
Abstract
Conventional angiogenic factors, such as vascular endothelial growth factor (VEGF), regulate both pathological and physiological angiogenesis indiscriminately, and their inhibitors may elicit adverse side effects. Secretogranin III (Scg3) was recently reported to be a diabetes-restricted VEGF-independent angiogenic factor, but the disease selectivity of Scg3 in retinopathy of prematurity (ROP), a retinal disease in preterm infants with concurrent pathological and physiological angiogenesis, was not defined. Here, using oxygen-induced retinopathy (OIR) mice, a surrogate model of ROP, we quantified an exclusive binding of Scg3 to diseased versus healthy developing neovessels that contrasted sharply with the ubiquitous binding of VEGF. Functional immunohistochemistry visualized Scg3 binding exclusively to disease-related disorganized retinal neovessels and neovascular tufts, whereas VEGF bound to both disorganized and well-organized neovessels. Homozygous deletion of the Scg3 gene showed undetectable effects on physiological retinal neovascularization but markedly reduced the severity of OIR-induced pathological angiogenesis. Furthermore, anti-Scg3 humanized antibody Fab (hFab) inhibited pathological angiogenesis with similar efficacy to anti-VEGF aflibercept. Aflibercept dose-dependently blocked physiological angiogenesis in neonatal retinas, whereas anti-Scg3 hFab was without adverse effects at any dose and supported a therapeutic window at least 10X wider than that of aflibercept. Therefore, Scg3 stringently regulates pathological but not physiological angiogenesis, and anti-Scg3 hFab satisfies essential criteria for development as a safe and effective disease-targeted anti-angiogenic therapy for ROP.
Collapse
Affiliation(s)
- Chang Dai
- Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, USA.,Bascom Palmer Eye Institute, University of Miami School of Medicine, Miami, Florida, USA
| | - Prabuddha Waduge
- Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, USA.,Bascom Palmer Eye Institute, University of Miami School of Medicine, Miami, Florida, USA
| | - Liyang Ji
- Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, USA.,Bascom Palmer Eye Institute, University of Miami School of Medicine, Miami, Florida, USA
| | - Chengchi Huang
- Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, USA
| | - Ye He
- Bascom Palmer Eye Institute, University of Miami School of Medicine, Miami, Florida, USA
| | - Hong Tian
- Everglades Biopharma, LLC, Houston, Texas, USA
| | - Elizabeth Zuniga-Sanchez
- Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, USA
| | - Amit Bhatt
- Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, USA.,Texas Children Hospital, Baylor College of Medicine, Houston, Texas, USA
| | - Iok-Hou Pang
- Dept. of Pharmaceutical Sciences, North Texas Eye Research Institute, University of North Texas, Fort Worth, Texas, USA
| | - Guanfang Su
- Department of Ophthalmology, The Second Hospital of Jilin University, #218 Ziqiang Street, Changchun, Jilin, China
| | - Keith A. Webster
- Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, USA.,Bascom Palmer Eye Institute, University of Miami School of Medicine, Miami, Florida, USA.,Everglades Biopharma, LLC, Houston, Texas, USA
| | - Wei Li
- Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, USA.,Bascom Palmer Eye Institute, University of Miami School of Medicine, Miami, Florida, USA
| |
Collapse
|
17
|
Klaska IP, White A, Villacampa P, Hoke J, Hervás LA, Maswood RN, Ali RR, Bunce C, Unwin RD, Cooper GJS, Bishop PN, Bainbridge JW. Intravitreal administration of recombinant human opticin protects against hyperoxia-induced pre-retinal neovascularization. Exp Eye Res 2021; 215:108908. [PMID: 34954204 PMCID: PMC8935380 DOI: 10.1016/j.exer.2021.108908] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/29/2021] [Accepted: 12/20/2021] [Indexed: 01/01/2023]
Abstract
Opticin is an extracellular glycoprotein present in the vitreous. Its antiangiogenic properties offer the potential for therapeutic intervention in conditions such as proliferative diabetic retinopathy and retinopathy of prematurity. Here, we investigated the hypothesis that intravitreal administration of recombinant human opticin can safely protect against the development of pathological angiogenesis and promote its regression. We generated and purified recombinant human opticin and investigated its impact on the development and regression of pathological retinal neovascularization following intravitreal administration in murine oxygen-induced retinopathy. We also investigated its effect on normal retinal vascular development and function, following intravitreal injection in neonatal mice, by histological examination and electroretinography. In oxygen-induced retinopathy, intravitreal administration of human recombinant opticin protected against the development of retinal neovascularization to similar extent as aflibercept, which targets VEGF. Opticin also accelerated regression of established retinal neovascularization, though the effect at 18 h was less than that of aflibercept. Intravitreal administration of human recombinant opticin in neonatal mice caused no detectable perturbation of subsequent retinal vascular development or function. In summary we found that intraocular administration of recombinant human opticin protects against the development of pathological angiogenesis in mice and promotes its regression.
Collapse
Affiliation(s)
- Izabela P Klaska
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London, EC1V 9EL, UK; KCL Centre for Cell and Gene Therapy, Tower Wing, Guy's Hospital, Great Maze Pond, London, SE1 9RT, UK
| | - Anne White
- Division of Evolution & Genomic Sciences, School of Biological Sciences, FBMH, University of Manchester, Manchester, UK
| | - Pilar Villacampa
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London, EC1V 9EL, UK; Josep Carreras Leukaemia Research Institute, Ctra de Can Ruti, Barcelona, Spain
| | - Justin Hoke
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London, EC1V 9EL, UK
| | - Laura A Hervás
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London, EC1V 9EL, UK
| | - Ryea N Maswood
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London, EC1V 9EL, UK
| | - Robin R Ali
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London, EC1V 9EL, UK; KCL Centre for Cell and Gene Therapy, Tower Wing, Guy's Hospital, Great Maze Pond, London, SE1 9RT, UK
| | - Catey Bunce
- School of Population Health and Environmental Sciences, Faculty of Life Sciences and Medicine, King's College London, Addison House, London, SE1 1UL, UK
| | - Richard D Unwin
- Division of Cardiovascular Sciences, School of Medical Sciences, FBMH, University of Manchester, Manchester, UK; Stoller Biomarker Discovery Centre and Division of Cancer Sciences, School of Medical Sciences, FBMH, University of Manchester, Manchester, UK
| | - Garth J S Cooper
- Division of Cardiovascular Sciences, School of Medical Sciences, FBMH, University of Manchester, Manchester, UK
| | - Paul N Bishop
- Division of Evolution & Genomic Sciences, School of Biological Sciences, FBMH, University of Manchester, Manchester, UK; Manchester Royal Eye Hospital, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - James W Bainbridge
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London, EC1V 9EL, UK.
| |
Collapse
|
18
|
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: 0.7] [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
|
19
|
Chen DY, Sun NH, Chen X, Gong JJ, Yuan ST, Hu ZZ, Lu NN, Körbelin J, Fukunaga K, Liu QH, Lu YM, Han F. Endothelium-derived semaphorin 3G attenuates ischemic retinopathy by coordinating β-catenin-dependent vascular remodeling. J Clin Invest 2021; 131:135296. [PMID: 33586674 DOI: 10.1172/jci135296] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 12/10/2020] [Indexed: 12/14/2022] Open
Abstract
Abnormal angiogenesis and regression of the diseased retinal vasculature are key processes associated with ischemic retinopathies, but the underlying mechanisms that regulate vascular remodeling remain poorly understood. Here, we confirmed the specific expression of semaphorin 3G (Sema3G) in retinal endothelial cells (ECs), which was required for vascular remodeling and the amelioration of ischemic retinopathy. We found that Sema3G was elevated in the vitreous fluid of patients with proliferative diabetic retinopathy (PDR) and in the neovascularization regression phase of oxygen-induced retinopathy (OIR). Endothelial-specific Sema3G knockout mice exhibited decreased vessel density and excessive matrix deposition in the retinal vasculature. Moreover, loss of Sema3G aggravated pathological angiogenesis in mice with OIR. Mechanistically, we demonstrated that HIF-2α directly regulated Sema3G transcription in ECs under hypoxia. Sema3G coordinated the functional interaction between β-catenin and VE-cadherin by increasing β-catenin stability in the endothelium through the neuropilin-2 (Nrp2)/PlexinD1 receptor. Furthermore, Sema3G supplementation enhanced healthy vascular network formation and promoted diseased vasculature regression during blood vessel remodeling. Overall, we deciphered the endothelium-derived Sema3G-dependent events involved in modulating physiological vascular remodeling and regression of pathological blood vessels for reparative vascular regeneration. Our findings shed light on the protective effect of Sema3G in ischemic retinopathies.
