1
|
Xiao R, Huang X, Gao S, Duan J, Zhang Y, Zhang M. Microglia in retinal diseases: From pathogenesis towards therapeutic strategies. Biochem Pharmacol 2024; 230:116550. [PMID: 39307318 DOI: 10.1016/j.bcp.2024.116550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 08/21/2024] [Accepted: 09/19/2024] [Indexed: 10/01/2024]
Abstract
Microglia, a widely dispersed cohort of immune cells in the retina, are intricately involved in a diverse range of pivotal biological processes, including inflammation, vascular development, complement activation, antigen presentation, and phagocytosis. Within the retinal milieu, microglia are crucial for the clearance of dead cells and cellular debris, release of anti-inflammatory agents, and orchestration of vascular network remodeling to maintain homeostasis. In addition, microglia are key mediators of neuroinflammation. Triggered by oxidative stress, elevated intraocular pressure, genetic anomalies, and immune dysregulation, microglia release numerous inflammatory cytokines, contributing to the pathogenesis of various retinal disorders. Recent studies on the ontogeny and broad functions of microglia in the retina have elucidated their characteristics during retinal development, homeostasis, and disease. Furthermore, therapeutic strategies that target microglia and their effector cytokines have been developed and shown positive results for some retinal diseases. Therefore, we systematically review the microglial ontogeny in the retina, elucidate their dual roles in retinal homeostasis and disease pathogenesis, and demonstrate microglia-based targeted therapeutic strategies for retinal diseases.
Collapse
Affiliation(s)
- Ruihan Xiao
- The Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, 610041, China; The Department of Ophthalmology and Research Laboratory of Macular Disease, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xi Huang
- The Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, 610041, China; The Department of Ophthalmology and Research Laboratory of Macular Disease, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Sheng Gao
- The Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, 610041, China; The Department of Ophthalmology and Research Laboratory of Macular Disease, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jianan Duan
- The Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, 610041, China; The Department of Ophthalmology and Research Laboratory of Macular Disease, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yun Zhang
- The Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, 610041, China; The Department of Ophthalmology and Research Laboratory of Macular Disease, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Meixia Zhang
- The Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, 610041, China; The Department of Ophthalmology and Research Laboratory of Macular Disease, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
2
|
Zheng J, Zhang W, Xu R, Liu L. The role of adiponectin and its receptor signaling in ocular inflammation-associated diseases. Biochem Biophys Res Commun 2024; 717:150041. [PMID: 38710142 DOI: 10.1016/j.bbrc.2024.150041] [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: 02/29/2024] [Revised: 04/13/2024] [Accepted: 04/29/2024] [Indexed: 05/08/2024]
Abstract
Ocular inflammation-associated diseases are leading causes of global visual impairment, with limited treatment options. Adiponectin, a hormone primarily secreted by adipose tissue, binds to its receptors, which are widely distributed throughout the body, exerting powerful physiological regulatory effects. The protective role of adiponectin in various inflammatory diseases has gained increasing attention in recent years. Previous studies have confirmed the presence of adiponectin and its receptors in the eyes. Furthermore, adiponectin and its analogs have shown potential as novel drugs for the treatment of inflammatory eye diseases. This article summarizes the evidence for the interplay between adiponectin and inflammatory eye diseases and provides new perspectives on the diagnostic and therapeutic possibilities of adiponectin.
Collapse
Affiliation(s)
- Jing Zheng
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China; Department of Optometry and Visual Science, West China Hospital, Sichuan University, Chengdu, China
| | - Wenqiu Zhang
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China; Department of Optometry and Visual Science, West China Hospital, Sichuan University, Chengdu, China
| | - Ran Xu
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China; Department of Optometry and Visual Science, West China Hospital, Sichuan University, Chengdu, China
| | - Longqian Liu
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China; Department of Optometry and Visual Science, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
3
|
Ferah Okkay I, Okkay U, Cicek B, Karatas O, Yilmaz A, Yesilyurt F, Hacimuftuoglu A. Syringic acid guards against indomethacin-induced gastric ulcer by alleviating inflammation, oxidative stress and apoptosis. Biotech Histochem 2024; 99:147-156. [PMID: 38644776 DOI: 10.1080/10520295.2024.2344477] [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] [Indexed: 04/23/2024] Open
Abstract
The purpose of this study was to evaluate the effects of syringic acid, an anti-oxidant, on indomethacin induced gastric ulcers in rats. Experimental groups were control, ulcer, ulcer treated with 20 mg/kg esomeprazole (a proton pump inhibitor that reduces acid secretion), and ulcer treated with 100 mg/kg syringic acid. Rats were pretreated with esomeprazole or syringic acid two weeks before ulcer induction. Our histopathological observations showed that either syringic acid or esomeprazole attenuated the severity of gastric mucosal damage. Moreover, syringic acid and esomeprazole pretreatments alleviated indomethacin-induced damage by regulating oxidative stress, inflammatory response, the level of transforming growth factor-β (TGF-β), expressions of COX and prostaglandin E2, cell proliferation, apoptosis and regulation of the NF-κB signaling pathway. We conclude that either esomeprazole or syringic acid administration protected the gastric mucosa from harmful effects of indomethacin. Syringic acid might, therefore be a potential therapeutic agent for preventing and treating indomethacin-induced gastric damage.