Collapse
Affiliation(s)
- Dan-Yang Chen
- Department of Physiology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China.,Key Laboratory of Cardiovascular & Cerebrovascular Medicine, Drug Target and Drug Discovery Center, School of Pharmacy, Nanjing Medical University, Nanjing, China.,Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Ning-He Sun
- Department of Physiology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China.,Key Laboratory of Cardiovascular & Cerebrovascular Medicine, Drug Target and Drug Discovery Center, School of Pharmacy, Nanjing Medical University, Nanjing, China.,Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Xiang Chen
- Key Laboratory of Cardiovascular & Cerebrovascular Medicine, Drug Target and Drug Discovery Center, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Jun-Jie Gong
- Key Laboratory of Cardiovascular & Cerebrovascular Medicine, Drug Target and Drug Discovery Center, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Song-Tao Yuan
- Department of Ophthalmology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zi-Zhong Hu
- Department of Ophthalmology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Nan-Nan Lu
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Jakob Körbelin
- Department of Oncology and Hematology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kohji Fukunaga
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Qing-Huai Liu
- Department of Ophthalmology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ying-Mei Lu
- Department of Physiology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Feng Han
- Key Laboratory of Cardiovascular & Cerebrovascular Medicine, Drug Target and Drug Discovery Center, School of Pharmacy, Nanjing Medical University, Nanjing, China.,Institute of Brain Science, the Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
20
|
De Rossi G, Vähätupa M, Cristante E, Arokiasamy S, Liyanage SE, May U, Pellinen L, Uusitalo-Järvinen H, Bainbridge JW, Järvinen TA, Whiteford JR. Pathological Angiogenesis Requires Syndecan-4 for Efficient VEGFA-Induced VE-Cadherin Internalization. Arterioscler Thromb Vasc Biol 2021; 41:1374-1389. [PMID: 33596666 PMCID: PMC7613699 DOI: 10.1161/atvbaha.121.315941] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Giulia De Rossi
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, United Kingdom
- UCL Institute of Ophthalmology, Department of Cell Biology, 11-43 Bath Street, London EC1V 9EL, UK
| | - Maria Vähätupa
- Faculty of Medicine & Health Technology, Tampere University, 33014 Tampere, Finland & Departments of Orthopedics & Traumatology and Tampere Eye Centre, Tampere University Hospital, 33521 Tampere, Finland
| | - Enrico Cristante
- UCL Institute of Ophthalmology, Genetics department, 11-43 Bath Street, London EC1V 9EL, UK
| | - Samantha Arokiasamy
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, United Kingdom
| | - Sidath E. Liyanage
- UCL Institute of Ophthalmology, Genetics department, 11-43 Bath Street, London EC1V 9EL, UK
| | - Ulrike May
- Faculty of Medicine & Health Technology, Tampere University, 33014 Tampere, Finland & Departments of Orthopedics & Traumatology and Tampere Eye Centre, Tampere University Hospital, 33521 Tampere, Finland
| | - Laura Pellinen
- Faculty of Medicine & Health Technology, Tampere University, 33014 Tampere, Finland & Departments of Orthopedics & Traumatology and Tampere Eye Centre, Tampere University Hospital, 33521 Tampere, Finland
| | - Hannele Uusitalo-Järvinen
- Faculty of Medicine & Health Technology, Tampere University, 33014 Tampere, Finland & Departments of Orthopedics & Traumatology and Tampere Eye Centre, Tampere University Hospital, 33521 Tampere, Finland
| | - James W. Bainbridge
- UCL Institute of Ophthalmology, Genetics department, 11-43 Bath Street, London EC1V 9EL, UK
- NIHR Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust, City Road, London EC1V 2PD, UK
| | - Tero A.H. Järvinen
- Faculty of Medicine & Health Technology, Tampere University, 33014 Tampere, Finland & Departments of Orthopedics & Traumatology and Tampere Eye Centre, Tampere University Hospital, 33521 Tampere, Finland
| | - James R. Whiteford
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, United Kingdom
| |
Collapse
|
21
|
Li J, Yu S, Lu X, Cui K, Tang X, Xu Y, Liang X. The phase changes of M1/M2 phenotype of microglia/macrophage following oxygen-induced retinopathy in mice. Inflamm Res 2021; 70:183-192. [PMID: 33386422 DOI: 10.1007/s00011-020-01427-w] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 10/27/2020] [Accepted: 11/24/2020] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE Microglia/macrophage activation is previously reported to be involved in various ocular diseases. However, the separate role of M1/M2 phenotype microglia/macrophage in the pathological process of oxygen-induced retinopathy (OIR) remains unknown. In this research, we explored the role and regulatory mechanism of M1/M2 microglia/macrophage in OIR in C57BL/6J mice. Furthermore, we demonstrated the time phase of M1/M2 shifting of microglia/macrophage during the natural process of OIR, which is very essential for further investigations. MATERIALS AND METHODS C57BL/6j pups were exposed to hyperoxia environment from postnatal 7(P7) to P12 then returned to normoxia. The mice were then euthanized, and the eyes were harvested at a series of time points for further investigation. The M1/M2 phenotype microglia/macrophage activity was presented by immunofluorescent staining and real-time quantitative polymerase chain reaction (qPCR). The NF-κb-STAT3 signaling and IL-4-STAT6-PPAR-γ signaling pathway activity was examined by western blot analysis. RESULTS The microglia/macrophage were activated when the OIR model was set up after P12. The M1 microglia/macrophage activation was found in neovascularization (NV) tufts in both central and peripheral retina, which started from P12 when the mice were returned to normoxia environment and peaked at P17. During this period of time, the NF-κb-STAT3 signaling pathway was activated, resulting in the upregulated M1 phenotype microglia/macrophage polarization, along with the enhanced inflammatory cytokine expression including tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and IL-1β. Consequently, the NV tufts were observed from P12 and the volume continued to increase until P17. However, the M2 phenotype microglia/macrophage activity took over during the late phase of OIR started from P17. The IL-4-STAT6-PPAR-γ signaling activity was upregulated from P17 and peaked at P20, inducing M2 phenotype microglia polarization, which consequently led to the inhibition of inflammatory cytokines and spontaneous regression of NV tufts. CONCLUSIONS Microglia/macrophage participate actively in the natural process of OIR in mice, and two phenotypes exert different functions. Treatment modulating microglia/macrophage polarize toward M2 phenotype might be a novel and promising method for ocular neovascular diseases such as retinopathy of prematurity (ROP), wet age-related macular degeneration (wAMD), and diabetic retinopathy (DR).
Collapse
Affiliation(s)
- Jia Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, 510030, Guangdong, China
| | - Shanshan Yu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, 510030, Guangdong, China
| | - Xi Lu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, 510030, Guangdong, China
| | - Kaixuan Cui
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, 510030, Guangdong, China
| | - Xiaoyu Tang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, 510030, Guangdong, China
| | - Yue Xu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, 510030, Guangdong, China.
| | - Xiaoling Liang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, 510030, Guangdong, China.
| |
Collapse
|
22
|
Lymphocytic microparticles suppress retinal angiogenesis via targeting Müller cells in the ischemic retinopathy mouse model. Exp Cell Res 2021; 399:112470. [PMID: 33434529 DOI: 10.1016/j.yexcr.2021.112470] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 12/14/2020] [Accepted: 12/30/2020] [Indexed: 11/22/2022]
Abstract
Retinopathy of prematurity (ROP) is the primary cause of visual impairment and vision loss in premature infants, which results from the formation of aberrant retinal neovascularization (NV). An emerging body of evidence has shown that Müller cells are the predominant source of vascular endothelial growth factor (VEGF), which also serves as a chemoattractant for monocyte/macrophage lineage. The recruitment of macrophages is increased during retinal NV, and they exert a pro-angiogenic role in ROP. We have shown that lymphocytic microparticles (microvesicles; LMPs) derived from apoptotic human T lymphocytes possess strong angiogenesis-inhibiting properties. Here, we investigated the effect of LMPs on the chemotactic capacity of Müller cells in vitro using rat Müller cell rMC-1 and mouse macrophage RAW 264.7. In addition, the impact of LMPs was determined in vivo using a mouse model of oxygen-induced ischemic retinopathy (OIR). The results revealed that LMPs were internalized by rMC-1 and reduced their cell proliferation dose-dependently without inducing cell apoptosis. LMPs inhibited the chemotactic capacity of rMC-1 on RAW 264.7 via reducing the expression of VEGF. Moreover, LMPs attenuated pathological retinal NV and the infiltration of macrophages in vivo. LMPs downregulated ERK1/2 and HIF-1α both in vitro and in vivo. These findings expand our understanding of the effects of LMPs, providing evidence of LMPs as a promising therapeutic approach for the treatment of retinal NV diseases.
Collapse
|
23
|
Tomita Y, Cagnone G, Fu Z, Cakir B, Kotoda Y, Asakage M, Wakabayashi Y, Hellström A, Joyal JS, Talukdar S, Smith LEH, Usui Y. Vitreous metabolomics profiling of proliferative diabetic retinopathy. Diabetologia 2021; 64:70-82. [PMID: 33099660 PMCID: PMC7718434 DOI: 10.1007/s00125-020-05309-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 09/10/2020] [Indexed: 02/08/2023]
Abstract
AIMS/HYPOTHESIS Proliferative diabetic retinopathy (PDR) with retinal neovascularisation (NV) is a leading cause of vision loss. This study identified a set of metabolites that were altered in the vitreous humour of PDR patients compared with non-diabetic control participants. We corroborated changes in vitreous metabolites identified in prior studies and identified novel dysregulated metabolites that may lead to treatment strategies for PDR. METHODS We analysed metabolites in vitreous samples from 43 PDR patients and 21 non-diabetic epiretinal membrane control patients from Japan (age 27-80 years) via ultra-high-performance liquid chromatography-mass spectrometry. We then investigated the association of a novel metabolite (creatine) with retinal NV in mouse oxygen-induced retinopathy (OIR). Creatine or vehicle was administered from postnatal day (P)12 to P16 (during induced NV) via oral gavage. P17 retinas were quantified for NV and vaso-obliteration. RESULTS We identified 158 metabolites in vitreous samples that were altered in PDR patients vs control participants. We corroborated increases in pyruvate, lactate, proline and allantoin in PDR, which were identified in prior studies. We also found changes in metabolites not previously identified, including creatine. In human vitreous humour, creatine levels were decreased in PDR patients compared with epiretinal membrane control participants (false-discovery rate <0.001). We validated that lower creatine levels were associated with vascular proliferation in mouse retina in the OIR model (p = 0.027) using retinal metabolomics. Oral creatine supplementation reduced NV compared with vehicle (P12 to P16) in OIR (p = 0.0024). CONCLUSIONS/INTERPRETATION These results suggest that metabolites from vitreous humour may reflect changes in metabolism that can be used to find pathways influencing retinopathy. Creatine supplementation could be useful to suppress NV in PDR. Graphical abstract.
Collapse
Affiliation(s)
- Yohei Tomita
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Gael Cagnone
- Department of Pediatrics, Pharmacology and Ophthalmology, CHU Sainte-Justine Research Center, Université de Montréal, Montreal, QC, Canada
| | - Zhongjie Fu
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Manton Center for Orphan Disease, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Bertan Cakir
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Yumi Kotoda
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Masaki Asakage
- Department of Ophthalmology, Tokyo Medical University Hospital, Tokyo, Japan
| | | | - Ann Hellström
- Pediatric Ophthalmology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, Gothenburg University, Gothenburg, Sweden
| | - Jean-Sébastien Joyal
- Department of Pediatrics, Pharmacology and Ophthalmology, CHU Sainte-Justine Research Center, Université de Montréal, Montreal, QC, Canada
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | | | - Lois E H Smith
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Yoshihiko Usui
- Department of Ophthalmology, Tokyo Medical University Hospital, Tokyo, Japan.
| |
Collapse
|
24
|
Smith RO, Ninchoji T, Gordon E, André H, Dejana E, Vestweber D, Kvanta A, Claesson-Welsh L. Vascular permeability in retinopathy is regulated by VEGFR2 Y949 signaling to VE-cadherin. eLife 2020; 9:54056. [PMID: 32312382 PMCID: PMC7188482 DOI: 10.7554/elife.54056] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 04/05/2020] [Indexed: 01/01/2023] Open
Abstract
Edema stemming from leaky blood vessels is common in eye diseases such as age-related macular degeneration and diabetic retinopathy. Whereas therapies targeting vascular endothelial growth factor A (VEGFA) can suppress leakage, side-effects include vascular rarefaction and geographic atrophy. By challenging mouse models representing different steps in VEGFA/VEGF receptor 2 (VEGFR2)-induced vascular permeability, we show that targeting signaling downstream of VEGFR2 pY949 limits vascular permeability in retinopathy induced by high oxygen or by laser-wounding. Although suppressed permeability is accompanied by reduced pathological neoangiogenesis in oxygen-induced retinopathy, similarly sized lesions leak less in mutant mice, separating regulation of permeability from angiogenesis. Strikingly, vascular endothelial (VE)-cadherin phosphorylation at the Y685, but not Y658, residue is reduced when VEGFR2 pY949 signaling is impaired. These findings support a mechanism whereby VE-cadherin Y685 phosphorylation is selectively associated with excessive vascular leakage. Therapeutically, targeting VEGFR2-regulated VE-cadherin phosphorylation could suppress edema while leaving other VEGFR2-dependent functions intact. The number of people with impaired vision and blindness is increasing in Western society due to the aging population and the increased prevalence of diabetes. This has led to eye diseases, such as age-related macular degeneration and diabetic retinopathy becoming more common. In both these eye diseases, new blood vessels grow in the retina – the light-sensitive part of the eye – to bring oxygen and nutrients to the tissue. However, these new blood vessels are leaky and allow molecules to leave the bloodstream and enter the retinal tissue. This causes the retina to swell and impair a person’s vision. The leaky blood supply also reduces the amount of oxygen that gets to the tissue, resulting in further damage to the retina. When tissues experience low levels of oxygen, cells start making a protein called vascular endothelial growth factor (or VEGF for short). Whilst this protein is important for helping form new blood vessels, it also makes these vessels leaky. Current treatments for age-related macular degeneration and diabetic retinopathy decrease swelling in the eye by blocking the action of VEGF. However, these treatments also cause existing blood vessels and nerve cells to die, leading to irreversible damage. Now, Smith et al. have set out to find whether the effects of VEGF can be blocked without causing further damage to existing cells. To investigate this possibility, the eyes and retinas of mice were treated with a laser or exposed to changing oxygen levels to create injuries that resembled human age-related macular degeneration and diabetic retinopathy. Each of the tested mice had specific mutations in proteins known to interact with VEGF. Fluorescent particles were injected into the bloodstream of the mice to assess how these different mutations affected blood vessel leakage: if fluorescent particles could no longer be detected outside the blood vessels, this suggested that the mutation had stopped the vessels from leaking. Further experiments showed these specific mutations affected leakage and did not prevent new blood vessels from forming. In the future it will be important to see if drugs, rather than mutations, can also decrease the leakiness of blood vessels in the retina. Such chemical compounds could then be tested in mouse experiments. If successful, these drugs might be used to treat patients with age-related macular degeneration and diabetic retinopathy.
Collapse
Affiliation(s)
- Ross O Smith
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Science for Life Laboratory Uppsala University, Uppsala, Sweden
| | - Takeshi Ninchoji
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Science for Life Laboratory Uppsala University, Uppsala, Sweden
| | - Emma Gordon
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Science for Life Laboratory Uppsala University, Uppsala, Sweden
| | - Helder André
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Elisabetta Dejana
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Science for Life Laboratory Uppsala University, Uppsala, Sweden.,IFOM-IEO Campus Via Adamello, Milan, Italy
| | | | - Anders Kvanta
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Lena Claesson-Welsh
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Science for Life Laboratory Uppsala University, Uppsala, Sweden
| |
Collapse
|
25
|
Boeck M, Thien A, Wolf J, Hagemeyer N, Laich Y, Yusuf D, Backofen R, Zhang P, Boneva S, Stahl A, Hilgendorf I, Agostini H, Prinz M, Wieghofer P, Schlunck G, Schlecht A, Lange C. Temporospatial distribution and transcriptional profile of retinal microglia in the oxygen‐induced retinopathy mouse model. Glia 2020; 68:1859-1873. [DOI: 10.1002/glia.23810] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 02/12/2020] [Accepted: 02/19/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Myriam Boeck
- Eye Center, Medical Center, Faculty of MedicineUniversity of Freiburg Freiburg im Breisgau Germany
| | - Adrian Thien
- Eye Center, Medical Center, Faculty of MedicineUniversity of Freiburg Freiburg im Breisgau Germany
| | - Julian Wolf
- Eye Center, Medical Center, Faculty of MedicineUniversity of Freiburg Freiburg im Breisgau Germany
| | - Nora Hagemeyer
- Institute of Neuropathology, Faculty of MedicineUniversity of Freiburg Freiburg im Breisgau Germany
| | - Yannik Laich
- Eye Center, Medical Center, Faculty of MedicineUniversity of Freiburg Freiburg im Breisgau Germany
| | - Dilmurat Yusuf
- Department of BioinformaticsUniversity of Freiburg Freiburg im Breisgau Germany
| | - Rolf Backofen
- Department of BioinformaticsUniversity of Freiburg Freiburg im Breisgau Germany
| | - Peipei Zhang
- Eye Center, Medical Center, Faculty of MedicineUniversity of Freiburg Freiburg im Breisgau Germany
| | - Stefaniya Boneva
- Eye Center, Medical Center, Faculty of MedicineUniversity of Freiburg Freiburg im Breisgau Germany
| | - Andreas Stahl
- Department of OphthalmologyUniversity Medical Center Greifswald Greifswald Germany
| | - Ingo Hilgendorf
- Cardiology and AngiologyUniversity Heart Center, University of Freiburg Freiburg im Breisgau Germany
| | - Hansjürgen Agostini
- Eye Center, Medical Center, Faculty of MedicineUniversity of Freiburg Freiburg im Breisgau Germany
| | - Marco Prinz
- Institute of Neuropathology, Faculty of MedicineUniversity of Freiburg Freiburg im Breisgau Germany
- Signalling Research Centres BIOSS and CIBSSUniversity of Freiburg Freiburg im Breisgau Germany
- Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of MedicineUniversity of Freiburg Freiburg im Breisgau Germany
| | - Peter Wieghofer
- Institute of AnatomyUniversity of Leipzig Freiburg im Breisgau Germany
| | - Günther Schlunck
- Eye Center, Medical Center, Faculty of MedicineUniversity of Freiburg Freiburg im Breisgau Germany
| | - Anja Schlecht
- Eye Center, Medical Center, Faculty of MedicineUniversity of Freiburg Freiburg im Breisgau Germany
| | - Clemens Lange
- Eye Center, Medical Center, Faculty of MedicineUniversity of Freiburg Freiburg im Breisgau Germany
| |
Collapse
|
26
|
Dalvin LA, Hartnett ME, Bretz CA, Hann CR, Cui RZ, Marmorstein AD, Sheikh-Hamad D, Fautsch MP, Roddy GW. Stanniocalcin-1 is a Modifier of Oxygen-Induced Retinopathy Severity. Curr Eye Res 2020; 45:46-51. [PMID: 31314602 PMCID: PMC6898771 DOI: 10.1080/02713683.2019.1645184] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 07/08/2019] [Accepted: 07/10/2019] [Indexed: 01/21/2023]
Abstract
Purpose/Aim: Abnormal activation of signaling pathways related to angiogenesis, inflammation, and oxidative stress has been implicated in the pathophysiology of retinopathy of prematurity (ROP), a leading cause of blindness in pre-term infants. Therapies for ROP include laser and anti-vascular endothelial growth factor agents. However, these therapies have side effects, and even with adequate treatment, visual acuity can be impaired. Novel therapeutic options are needed. Stanniocalcin-1 (STC-1) is a neuroprotective protein with anti-inflammatory and anti-oxidative stress properties. Rodent models of oxygen-induced retinopathy (OIR) were selected to determine whether STC-1 plays a role in the development of OIR.Materials and methods: STC-1 gene and protein expression was first evaluated in the Sprague Dawley rat OIR model that is most similar to human ROP. OIR was then induced in wild-type and Stc-1-/- mice. Retinas were isolated and evaluated for avascular and neovascular area on retinal flat mounts. Quantification of gene expression by quantitative real-time PCR was performed. VEGF was assayed by ELISA in media obtained from induced pluripotent stem-cell-derived retinal pigment epithelial (iPS-RPE) cells following treatment with recombinant STC-1.Results: STC-1 was significantly upregulated in a rat model of OIR compared to room air controls at the gene (P < .05) and protein (P < .001) level. Stc-1-/- OIR mice showed significantly worse ROP compared to wild-type mice as assessed by avascular (20.2 ± 2.4% vs 15.2 ± 2.5%; P = .02) and neovascular area (14.3 ± 2.7% vs 8.8 ± 3.7%; P < .05). Transcript levels of vascular endothelial growth factor-A were significantly higher in Stc-1-/- OIR mice compared to wild-type controls (P = .03). STC-1 reduced VEGF production in iPS-RPE cells (P = .01).Conclusions: STC-1 plays a role in the OIR stress response and development of pathologic vascular features in rodent OIR models by regulating VEGF levels.
Collapse
Affiliation(s)
| | | | | | - Cheryl R. Hann
- Department of Ophthalmology, Mayo Clinic, Rochester, MN,
55905
| | - Ricky Z Cui
- West Suburban Medical Center, Oak Park, IL 60302
| | | | - David Sheikh-Hamad
- Department of Nephrology, Baylor College of Medicine,
Houston, TX, 77030
| | | | - Gavin W. Roddy
- Department of Ophthalmology, Mayo Clinic, Rochester, MN,
55905
| |
Collapse
|
27
|
Bats ML, Bougaran P, Peghaire C, Gueniot F, Abelanet A, Chan H, Séguy C, Jeanningros S, Jaspard-Vinassa B, Couffinhal T, Duplàa C, Dufourcq P. Therapies targeting Frizzled-7/β-catenin pathway prevent the development of pathological angiogenesis in an ischemic retinopathy model. FASEB J 2019; 34:1288-1303. [PMID: 31914666 DOI: 10.1096/fj.201901886r] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Revised: 10/28/2019] [Accepted: 11/11/2019] [Indexed: 12/24/2022]
Abstract
Retinopathies remain major causes of visual impairment in diabetic patients and premature infants. Introduction of anti-angiogenic drugs targeting vascular endothelial growth factor (VEGF) has transformed therapy for these proliferative retinopathies. However, limitations associated with anti-VEGF medications require to unravel new pathways of vessel growth to identify potential drug targets. Here, we investigated the role of Wnt/Frizzled-7 (Fzd7) pathway in a mouse model of oxygen-induced retinopathy (OIR). Using transgenic mice, which enabled endothelium-specific and time-specific Fzd7 deletion, we demonstrated that Fzd7 controls both vaso-obliteration and neovascular phases (NV). Deletion of Fzd7 at P12, after the ischemic phase of OIR, prevented formation of aberrant neovessels into the vitreous by suppressing proliferation of endothelial cells (EC) in tufts. Next we validated in vitro two Frd7 blocking strategies: a monoclonal antibody (mAbFzd7) against Fzd7 and a soluble Fzd7 receptor (CRD). In vivo a single intravitreal microinjection of mAbFzd7 or CRD significantly attenuated retinal neovascularization (NV) in mice with OIR. Molecular analysis revealed that Fzd7 may act through the activation of Wnt/β-catenin and Jagged1 expression to control EC proliferation in extra-retinal neovessels. We identified Fzd7/β-catenin signaling as new regulator of pathological retinal NV. Fzd7 appears to be a potent pharmacological target to prevent or treat aberrant angiogenesis of ischemic retinopathies.