Collapse
Affiliation(s)
- Irmak Ferah Okkay
- Department of Pharmacology, Faculty of Pharmacy, Ataturk University, Erzurum, Turkey
| | - Ufuk Okkay
- Department of Medical Pharmacology, Faculty of Medicine, Ataturk University, Erzurum, Turkey
| | - Betul Cicek
- Department of Physiology, Faculty of Medicine, Erzincan Binali Yildirim University, Erzincan, Turkey
| | - Ozhan Karatas
- Department of Pathology, Faculty of Veterinary Medicine, Sivas Cumhuriyet University, Sivas, Turkey
| | - Aysegul Yilmaz
- Department of Medical Pharmacology, Faculty of Medicine, Ataturk University, Erzurum, Turkey
| | - Fatma Yesilyurt
- Department of Medical Pharmacology, Faculty of Medicine, Ataturk University, Erzurum, Turkey
| | - Ahmet Hacimuftuoglu
- Department of Medical Pharmacology, Faculty of Medicine, Ataturk University, Erzurum, Turkey
| |
Collapse
|
4
|
Edgar KS, Cunning C, Gardiner TA, McDonald DM. BH4 supplementation reduces retinal cell death in ischaemic retinopathy. Sci Rep 2023; 13:21292. [PMID: 38042898 PMCID: PMC10693630 DOI: 10.1038/s41598-023-48167-5] [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: 02/27/2023] [Accepted: 11/22/2023] [Indexed: 12/04/2023] Open
Abstract
Dysregulation of nitric oxide (NO) production can cause ischaemic retinal injury and result in blindness. How this dysregulation occurs is poorly understood but thought to be due to an impairment in NO synthase function (NOS) and nitro-oxidative stress. Here we investigated the possibility of correcting this defective NOS activity by supplementation with the cofactor tetrahydrobiopterin, BH4. Retinal ischaemia was examined using the oxygen-induced retinopathy model and BH4 deficient Hph-1 mice used to establish the relationship between NOS activity and BH4. Mice were treated with the stable BH4 precursor sepiapterin at the onset of hypoxia and their retinas assessed 48 h later. HPLC analysis confirmed elevated BH4 levels in all sepiapterin supplemented groups and increased NOS activity. Sepiapterin treatment caused a significant decrease in neuronal cell death in the inner nuclear layer that was most notable in WT animals and was associated with significantly diminished superoxide and local peroxynitrite formation. Interestingly, sepiapterin also increased inflammatory cytokine levels but not microglia cell number. BH4 supplementation by sepiapterin improved both redox state and neuronal survival during retinal ischaemia, in spite of a paradoxical increase in inflammatory cytokines. This implicates nitro-oxidative stress in retinal neurones as the cytotoxic element in ischaemia, rather than enhanced pro-inflammatory signalling.
Collapse
Affiliation(s)
- Kevin S Edgar
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, 97 Lisburn Road, BT9 7BL, UK
| | - Ciara Cunning
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, 97 Lisburn Road, BT9 7BL, UK
| | - Tom A Gardiner
- School of Medicine, Dentistry and Biomedical Sciences, Centre for Biomedical Sciences Education, Queen's University Belfast, Belfast, UK
| | - Denise M McDonald
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, 97 Lisburn Road, BT9 7BL, UK.