Collapse
Affiliation(s)
- Marie-Lise Bats
- Biology of Cardiovascular Diseases, Inserm U1034, Pessac, France.,Biology of Cardiovascular Diseases, University of Bordeaux U1034, Bordeaux, France.,Service de Biochimie clinique, CHU de Bordeaux, Bordeaux, France
| | - Pauline Bougaran
- Biology of Cardiovascular Diseases, Inserm U1034, Pessac, France.,Biology of Cardiovascular Diseases, University of Bordeaux U1034, Bordeaux, France
| | - Claire Peghaire
- Biology of Cardiovascular Diseases, Inserm U1034, Pessac, France.,NHLI-Vascular Science, Imperial College London, London, UK
| | - Florian Gueniot
- Biology of Cardiovascular Diseases, Inserm U1034, Pessac, France.,Biology of Cardiovascular Diseases, University of Bordeaux U1034, Bordeaux, France
| | - Alice Abelanet
- Biology of Cardiovascular Diseases, Inserm U1034, Pessac, France.,Biology of Cardiovascular Diseases, University of Bordeaux U1034, Bordeaux, France
| | - Hélène Chan
- Biology of Cardiovascular Diseases, Inserm U1034, Pessac, France
| | - Camille Séguy
- Biology of Cardiovascular Diseases, Inserm U1034, Pessac, France
| | | | - Béatrice Jaspard-Vinassa
- Biology of Cardiovascular Diseases, Inserm U1034, Pessac, France.,Biology of Cardiovascular Diseases, University of Bordeaux U1034, Bordeaux, France
| | - Thierry Couffinhal
- Biology of Cardiovascular Diseases, Inserm U1034, Pessac, France.,Biology of Cardiovascular Diseases, University of Bordeaux U1034, Bordeaux, France.,Service des Maladies cardiaques et vasculaires, CHU de Bordeaux, Bordeaux, France
| | - Cécile Duplàa
- Biology of Cardiovascular Diseases, Inserm U1034, Pessac, France.,Biology of Cardiovascular Diseases, University of Bordeaux U1034, Bordeaux, France
| | - Pascale Dufourcq
- Biology of Cardiovascular Diseases, Inserm U1034, Pessac, France.,Biology of Cardiovascular Diseases, University of Bordeaux U1034, Bordeaux, France
| |
Collapse
|
28
|
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: 1.4] [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]
|
29
|
Park B, Corson TW. Soluble Epoxide Hydrolase Inhibition for Ocular Diseases: Vision for the Future. Front Pharmacol 2019; 10:95. [PMID: 30792659 PMCID: PMC6374558 DOI: 10.3389/fphar.2019.00095] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 01/24/2019] [Indexed: 12/16/2022] Open
Abstract
Ocular diseases cause visual impairment and blindness, imposing a devastating impact on quality of life and a substantial societal economic burden. Many such diseases lack universally effective pharmacotherapies. Therefore, understanding the mediators involved in their pathophysiology is necessary for the development of therapeutic strategies. To this end, the hydrolase activity of soluble epoxide hydrolase (sEH) has been explored in the context of several eye diseases, due to its implications in vascular diseases through metabolism of bioactive epoxygenated fatty acids. In this mini-review, we discuss the mounting evidence associating sEH with ocular diseases and its therapeutic value as a target. Substantial data link sEH with the retinal and choroidal neovascularization underlying diseases such as wet age-related macular degeneration, retinopathy of prematurity, and proliferative diabetic retinopathy, although some conflicting results pose challenges for the synthesis of a common mechanism. sEH also shows therapeutic relevance in non-proliferative diabetic retinopathy and diabetic keratopathy, and sEH inhibition has been tested in a uveitis model. Various approaches have been implemented to assess sEH function in the eye, including expression analyses, genetic manipulation, pharmacological targeting of sEH, and modulation of certain lipid metabolites that are upstream and downstream of sEH. On balance, sEH inhibition shows considerable promise for treating multiple eye diseases. The possibility of local delivery of inhibitors makes the eye an appealing target for future sEH drug development initiatives.
Collapse
Affiliation(s)
- Bomina Park
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine, Indianapolis, IN, United States.,Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Timothy W Corson
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine, Indianapolis, IN, United States.,Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, United States.,Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
30
|
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: 1.6] [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
|
31
|
Abstract
Ischemic retinal diseases can be caused by various pathologies, which often lead to formation of preretinal neovascularization. A very common and well-established model to study normal as well as pathological angiogenic mechanisms in retina is the oxygen-induced retinopathy model in mice. This model is based on oxygen exposure of mouse pups during retinal vascular development, leading to a depletion of retinal capillaries. Upon return to room air, the lack of retinal vasculature results in hypoxia, which in turn induces vascular repair and preretinal neovascularization. In this review, we will focus on the scientific options, practical implementation, and quantification of the OIR model and its potential pitfalls.
Collapse
Affiliation(s)
- Raffael Liegl
- Department of Ophthalmology, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Claudia Priglinger
- Department of Ophthalmology, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Andreas Ohlmann
- Department of Ophthalmology, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany.
| |
Collapse
|
32
|
Nowak-Sliwinska P, Alitalo K, Allen E, Anisimov A, Aplin AC, Auerbach R, Augustin HG, Bates DO, van Beijnum JR, Bender RHF, Bergers G, Bikfalvi A, Bischoff J, Böck BC, Brooks PC, Bussolino F, Cakir B, Carmeliet P, Castranova D, Cimpean AM, Cleaver O, Coukos G, Davis GE, De Palma M, Dimberg A, Dings RPM, Djonov V, Dudley AC, Dufton NP, Fendt SM, Ferrara N, Fruttiger M, Fukumura D, Ghesquière B, Gong Y, Griffin RJ, Harris AL, Hughes CCW, Hultgren NW, Iruela-Arispe ML, Irving M, Jain RK, Kalluri R, Kalucka J, Kerbel RS, Kitajewski J, Klaassen I, Kleinmann HK, Koolwijk P, Kuczynski E, Kwak BR, Marien K, Melero-Martin JM, Munn LL, Nicosia RF, Noel A, Nurro J, Olsson AK, Petrova TV, Pietras K, Pili R, Pollard JW, Post MJ, Quax PHA, Rabinovich GA, Raica M, Randi AM, Ribatti D, Ruegg C, Schlingemann RO, Schulte-Merker S, Smith LEH, Song JW, Stacker SA, Stalin J, Stratman AN, Van de Velde M, van Hinsbergh VWM, Vermeulen PB, Waltenberger J, Weinstein BM, Xin H, Yetkin-Arik B, Yla-Herttuala S, Yoder MC, Griffioen AW. Consensus guidelines for the use and interpretation of angiogenesis assays. Angiogenesis 2018; 21:425-532. [PMID: 29766399 PMCID: PMC6237663 DOI: 10.1007/s10456-018-9613-x] [Citation(s) in RCA: 419] [Impact Index Per Article: 69.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The formation of new blood vessels, or angiogenesis, is a complex process that plays important roles in growth and development, tissue and organ regeneration, as well as numerous pathological conditions. Angiogenesis undergoes multiple discrete steps that can be individually evaluated and quantified by a large number of bioassays. These independent assessments hold advantages but also have limitations. This article describes in vivo, ex vivo, and in vitro bioassays that are available for the evaluation of angiogenesis and highlights critical aspects that are relevant for their execution and proper interpretation. As such, this collaborative work is the first edition of consensus guidelines on angiogenesis bioassays to serve for current and future reference.
Collapse
Affiliation(s)
- Patrycja Nowak-Sliwinska
- Molecular Pharmacology Group, School of Pharmaceutical Sciences, Faculty of Sciences, University of Geneva, University of Lausanne, Rue Michel-Servet 1, CMU, 1211, Geneva 4, Switzerland.
- Translational Research Center in Oncohaematology, University of Geneva, Geneva, Switzerland.
| | - Kari Alitalo
- Wihuri Research Institute and Translational Cancer Biology Program, University of Helsinki, Helsinki, Finland
| | - Elizabeth Allen
- Laboratory of Tumor Microenvironment and Therapeutic Resistance, Department of Oncology, VIB-Center for Cancer Biology, KU Leuven, Louvain, Belgium
| | - Andrey Anisimov
- Wihuri Research Institute and Translational Cancer Biology Program, University of Helsinki, Helsinki, Finland
| | - Alfred C Aplin
- Department of Pathology, University of Washington, Seattle, WA, USA
| | | | - Hellmut G Augustin
- European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
- Division of Vascular Oncology and Metastasis Research, German Cancer Research Center, Heidelberg, Germany
- German Cancer Consortium, Heidelberg, Germany
| | - David O Bates
- Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, UK
| | - Judy R van Beijnum
- Angiogenesis Laboratory, Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - R Hugh F Bender
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, USA
| | - Gabriele Bergers
- Laboratory of Tumor Microenvironment and Therapeutic Resistance, Department of Oncology, VIB-Center for Cancer Biology, KU Leuven, Louvain, Belgium
- Department of Neurological Surgery, Brain Tumor Research Center, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Andreas Bikfalvi
- Angiogenesis and Tumor Microenvironment Laboratory (INSERM U1029), University Bordeaux, Pessac, France
| | - Joyce Bischoff
- Vascular Biology Program and Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Barbara C Böck
- European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
- Division of Vascular Oncology and Metastasis Research, German Cancer Research Center, Heidelberg, Germany
- German Cancer Consortium, Heidelberg, Germany
| | - Peter C Brooks
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, ME, USA
| | - Federico Bussolino
- Department of Oncology, University of Torino, Turin, Italy
- Candiolo Cancer Institute-FPO-IRCCS, 10060, Candiolo, Italy
| | - Bertan Cakir
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology and Leuven Cancer Institute (LKI), KU Leuven, Leuven, Belgium
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
| | - Daniel Castranova
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Anca M Cimpean
- Department of Microscopic Morphology/Histology, Angiogenesis Research Center, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania
| | - Ondine Cleaver
- Department of Molecular Biology, Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - George Coukos
- Ludwig Institute for Cancer Research, Department of Oncology, University of Lausanne, Lausanne, Switzerland
| | - George E Davis
- Department of Medical Pharmacology and Physiology, University of Missouri, School of Medicine and Dalton Cardiovascular Center, Columbia, MO, USA
| | - Michele De Palma
- School of Life Sciences, Swiss Federal Institute of Technology, Lausanne, Switzerland
| | - Anna Dimberg
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Ruud P M Dings
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | | | - Andrew C Dudley
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, USA
- Emily Couric Cancer Center, The University of Virginia, Charlottesville, VA, USA
| | - Neil P Dufton
- Vascular Sciences, Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, Imperial College London, London, UK
| | - Sarah-Maria Fendt
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB Center for Cancer Biology, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute, Leuven, Belgium
| | | | - Marcus Fruttiger
- Institute of Ophthalmology, University College London, London, UK
| | - Dai Fukumura
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Bart Ghesquière
- Metabolomics Expertise Center, VIB Center for Cancer Biology, VIB, Leuven, Belgium
- Department of Oncology, Metabolomics Expertise Center, KU Leuven, Leuven, Belgium
| | - Yan Gong
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Robert J Griffin
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Adrian L Harris
- Molecular Oncology Laboratories, Oxford University Department of Oncology, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford, UK
| | - Christopher C W Hughes
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, USA
| | - Nan W Hultgren
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, USA
| | | | - Melita Irving
- Ludwig Institute for Cancer Research, Department of Oncology, University of Lausanne, Lausanne, Switzerland
| | - Rakesh K Jain
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Raghu Kalluri
- Department of Cancer Biology, Metastasis Research Center, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Joanna Kalucka
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology and Leuven Cancer Institute (LKI), KU Leuven, Leuven, Belgium
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
| | - Robert S Kerbel
- Department of Medical Biophysics, Biological Sciences Platform, Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| | - Jan Kitajewski
- Department of Physiology and Biophysics, University of Illinois, Chicago, IL, USA
| | - Ingeborg Klaassen
- Ocular Angiogenesis Group, Departments of Ophthalmology and Medical Biology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Hynda K Kleinmann
- The George Washington University School of Medicine, Washington, DC, USA
| | - Pieter Koolwijk
- Department of Ophthalmology, University of Lausanne, Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, Lausanne, Switzerland
| | - Elisabeth Kuczynski
- Department of Medical Biophysics, Biological Sciences Platform, Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| | - Brenda R Kwak
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | | | - Juan M Melero-Martin
- Department of Cardiac Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Lance L Munn
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Roberto F Nicosia
- Department of Pathology, University of Washington, Seattle, WA, USA
- Pathology and Laboratory Medicine Service, VA Puget Sound Health Care System, Seattle, WA, USA
| | - Agnes Noel
- Laboratory of Tumor and Developmental Biology, GIGA-Cancer, University of Liège, Liège, Belgium
| | - Jussi Nurro
- Department of Biotechnology and Molecular Medicine, University of Eastern Finland, Kuopio, Finland
| | - Anna-Karin Olsson
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala Biomedical Center, Uppsala University, Uppsala, Sweden
| | - Tatiana V Petrova
- Department of oncology UNIL-CHUV, Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
| | - Kristian Pietras
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund, Sweden
| | - Roberto Pili
- Genitourinary Program, Indiana University-Simon Cancer Center, Indianapolis, IN, USA
| | - Jeffrey W Pollard
- Medical Research Council Centre for Reproductive Health, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, UK
| | - Mark J Post
- Department of Physiology, Maastricht University, Maastricht, The Netherlands
| | - Paul H A Quax
- Einthoven Laboratory for Experimental Vascular Medicine, Department Surgery, LUMC, Leiden, The Netherlands
| | - Gabriel A Rabinovich
- Laboratory of Immunopathology, Institute of Biology and Experimental Medicine, National Council of Scientific and Technical Investigations (CONICET), Buenos Aires, Argentina
| | - Marius Raica
- Department of Microscopic Morphology/Histology, Angiogenesis Research Center, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania
| | - Anna M Randi
- Vascular Sciences, Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, Imperial College London, London, UK
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy
- National Cancer Institute "Giovanni Paolo II", Bari, Italy
| | - Curzio Ruegg
- Department of Oncology, Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Reinier O Schlingemann
- Ocular Angiogenesis Group, Departments of Ophthalmology and Medical Biology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Department of Ophthalmology, University of Lausanne, Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, Lausanne, Switzerland
| | - Stefan Schulte-Merker
- Institute of Cardiovascular Organogenesis and Regeneration, Faculty of Medicine, WWU, Münster, Germany
| | - Lois E H Smith
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Jonathan W Song
- Department of Mechanical and Aerospace Engineering, The Ohio State University, Columbus, OH, USA
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Steven A Stacker
- Tumour Angiogenesis and Microenvironment Program, Peter MacCallum Cancer Centre and The Sir Peter MacCallum, Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Jimmy Stalin
- Institute of Cardiovascular Organogenesis and Regeneration, Faculty of Medicine, WWU, Münster, Germany
| | - Amber N Stratman
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Maureen Van de Velde
- Laboratory of Tumor and Developmental Biology, GIGA-Cancer, University of Liège, Liège, Belgium
| | - Victor W M van Hinsbergh
- Department of Ophthalmology, University of Lausanne, Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, Lausanne, Switzerland
| | - Peter B Vermeulen
- HistoGeneX, Antwerp, Belgium
- Translational Cancer Research Unit, GZA Hospitals, Sint-Augustinus & University of Antwerp, Antwerp, Belgium
| | - Johannes Waltenberger
- Medical Faculty, University of Münster, Albert-Schweitzer-Campus 1, Münster, Germany
| | - Brant M Weinstein
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Hong Xin
- University of California, San Diego, La Jolla, CA, USA
| | - Bahar Yetkin-Arik
- Ocular Angiogenesis Group, Departments of Ophthalmology and Medical Biology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Seppo Yla-Herttuala
- Department of Biotechnology and Molecular Medicine, University of Eastern Finland, Kuopio, Finland
| | - Mervin C Yoder
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Arjan W Griffioen
- Angiogenesis Laboratory, Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.
| |
Collapse
|
33
|
Zhang HB, Wang XD, Xu K, Li XG. The progress of prophylactic treatment in retinopathy of prematurity. Int J Ophthalmol 2018; 11:858-873. [PMID: 29862189 DOI: 10.18240/ijo.2018.05.24] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 09/28/2017] [Indexed: 12/19/2022] Open
Abstract
Retinopathy of prematurity (ROP) is a retinal vascular disorder frequently found in premature infants. Different therapeutic strategies have been developed to treat ROP. However, there are still many children with ROP suffering by severe limitations in vision or even blindness. Recently, ROP has been suggested to be caused by abnormal development of the retinal vasculature, but not simply resulted by retinal neovascularization which takes about 4 to 6wk after birth in premature infants. Thus, instead of focusing on how to reduce retinal neovascularization, understanding the pathological changes and mechanisms that occur prior to retinal neovascularization is meaningful, which may lead to identify novel target(s) for the development of novel strategy to promote the healthy growth of retinal blood vessels rather than passively waiting for the appearance of retinal neovascularization and removing it by force. In this review, we discussed recent studies about, 1) the pathogenesis prior to retinal neovascularization in oxygen-induced retinopathy (OIR; a ROP in animal model) and in premature infants with ROP; 2) the preclinical and clinical research on preventive treatment of early OIR and ROP. We will not only highlight the importance of the mechanisms and signalling pathways in regulating early stage of ROP but also will provide guidance for actively exploring novel mechanisms and discovering novel treatments for early phase OIR and ROP prior to retinal neovascularization in the future.
Collapse
Affiliation(s)
- Hong-Bing Zhang
- Eye Institute of Shaanxi Province; Xi'an First Hospital, Xi'an 710002, Shaanxi Province, China
| | - Xiao-Dong Wang
- Eye Institute of Shaanxi Province; Xi'an First Hospital, Xi'an 710002, Shaanxi Province, China
| | - Kun Xu
- Eye Institute of Shaanxi Province; Xi'an First Hospital, Xi'an 710002, Shaanxi Province, China
| | - Xiao-Gang Li
- Department of Internal Medicine; Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
34
|
Yang C, Tahiri H, Cai C, Gu M, Gagnon C, Hardy P. microRNA-181a inhibits ocular neovascularization by interfering with vascular endothelial growth factor expression. Cardiovasc Ther 2018; 36:e12329. [PMID: 29608244 DOI: 10.1111/1755-5922.12329] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 03/13/2018] [Accepted: 03/26/2018] [Indexed: 12/24/2022] Open
Abstract
AIM Excess angiogenesis or neovascularization plays a key role in the pathophysiology of several ocular diseases such as retinopathy of prematurity, diabetic retinopathy, and exudative age-related macular degeneration. microRNA-181a (miR-181a) was found highly expressed in retina and choroidal tissues. This study intends to investigate the role of miR-181a in the regulation of ocular neovascularization in different pathophysiological conditions. METHOD We performed the RNA sequence to identify the microRNAs components of anti-angiogenic lymphocyte-derived microparticles (LMPs). The effect of miR-181a on human retinal endothelial cells proliferation was assessed in vitro. The impact of miR-181a on angiogenesis was confirmed using in vitro angiogenesis assay, ex vivo choroidal explant, and in vivo retinal neovascularization. The expression of major angiogenic factors was assessed by real-time qPCR. RESULTS RNA sequence revealed that miR-181a is selectively enriched in LMPs. Importantly, the inhibition of miR-181a significantly abrogated the effect of LMPs on endothelial viability, but overexpression of miR-181a reduced endothelial cell viability in a dose-dependent manner. miR-181a strongly inhibited in vitro angiogenesis and ex vivo choroidal neovascularization. The strong anti-angiogenic effect of miR-181a was also displayed on the retinal neovascularization of the in vivo mouse model of oxygen-induced retinopathy. In keeping with its effect, several angiogenesis-related genes were dysregulated in the miR-181a overexpressed endothelial cells. CONCLUSION These data may open unexpected avenues for the development of miR-181a as a novel therapeutic strategy that would be particularly useful and relevant for the treatment of neovascular diseases.
Collapse
Affiliation(s)
- Chun Yang
- Departments of Pediatrics, Physiology and Pharmacology, University of Montreal, Montreal, QC, Canada
| | - Houda Tahiri
- Departments of Pediatrics, Physiology and Pharmacology, University of Montreal, Montreal, QC, Canada
| | - Chenrongrong Cai
- Departments of Pediatrics, Physiology and Pharmacology, University of Montreal, Montreal, QC, Canada
| | - Muqing Gu
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Carmen Gagnon
- Departments of Pediatrics, Physiology and Pharmacology, University of Montreal, Montreal, QC, Canada
| | - Pierre Hardy
- Departments of Pediatrics, Physiology and Pharmacology, University of Montreal, Montreal, QC, Canada
| |
Collapse
|
35
|
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: 32] [Impact Index Per Article: 4.6] [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
|
36
|
Oubaha M, Miloudi K, Dejda A, Guber V, Mawambo G, Germain MA, Bourdel G, Popovic N, Rezende FA, Kaufman RJ, Mallette FA, Sapieha P. Senescence-associated secretory phenotype contributes to pathological angiogenesis in retinopathy. Sci Transl Med 2017; 8:362ra144. [PMID: 27797960 DOI: 10.1126/scitranslmed.aaf9440] [Citation(s) in RCA: 174] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Accepted: 06/17/2016] [Indexed: 12/13/2022]
Abstract
Pathological angiogenesis is the hallmark of diseases such as cancer and retinopathies. Although tissue hypoxia and inflammation are recognized as central drivers of vessel growth, relatively little is known about the process that bridges the two. In a mouse model of ischemic retinopathy, we found that hypoxic regions of the retina showed only modest rates of apoptosis despite severely compromised metabolic supply. Using transcriptomic analysis and inducible loss-of-function genetics, we demonstrated that ischemic retinal cells instead engage the endoplasmic reticulum stress inositol-requiring enzyme 1α (IRE1α) pathway that, through its endoribonuclease activity, induces a state of senescence in which cells adopt a senescence-associated secretory phenotype (SASP). We also detected SASP-associated cytokines (plasminogen activator inhibitor 1, interleukin-6, interleukin-8, and vascular endothelial growth factor) in the vitreous humor of patients suffering from proliferative diabetic retinopathy. Therapeutic inhibition of the SASP through intravitreal delivery of metformin or interference with effectors of senescence (semaphorin 3A or IRE1α) in mice reduced destructive retinal neovascularization in vivo. We conclude that the SASP contributes to pathological vessel growth, with ischemic retinal cells becoming prematurely senescent and secreting inflammatory cytokines that drive paracrine senescence, exacerbate destructive angiogenesis, and hinder reparative vascular regeneration. Reversal of this process may be therapeutically beneficial.
Collapse
Affiliation(s)
- Malika Oubaha
- Department of Biochemistry, Maisonneuve-Rosemont Hospital Research Centre, Université de Montréal, Montreal, Quebec H1T 2M4, Canada.,Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec H3A 2B4 Canada
| | - Khalil Miloudi
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec H3A 2B4 Canada
| | - Agnieszka Dejda
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, Université de Montréal, Montreal, Quebec H1T 2M4, Canada
| | - Vera Guber
- Department of Biochemistry, Maisonneuve-Rosemont Hospital Research Centre, Université de Montréal, Montreal, Quebec H1T 2M4, Canada
| | - Gaëlle Mawambo
- Department of Biochemistry, Maisonneuve-Rosemont Hospital Research Centre, Université de Montréal, Montreal, Quebec H1T 2M4, Canada
| | - Marie-Anne Germain
- Department of Biochemistry, Maisonneuve-Rosemont Hospital Research Centre, Université de Montréal, Montreal, Quebec H1T 2M4, Canada.,Department of Medicine, Maisonneuve-Rosemont Hospital Research Centre, Université de Montréal, Montreal, Quebec H1T 2M4, Canada
| | - Guillaume Bourdel
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, Université de Montréal, Montreal, Quebec H1T 2M4, Canada
| | - Natalija Popovic
- Department of Biochemistry, Maisonneuve-Rosemont Hospital Research Centre, Université de Montréal, Montreal, Quebec H1T 2M4, Canada
| | - Flavio A Rezende
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, Université de Montréal, Montreal, Quebec H1T 2M4, Canada
| | - Randal J Kaufman
- Degenerative Diseases Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Frédérick A Mallette
- Department of Biochemistry, Maisonneuve-Rosemont Hospital Research Centre, Université de Montréal, Montreal, Quebec H1T 2M4, Canada. .,Department of Medicine, Maisonneuve-Rosemont Hospital Research Centre, Université de Montréal, Montreal, Quebec H1T 2M4, Canada
| | - Przemyslaw Sapieha
- Department of Biochemistry, Maisonneuve-Rosemont Hospital Research Centre, Université de Montréal, Montreal, Quebec H1T 2M4, Canada. .,Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec H3A 2B4 Canada.,Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, Université de Montréal, Montreal, Quebec H1T 2M4, Canada
| |
Collapse
|
37
|
Retinal vasculature development in health and disease. Prog Retin Eye Res 2017; 63:1-19. [PMID: 29129724 DOI: 10.1016/j.preteyeres.2017.11.001] [Citation(s) in RCA: 196] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 11/02/2017] [Accepted: 11/06/2017] [Indexed: 12/17/2022]
Abstract
Development of the retinal vasculature is based on highly coordinated signalling between different cell types of the retina, integrating internal metabolic requirements with external influences such as the supply of oxygen and nutrients. The developing mouse retinal vasculature is a useful model system to study these interactions because it is experimentally accessible for intra ocular injections and genetic manipulations, can be easily imaged and develops in a similar fashion to that of humans. Research using this model has provided insights about general principles of angiogenesis as well as pathologies that affect the developing retinal vasculature. In this review, we discuss recent advances in our understanding of the molecular and cellular mechanisms that govern the interactions between neurons, glial and vascular cells in the developing retina. This includes a review of mechanisms that shape the retinal vasculature, such as sprouting angiogenesis, vascular network remodelling and vessel maturation. We also explore how the disruption of these processes in mice can lead to pathology - such as oxygen induced retinopathy - and how this translates to human retinopathy of prematurity.
Collapse
|
38
|
Vähätupa M, Cordova ZM, Barker H, Aittomäki S, Uusitalo H, Järvinen TAH, Pesu M, Uusitalo-Järvinen H. Furin deficiency in myeloid cells leads to attenuated revascularization in a mouse-model of oxygen-induced retinopathy. Exp Eye Res 2017; 166:160-167. [PMID: 29031855 DOI: 10.1016/j.exer.2017.10.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 09/01/2017] [Accepted: 10/11/2017] [Indexed: 12/12/2022]
Abstract
Ischemic retinopathy is a vision-threatening disease associated with chronic retinal inflammation and hypoxia leading to abnormal angiogenesis. Furin, a member of the proprotein convertase family of proteins, has been implicated in the regulation of angiogenesis due to its essential role in the activation of several angiogenic growth factors, including vascular endothelial growth factor-C (VEGF-C), VEGF-D and transforming growth factor - β (TGF- β). In the present study, we evaluated expression of furin in the retina and its role in retinal angiogenesis. As both inflammation and hypoxia contribute to angiogenesis, the role of furin was evaluated using myeloid-cell specific furin knockout (KO) mice (designated LysMCre-fur(fl/fl)) both in developmental retinal angiogenesis as well as in hypoxia-driven angiogenesis using the oxygen-induced retinopathy (OIR) model. In the retina, furin expression was detected in endothelial cells, macrophages and, to some extent, in neurons. The rate of angiogenesis was not different in LysMCre-fur(fl/fl) mice when compared to their wild-type littermates during development. In the OIR model, the revascularization of retina was significantly delayed in LysMCre-fur(fl/fl) mice compared to their wild-type littermates, while there was no compensatory increase in the preretinal neovascularization in LysMCre-fur(fl/fl) mice. These results demonstrate that furin expression in myeloid cells plays a significant role in hypoxia-induced angiogenesis in retina.
Collapse
Affiliation(s)
- Maria Vähätupa
- Faculty of Medicine & Life Sciences, University of Tampere, Tampere, Finland
| | - Zuzet Martinez Cordova
- Faculty of Medicine & Life Sciences, University of Tampere, Tampere, Finland; Immunoregulation, Institute of Biosciences and Medical Technology (BioMediTech), University of Tampere, Tampere, Finland
| | - Harlan Barker
- Faculty of Medicine & Life Sciences, University of Tampere, Tampere, Finland
| | - Saara Aittomäki
- Faculty of Medicine & Life Sciences, University of Tampere, Tampere, Finland; Immunoregulation, Institute of Biosciences and Medical Technology (BioMediTech), University of Tampere, Tampere, Finland
| | - Hannu Uusitalo
- Faculty of Medicine & Life Sciences, University of Tampere, Tampere, Finland; Eye Centre, Tampere University Hospital, Tampere, Finland
| | - Tero A H Järvinen
- Faculty of Medicine & Life Sciences, University of Tampere, Tampere, Finland; Departments of Musculoskeletal Disorders, Tampere University Hospital, Tampere, Finland
| | - Marko Pesu
- Faculty of Medicine & Life Sciences, University of Tampere, Tampere, Finland; Immunoregulation, Institute of Biosciences and Medical Technology (BioMediTech), University of Tampere, Tampere, Finland; Departments of Dermatology, Tampere University Hospital, Tampere, Finland
| | - Hannele Uusitalo-Järvinen
- Faculty of Medicine & Life Sciences, University of Tampere, Tampere, Finland; Eye Centre, Tampere University Hospital, Tampere, Finland.
| |
Collapse
|
39
|
Foxp3 + Tregs are recruited to the retina to repair pathological angiogenesis. Nat Commun 2017; 8:748. [PMID: 28963474 PMCID: PMC5622066 DOI: 10.1038/s41467-017-00751-w] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Accepted: 07/25/2017] [Indexed: 02/03/2023] Open
Abstract
Neovascular retinopathies are major causes of vision loss; yet treatments to prevent the condition are inadequate. The role of regulatory T cells in neovascular retinopathy is unknown. Here we show that in retinopathy regulatory T cells are transiently increased in lymphoid organs and the retina, but decline when neovascularization is established. The decline is prevented following regulatory T cells expansion with an IL-2/anti-IL-2 mAb complex or the adoptive transfer of regulatory T cells. Further, both approaches reduce vasculopathy (vaso-obliteration, neovascularization, vascular leakage) and alter the activation of Tmem119+ retinal microglia. Our in vitro studies complement these findings, showing that retinal microglia co-cultured with regulatory T cells exhibit a reduction in co-stimulatory molecules and pro-inflammatory mediators that is attenuated by CTLA-4 blockade. Collectively, we demonstrate that regulatory T cells are recruited to the retina and, when expanded in number, repair the vasculature. Manipulation of regulatory T cell numbers is a previously unrecognized, and promising avenue for therapies to prevent blinding neovascular retinopathies. The local immune responses in the eye are attenuated to preserve sight. Surprisingly, Deliyanti et al. show that regulatory T cells (Tregs) take an active role in protecting the eye from neovascularization in oxygen-induced retinopathy, and that interventions that augment the retinal Treg numbers reduce neovascular retinopathy in mice.
Collapse
|
40
|
Matsumoto T, Saito Y, Itokawa T, Shiba T, Oba MS, Takahashi H, Hori Y. Retinal VEGF levels correlate with ocular circulation measured by a laser speckle-micro system in an oxygen-induced retinopathy rat model. Graefes Arch Clin Exp Ophthalmol 2017; 255:1981-1990. [PMID: 28791491 DOI: 10.1007/s00417-017-3756-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 06/12/2017] [Accepted: 07/17/2017] [Indexed: 01/12/2023] Open
Abstract
PURPOSE We used a Laser speckle flowgraphy (LSFG)-micro system to examine the relationship between ocular blood flow and retinal vascular endothelial growth factor (VEGF) at retinopathy onset in oxygen-induced ischemic retinopathy (OIR) model rats. METHODS Sixteen 50/10 OIR rats were compared with 17 control rats reared in room air. In postnatal day 14 (P14) and P18 rats, we measured and analyzed the left eye's mean blur rate (MBR) by setting a rubber band on the optic nerve head center, using the LSFG-Micro. At P18, the rats were sacrificed and their left-eye retinas were fixed, flat-mounted and stained with adenosine diphosphatase (ADPase). The right-eye retinas were homogenized; the lysate was centrifuged for an enzyme-linked immunosorbent assay (ELISA). The avascular area was measured as the percentage (%AVA) of the total retinal area. Retinal VEGF was measured by an ELISA. RESULTS The examination's reproducibility was good. Our multivariate linear mixed model analysis revealed significantly high MBRs in the OIR rats (p = 0.0017). In the P18 OIR rats, significant correlations were seen between the MBR and %AVA (r = 0.80, p = 0.0002) and between the MBR and VEGF (r = 0.76, p = 0.0006). CONCLUSIONS The LSFG-Micro provided reproducible blood flow measurements in neonatal rats. Because of the vitreous blood vessels, measurement of only the retinal vessels was not possible. However, the MBR was higher in the OIR rats than in the control rats, and the MBR and %AVA were correlated, as were the MBR and retinal VEGF. The MBR may thus serve as an indicator of OIR severity.
Collapse
Affiliation(s)
- Tadashi Matsumoto
- Department of Ophthalmology, School of Medicine, Toho University, 6-11-1 Omori-Nishi, Ota-ku, Tokyo, 143-8541, Japan.
| | - Yuta Saito
- Department of Ophthalmology, School of Medicine, Showa University, Tokyo, Japan
| | - Takashi Itokawa
- Department of Ophthalmology, School of Medicine, Toho University, 6-11-1 Omori-Nishi, Ota-ku, Tokyo, 143-8541, Japan
| | - Tomoaki Shiba
- Department of Ophthalmology, School of Medicine, Toho University, 6-11-1 Omori-Nishi, Ota-ku, Tokyo, 143-8541, Japan
| | - Mari S Oba
- Department of Medical Statistics Faculty of Medicine, Toho University, Tokyo, Japan
| | - Haruo Takahashi
- Department of Ophthalmology, School of Medicine, Showa University, Tokyo, Japan
| | - Yuichi Hori
- Department of Ophthalmology, School of Medicine, Toho University, 6-11-1 Omori-Nishi, Ota-ku, Tokyo, 143-8541, Japan
| |
Collapse
|
41
|
Sun Y, Liu CH, Wang Z, Meng SS, Burnim SB, SanGiovanni JP, Kamenecka TM, Solt LA, Chen J. RORα modulates semaphorin 3E transcription and neurovascular interaction in pathological retinal angiogenesis. FASEB J 2017. [PMID: 28646017 DOI: 10.1096/fj.201700172r] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Pathological proliferation of retinal blood vessels commonly causes vision impairment in proliferative retinopathies, including retinopathy of prematurity. Dysregulated crosstalk between the vasculature and retinal neurons is increasingly recognized as a major factor contributing to the pathogenesis of vascular diseases. Class 3 semaphorins (SEMA3s), a group of neuron-secreted axonal and vascular guidance factors, suppress pathological vascular growth in retinopathy. However, the upstream transcriptional regulators that mediate the function of SEMA3s in vascular growth are poorly understood. Here we showed that retinoic acid receptor-related orphan receptor α (RORα), a nuclear receptor and transcription factor, is a novel transcriptional regulator of SEMA3E-mediated neurovascular coupling in a mouse model of oxygen-induced proliferative retinopathy. We found that genetic deficiency of RORα substantially induced Sema3e expression in retinopathy. Both RORα and SEMA3E were expressed in retinal ganglion cells. RORα directly bound to a specific ROR response element on the promoter of Sema3e and negatively regulated Sema3e promoter-driven luciferase expression. Suppression of Sema3e using adeno-associated virus 2 carrying short hairpin RNA targeting Sema3e promoted disoriented pathological neovascularization and partially abolished the inhibitory vascular effects of RORα deficiency in retinopathy. Our findings suggest that RORα is a novel transcriptional regulator of SEMA3E-mediated neurovascular coupling in pathological retinal angiogenesis.-Sun, Y., Liu, C.-H., Wang, Z., Meng, S. S., Burnim, S. B., SanGiovanni, J. P., Kamenecka, T. M., Solt, L. A., Chen, J. RORα modulates semaphorin 3E transcription and neurovascular interaction in pathological retinal angiogenesis.
Collapse
Affiliation(s)
- Ye Sun
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Chi-Hsiu Liu
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Zhongxiao Wang
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Steven S Meng
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Samuel B Burnim
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, Massachusetts, USA
| | - John Paul SanGiovanni
- Section of Nutritional Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, USA.,Department of Biochemistry and Molecular and Cellular Biology, Georgetown School of Medicine, Washington, D.C., USA
| | - Theodore M Kamenecka
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, Florida, USA
| | - Laura A Solt
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, Florida, USA
| | - Jing Chen
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, Massachusetts, USA;
| |
Collapse
|
42
|
Walz JM, Bemme S, Pielen A, Aisenbrey S, Breuß H, Alex AF, Wagenfeld L, Schiedel S, Krohne TU, Stahl A. The German ROP Registry: data from 90 infants treated for retinopathy of prematurity. Acta Ophthalmol 2016; 94:e744-e752. [PMID: 27197876 DOI: 10.1111/aos.13069] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Accepted: 02/27/2016] [Indexed: 11/30/2022]
Abstract
PURPOSE The German retinopathy of prematurity (ROP) Registry collects data on treated ROP in a multicentre approach to analyse epidemiology and treatment patterns of severe ROP. METHODS Nine centres entered data from 90 treated ROP infants (born between January 2011 and December 2013) into a central database. Analysis included incidence rate of severe ROP, demographic data, stage of ROP, treatment patterns, recurrence rates, relevant comorbidities and ophthalmological or systemic complications associated with treatment. RESULTS Treatment rate for ROP was 3.2% of the screened population. The most frequent ROP stage at time of treatment was zone II, stage 3 + (137 eyes). Treatment was bilateral in 97% of infants. Treatment patterns changed over time from 7% anti-vascular endothelial growth factor (VEGF) monotherapy in 2011 to 32% in 2014. Overall, laser treatment was the predominant treatment. However, all infants with zone I disease received anti-VEGF treatment. About 19% of infants required retreatment (16% of laser-treated and 21% of anti-VEGF treated infants). Mean time between first and second treatment was 3.8 weeks (± 11 days) for laser-treated and 10.4 weeks (± 60 days) for anti-VEGF-treated infants. CONCLUSION This study is the first multicentre analysis of severe ROP in Germany. The identified treatment patterns find laser as the most prevalent form of therapy, with an increasing use of anti-VEGF therapy over recent years. Recurrence rates were relatively high overall with slightly higher recurrence rates and later recurrence times in the anti-VEGF group. Anti-VEGF was predominantly used for high-risk stages like AP-ROP and zone I disease.
Collapse
Affiliation(s)
- Johanna M. Walz
- Eye Center; University of Freiburg; Freiburg Germany
- Department of Pharmacology and Toxicology; University of Regensburg; Regensburg Germany
| | - Sebastian Bemme
- Department of Ophthalmology; University Medical Center Göttingen; Göttingen Germany
| | - Amelie Pielen
- University Eye Hospital; Hannover Medical School; Hannover Germany
| | - Sabine Aisenbrey
- University Eye Hospital; Eberhard-Karl University Tübingen; Tübingen Germany
- University Eye Hospital; University of Oldenburg; Oldenburg Germany
| | - Helge Breuß
- Department of Ophthalmology; HELIOS Klinikum Berlin-Buch; Berlin Germany
| | - Anne F. Alex
- Department of Ophthalmology; University of Muenster Medical Center; Muenster Germany
| | - Lars Wagenfeld
- Department of Ophthalmology; University Medical Center Hamburg-Eppendorf; Hamburg Germany
| | - Susanne Schiedel
- Department of Ophthalmology; Vivantes Klinikum Neukölln; Berlin Germany
| | - Tim U. Krohne
- Department of Ophthalmology; University of Bonn; Bonn Germany
| | - Andreas Stahl
- Eye Center; University of Freiburg; Freiburg Germany
| | | |
Collapse
|
43
|
Vähätupa M, Prince S, Vataja S, Mertimo T, Kataja M, Kinnunen K, Marjomäki V, Uusitalo H, Komatsu M, Järvinen TAH, Uusitalo-Järvinen H. Lack of R-Ras Leads to Increased Vascular Permeability in Ischemic Retinopathy. Invest Ophthalmol Vis Sci 2016; 57:4898-4909. [PMID: 27654416 PMCID: PMC5032915 DOI: 10.1167/iovs.16-19212] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Purpose The role of R-Ras in retinal angiogenesis and vascular permeability was evaluated in an oxygen-induced retinopathy (OIR) model using R-Ras knockout (KO) mice and in human diabetic neovascular membranes. Methods Mice deficient for R-Ras and their wild-type (WT) littermates were subjected to 75% oxygen from postnatal day 7 (P7) to P12 and then returned to room air. At P17 retinal vascularization was examined from whole mounts, and retinal vascular permeability was studied using Miles assay. Real-time RT-PCR, Western blotting, and immunohistochemistry were used to assess the expression of R-Ras in retina during development or in the OIR model. The degree of pericyte coverage and vascular endothelial (VE)-cadherin expression on WT and R-Ras KO retinal blood vessels was quantified using confocal microscopy. The correlation of R-Ras with vascular endothelial growth factor receptor 2 (VEGFR2) and human serum albumin on human proliferative diabetic retinopathy membranes was assessed using immunohistochemistry. Results In retina, R-Ras expression was mostly restricted to the vasculature. Retinal vessels in the R-Ras KO mice were significantly more permeable than WT controls in the OIR model. A significant reduction in the direct physical contact between pericytes and blood vessel endothelium as well as reduced VE-cadherin immunostaining was found in R-Ras–deficient mice. In human proliferative diabetic retinopathy neovascular membranes, R-Ras expression negatively correlated with increased vascular leakage and expression of VEGFR2, a marker of blood vessel immaturity. Conclusions Our results suggest that R-Ras has a role in controlling retinal vessel maturation and stabilization in ischemic retinopathy and provides a potential target for pharmacologic manipulation to treat diabetic retinopathy.
Collapse
Affiliation(s)
- Maria Vähätupa
- Department of Ophthalmology, University of Tampere, Tampere, Finland 2Department of Anatomy, University of Tampere, Tampere, Finland
| | - Stuart Prince
- Department of Anatomy, University of Tampere, Tampere, Finland
| | - Suvi Vataja
- Department of Ophthalmology, University of Tampere, Tampere, Finland
| | - Teija Mertimo
- Department of Ophthalmology, University of Tampere, Tampere, Finland
| | - Marko Kataja
- Eye Centre, Tampere University Hospital, Tampere, Finland
| | - Kati Kinnunen
- Department of Ophthalmology, Kuopio University Hospital, Kuopio, Finland
| | - Varpu Marjomäki
- Department of Biological and Environmental Science/Nanoscience Center, University of Jyväskylä, Jyväskylä, Finland
| | - Hannu Uusitalo
- Department of Ophthalmology, University of Tampere, Tampere, Finland 3Eye Centre, Tampere University Hospital, Tampere, Finland
| | - Masanobu Komatsu
- Sanford Burnham Prebys Medical Discovery Institute at Lake Nona, Orlando, Florida, United States
| | - Tero A H Järvinen
- Department of Anatomy, University of Tampere, Tampere, Finland 7Department of Musculoskeletal Disorders, Tampere University Hospital, Tampere, Finland
| | - Hannele Uusitalo-Järvinen
- Department of Ophthalmology, University of Tampere, Tampere, Finland 3Eye Centre, Tampere University Hospital, Tampere, Finland
| |
Collapse
|
44
|
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
|
45
|
Abstract
More than 450,000 babies are born prematurely in the USA every year. The improved survival of even the most vulnerable low body weight preterm infants has, despite improving health outcomes, led to the resurgence in preterm complications including one of the major causes for blindness in children, retinopathy of prematurity (ROP). The current mainstay in ROP therapy is laser photocoagulation and the injection of vascular endothelial growth factor (VEGF) antibodies in the late stages of the disease after the onset of neovascularization. Both are proven options for ophthalmologists to treat the severe forms of late ROP. However, laser photocoagulation destroys major parts of the retina, and the injection of VEGF antibodies, although rather simple to administer, may cause a systemic suppression of normal vascularization, which has not been studied in sufficient depth. However, the use of neither VEGF antibody nor laser treatment prevents ROP, which should be the long-term goal. It should be possible to prevent ROP by more closely mimicking the intrauterine environment after preterm birth. Such preventive measures include preventing the toxic postbirth influences (eg, oxygen excess) as well as providing the missing intrauterine factors (eg, insulin growth factor 1) and are likely to also reduce other complications of premature birth as well as ROP. This review is meant to summarize the current knowledge on the prevention of ROP with a particular emphasize on the use of insulin growth factor 1 supplementation.
Collapse
Affiliation(s)
- Raffael Liegl
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ann Hellström
- Department of Ophthalmology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Lois Eh Smith
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
46
|
Joyal JS, Sun Y, Gantner ML, Shao Z, Evans LP, Saba N, Fredrick T, Burnim S, Kim JS, Patel G, Juan AM, Hurst CG, Hatton CJ, Cui Z, Pierce KA, Bherer P, Aguilar E, Powner MB, Vevis K, Boisvert M, Fu Z, Levy E, Fruttiger M, Packard A, Rezende FA, Maranda B, Sapieha P, Chen J, Friedlander M, Clish CB, Smith LE. Retinal lipid and glucose metabolism dictates angiogenesis through the lipid sensor Ffar1. Nat Med 2016; 22:439-45. [PMID: 26974308 PMCID: PMC4823176 DOI: 10.1038/nm.4059] [Citation(s) in RCA: 177] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2015] [Accepted: 02/05/2016] [Indexed: 02/06/2023]
Abstract
Tissues with high metabolic rates often use lipids, as well as glucose, for energy, conferring a survival advantage during feast and famine. Current dogma suggests that high-energy-consuming photoreceptors depend on glucose. Here we show that the retina also uses fatty acid β-oxidation for energy. Moreover, we identify a lipid sensor, free fatty acid receptor 1 (Ffar1), that curbs glucose uptake when fatty acids are available. Very-low-density lipoprotein receptor (Vldlr), which is present in photoreceptors and is expressed in other tissues with a high metabolic rate, facilitates the uptake of triglyceride-derived fatty acid. In the retinas of Vldlr(-/-) mice with low fatty acid uptake but high circulating lipid levels, we found that Ffar1 suppresses expression of the glucose transporter Glut1. Impaired glucose entry into photoreceptors results in a dual (lipid and glucose) fuel shortage and a reduction in the levels of the Krebs cycle intermediate α-ketoglutarate (α-KG). Low α-KG levels promotes stabilization of hypoxia-induced factor 1a (Hif1a) and secretion of vascular endothelial growth factor A (Vegfa) by starved Vldlr(-/-) photoreceptors, leading to neovascularization. The aberrant vessels in the Vldlr(-/-) retinas, which invade normally avascular photoreceptors, are reminiscent of the vascular defects in retinal angiomatous proliferation, a subset of neovascular age-related macular degeneration (AMD), which is associated with high vitreous VEGFA levels in humans. Dysregulated lipid and glucose photoreceptor energy metabolism may therefore be a driving force in macular telangiectasia, neovascular AMD and other retinal diseases.
Collapse
Affiliation(s)
- Jean-Sébastien Joyal
- Department of Pediatrics, CHU Sainte-Justine Research Center, Université de Montréal, Montreal, Qc, Ca
- Department of Pharmacology, CHU Sainte-Justine Research Center, Université de Montréal, Montreal, Qc, Ca
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Qc, Ca
| | - Ye Sun
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Qc, Ca
| | | | - Zhuo Shao
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, MA, USA
| | - Lucy P. Evans
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, MA, USA
| | - Nicholas Saba
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, MA, USA
| | - Thomas Fredrick
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, MA, USA
| | - Samuel Burnim
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, MA, USA
| | - Jin Sung Kim
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Qc, Ca
| | - Gauri Patel
- Department of Pharmacology, CHU Sainte-Justine Research Center, Université de Montréal, Montreal, Qc, Ca
| | - Aimee M. Juan
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Qc, Ca
| | - Christian G. Hurst
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, MA, USA
| | - Colman J. Hatton
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, MA, USA
| | - Zhenghao Cui
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, MA, USA
| | - Kerry A. Pierce
- Metabolite Profiling Platform, The Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Patrick Bherer
- Department of Genetics, Université de Sherbrooke, Sherbrooke, Qc, Ca
| | - Edith Aguilar
- Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, CA, USA
| | | | - Kristis Vevis
- Institute of Ophthalmology, University College London, London, UK
| | - Michel Boisvert
- Department of Nutrition, CHU Sainte-Justine Research Center, Université de Montreal, Montreal, Qc, Ca
| | - Zhongjie Fu
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, MA, USA
| | - Emile Levy
- Department of Nutrition, CHU Sainte-Justine Research Center, Université de Montreal, Montreal, Qc, Ca
| | - Marcus Fruttiger
- Institute of Ophthalmology, University College London, London, UK
| | - Alan Packard
- Radiology, Harvard Medical School, Boston Children's Hospital, MA. USA
| | - Flavio A. Rezende
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, Université de Montréal, Montreal, Qc, Ca
| | - Bruno Maranda
- Department of Genetics, Université de Sherbrooke, Sherbrooke, Qc, Ca
| | - Przemyslaw Sapieha
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, Université de Montréal, Montreal, Qc, Ca
| | - Jing Chen
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, MA, USA
| | - Martin Friedlander
- Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Clary B. Clish
- Metabolite Profiling Platform, The Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Lois E.H. Smith
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, MA, USA
| |
Collapse
|
47
|
Lack of netrin-4 modulates pathologic neovascularization in the eye. Sci Rep 2016; 6:18828. [PMID: 26732856 PMCID: PMC4702134 DOI: 10.1038/srep18828] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 11/10/2015] [Indexed: 12/17/2022] Open
Abstract
Netrins are a family of matrix-binding proteins that function as guidance signals. Netrin-4 displays pathologic roles in tumorigenesis and neovascularization. To answer the question whether netrin-4 acts either pro- or anti-angiogenic, angiogenesis in the retina was assessed in Ntn-4−/− mice with oxygen-induced retinopathy (OIR) and laser-induced choroidal neovascularization (CNV), mimicking hypoxia-mediated neovascularization and inflammatory mediated angiogenesis. The basement membrane protein netrin-4 was found to be localised to mature retinal blood vessels. Netrin-4, but not netrin-1 mRNA expression, increased in response to relative hypoxia and recovered to normal levels at the end of blood vessel formation. No changes in the retina were found in normoxic Ntn-4−/− mice. In OIR, Ntn-4−/− mice initially displayed larger avascular areas which recovered faster to revascularization. Ganzfeld electroretinography showed faster recovery of retinal function in Ntn-4−/− mice. Expression of netrin receptors, Unc5H2 (Unc-5 homolog B, C. elegans) and DCC (deleted in colorectal carcinoma), was found in Müller cells and astrocytes. Laser-induced neovascularization in Nnt-4−/− mice did not differ to that in the controls. Our results indicate a role for netrin-4 as an angiogenesis modulating factor in O2-dependent vascular homeostasis while being less important during normal retinal developmental angiogenesis or during inflammatory neovascularization.
Collapse
|
48
|
Sun Y, Ju M, Lin Z, Fredrick TW, Evans LP, Tian KT, Saba NJ, Morss PC, Pu WT, Chen J, Stahl A, Joyal JS, Smith LEH. SOCS3 in retinal neurons and glial cells suppresses VEGF signaling to prevent pathological neovascular growth. Sci Signal 2015; 8:ra94. [PMID: 26396267 DOI: 10.1126/scisignal.aaa8695] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Neurons and glial cells in the retina contribute to neovascularization, or the formation of abnormal new blood vessels, in proliferative retinopathy, a condition that can lead to vision loss or blindness. We identified a mechanism by which suppressor of cytokine signaling 3 (SOCS3) in neurons and glial cells prevents neovascularization. We found that Socs3 expression was increased in the retinal ganglion cell and inner nuclear layers after oxygen-induced retinopathy. Mice with Socs3 deficiency in neuronal and glial cells had substantially reduced vaso-obliterated retinal areas and increased pathological retinal neovascularization in response to oxygen-induced retinopathy, suggesting that loss of neuronal/glial SOCS3 increased both retinal vascular regrowth and pathological neovascularization. Furthermore, retinal expression of Vegfa (which encodes vascular endothelial growth factor A) was higher in these mice than in Socs3 flox/flox controls, indicating that neuronal and glial SOCS3 suppressed Vegfa expression during pathological conditions. Lack of neuronal and glial SOCS3 resulted in greater phosphorylation and activation of STAT3, which led to increased expression of its gene target Vegfa, and increased endothelial cell proliferation. In summary, SOCS3 in neurons and glial cells inhibited the STAT3-mediated secretion of VEGF from these cells, which suppresses endothelial cell activation, resulting in decreased endothelial cell proliferation and angiogenesis. These results suggest that neuronal and glial cell SOCS3 limits pathological retinal angiogenesis by suppressing VEGF signaling.
Collapse
Affiliation(s)
- Ye Sun
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Meihua Ju
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Zhiqiang Lin
- Department of Cardiology, Harvard Medical School, Boston Children's Hospital, Boston, MA 02115, USA
| | - Thomas W Fredrick
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Lucy P Evans
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Katherine T Tian
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Nicholas J Saba
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Peyton C Morss
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - William T Pu
- Department of Cardiology, Harvard Medical School, Boston Children's Hospital, Boston, MA 02115, USA. Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Jing Chen
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Andreas Stahl
- University Eye Hospital Freiburg, Killianstr. 5, Freiburg 79106, Germany
| | - Jean-Sébastien Joyal
- Department of Pediatrics, Centre Hospitalier Universitaire (CHU) Sainte-Justine Research Center, Université de Montréal, Montréal, Québec H3T1C4, Canada
| | - Lois E H Smith
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA.
| |
Collapse
|
49
|
Edgar KS, Matesanz N, Gardiner TA, Katusic ZS, McDonald DM. Hyperoxia depletes (6R)-5,6,7,8-tetrahydrobiopterin levels in the neonatal retina: implications for nitric oxide synthase function in retinopathy. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:1769-82. [PMID: 25913075 DOI: 10.1016/j.ajpath.2015.02.021] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Revised: 01/20/2015] [Accepted: 02/10/2015] [Indexed: 02/08/2023]
Abstract
Retinopathy of prematurity is a sight-threatening complication of premature birth caused by nitro-oxidative insult to the developing retinal vasculature during therapeutic hyperoxia exposure and later ischemia-induced neovascularization on supplemental oxygen withdrawal. In the vasodegenerative phase, during hyperoxia, defective endothelial nitric oxide synthase (NOS) produces reactive oxygen and nitrogen free radicals rather than vasoprotective nitric oxide for unclear reasons. Crucially, normal NOS function depends on availability of the cofactor (6R)-5,6,7,8-tetrahydrobiopterin (BH4). Because BH4 synthesis is controlled enzymatically by GTP cyclohydrolase (GTPCH), we used GTPCH-depleted mice [hyperphenylalaninemia strain (hph1)] to investigate the impact of hyperoxia on BH4 bioavailability and retinal vascular pathology in the neonate. Hyperoxia decreased BH4 in retinas, lungs, and aortas in all experimental groups, resulting in a dose-dependent decrease in NOS activity and, in the wild-type group, elevated NOS-derived superoxide. Retinal dopamine levels were similarly diminished, consistent with the dependence of tyrosine hydroxylase on BH4. Despite greater depletion of BH4, the hph(+/-) and hph1(-/-) groups did not show exacerbated hyperoxia-induced vessel closure, but exhibited greater vascular protection and reduced progression to neovascular disease. This vasoprotective effect was independent of enhanced circulating vascular endothelial growth factor (VEGF), which was reduced by hyperoxia, but to local retinal ganglion cell layer-derived VEGF. In conclusion, a constitutively higher level of VEGF expression associated with retinal development protects GTPCH-deficient neonates from oxygen-induced vascular damage.
Collapse
Affiliation(s)
- Kevin S Edgar
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Nuria Matesanz
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Tom A Gardiner
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Zvonimir S Katusic
- Department of Pharmacology and Anesthesiology, Mayo Clinic, Rochester, Minnesota
| | - Denise M McDonald
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom.
| |
Collapse
|
50
|
Davydov DM, Lobanov AV, Morozov SG, Gribova IE, Murashev AN. Neurodevelopment and phenotype-modulating functions of S100B protein: a pilot study. Physiol Behav 2015; 140:188-96. [PMID: 25543091 DOI: 10.1016/j.physbeh.2014.12.037] [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] [Received: 05/05/2014] [Revised: 12/21/2014] [Accepted: 12/22/2014] [Indexed: 12/13/2022]
Abstract
The importance of certain neurotrophic proteins found in maternal blood and milk for breastfed infants has remained ambiguous. This study was conducted to present evidence of the impact of an induced deficit of active S100B protein on neonate development. Newborn mice from two groups of mothers, immunized or sham-immunized against S100B, were subjected to various behavioral tests, and the development of their morphological characteristics was recorded from birth until weaning. Morphological problems, including weight gain and fur coating, a delay in the maturation of neurobehavioral systems and a deficit in neuromotor functions, including visual abilities, somato-sensory and posture reactions, muscular strength, locomotion, and fear/orienting processes, were observed in pups of immunized mothers. The S100B protein of external or internal origin in infants may be considered to be a specific factor that determines neuro- and morphological development and a risk-avoidance ('homeward-bent' or fearful) phenotype. The suppression of activity of the S100B protein results in a slower neonatal development and the formation of a risk-tolerant (fearless) phenotype of the offspring. This study thus considers the mechanism of neuroplastic regulation on the extent of sensation-seeking or risk-taking (homeless-like or fearless) and sensation- or risk-avoidance (home-bound or fearful) features in individual phenotypes.
Collapse
Affiliation(s)
- D M Davydov
- Sholokhov Moscow State University for the Humanities, The Russian Institute for Advanced Study and Institute of Neurosciences and Cognitive Research, Verkhnyaya Radishevskaya 16-18, Moscow 109240, Russia; Institute of General Pathology and Pathophysiology RAMS, Baltiyskaia ul. 8, Moscow 125315, Russia.
| | - A V Lobanov
- Branch of Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, RAS, Pushchino, Moscow Region 142290, Russia.
| | - S G Morozov
- Institute of General Pathology and Pathophysiology RAMS, Baltiyskaia ul. 8, Moscow 125315, Russia.
| | - I E Gribova
- Institute of General Pathology and Pathophysiology RAMS, Baltiyskaia ul. 8, Moscow 125315, Russia.
| | - A N Murashev
- Branch of Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, RAS, Pushchino, Moscow Region 142290, Russia.
| |
Collapse
|