| |
Collapse
|
5
|
Deliyanti D, Suphapimol V, Ang P, Tang X, Jayasimhan A, Wilkinson-Berka JL. Early Depletion of Neutrophils Reduces Retinal Inflammation and Neovascularization in Mice with Oxygen-Induced Retinopathy. Int J Mol Sci 2023; 24:15680. [PMID: 37958664 PMCID: PMC10648252 DOI: 10.3390/ijms242115680] [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: 09/08/2023] [Revised: 10/22/2023] [Accepted: 10/26/2023] [Indexed: 11/15/2023] Open
Abstract
Retinal inflammation is a central feature of ocular neovascular diseases such as diabetic retinopathy and retinopathy of prematurity, but the contribution of neutrophils to this process is not fully understood. We studied oxygen-induced retinopathy (OIR) which develops in two phases, featuring hyperoxia-induced retinal vaso-obliteration in phase I, followed by retinal neovascularization in phase II. As neutrophils are acute responders to tissue damage, we evaluated whether neutrophil depletion with an anti-Ly6G mAb administered in phase I OIR influenced retinal inflammation and vascular injury. Neutrophils were measured in blood and spleen via flow cytometry, and myeloperoxidase, an indicator of neutrophil activity, was evaluated in the retina using Western blotting. Retinal vasculopathy was assessed by quantitating vaso-obliteration, neovascularization, vascular leakage, and VEGF levels. The inflammatory factors, TNF, MCP-1, and ICAM-1 were measured in retina. In the OIR controls, neutrophils were increased in the blood and spleen in phase I but not phase II OIR. In OIR, the anti-Ly6G mAb reduced neutrophils in the blood and spleen, and myeloperoxidase, inflammation, and vasculopathy in the retina. Our findings revealed that the early rise in neutrophils in OIR primes the retina for an inflammatory and angiogenic response that promotes severe damage to the retinal vasculature.
Collapse
Affiliation(s)
| | | | | | | | | | - Jennifer L. Wilkinson-Berka
- Department of Anatomy and Physiology, School of Biomedical Sciences, University of Melbourne, Parkville, VIC 3010, Australia; (D.D.); (V.S.); (P.A.); (X.T.); (A.J.)
| |
Collapse
|
6
|
Hu Z, Xu W, Yang X, Li Y, Ma R, Hei Y, Hu J, Zhang Z, Wang L, Wang Y. SIRT2 inhibition attenuates the vasculopathy and vision impairment via Akt signaling in retinopathy of prematurity. Exp Eye Res 2023:109547. [PMID: 37348672 DOI: 10.1016/j.exer.2023.109547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 06/01/2023] [Accepted: 06/19/2023] [Indexed: 06/24/2023]
Abstract
Despite decades of research, the underlying mechanism of retinopathy of prematurity (ROP) remains unclear. The role of Sirt2, which is involved in both angiogenesis and inflammation, both pivotal in ROP, was investigated in an animal model of ROP known as oxygen-induced retinopathy (OIR). Our study found that Sirt2 was overexpressed and colocalized with microglia in OIR. Furthermore, it demonstrated that the level of Sirt2 was upregulated in hypoxia microglia BV-2 in vitro. Subsequently, our results elucidated that administration of the Sirt2 antagonist AGK2 attenuated the avascular and neovascular area and downregulated the expression of IGF-1. The phosphorylation of Akt and the expression of IGF-1 were upregulated in hypoxia BV-2 and conditional media collected from BV-2 under hypoxia promoted the migration and tube formation of retinal capillary endothelial cells, which were suppressed with AGK2. Notably, our findings are the first to demonstrate the deleterious role of Sirt2 in ROP, as Sirt2 inhibition led to the downregulation of Akt/IGF-1 and ameliorated vasculopathy, ultimately improving visual function. These results suggest that Sirt2 may be a promising therapeutic target for ROP.
Collapse
Affiliation(s)
- Zhicha Hu
- Department of Ophthalmology of the Third Medical Center of PLA General Hospital, PR China
| | - Wenqi Xu
- Department of Ophthalmology of the Third Medical Center of PLA General Hospital, PR China
| | - Xinji Yang
- Department of Ophthalmology of the Third Medical Center of PLA General Hospital, PR China
| | - Yueyue Li
- Department of Ophthalmology of the Third Medical Center of PLA General Hospital, PR China
| | - Rui Ma
- Department of Ophthalmology of the Third Medical Center of PLA General Hospital, PR China
| | - Yan Hei
- Department of Ophthalmology of the Third Medical Center of PLA General Hospital, PR China
| | - Jian Hu
- Department of Ophthalmology of the Third Medical Center of PLA General Hospital, PR China
| | - Zifeng Zhang
- Department of Ophthalmology, Eye Institute of China PLA, Xijing Hospital, the Fourth Military Medical University, PR China.
| | - Liqiang Wang
- Department of Ophthalmology of the Third Medical Center of PLA General Hospital, PR China.
| | - Yusheng Wang
- Department of Ophthalmology, Eye Institute of China PLA, Xijing Hospital, the Fourth Military Medical University, PR China.
| |
Collapse
|
7
|
Shan S, Liu F, Ford E, Caldwell RB, Narayanan SP, Somanath PR. Triciribine attenuates pathological neovascularization and vascular permeability in a mouse model of proliferative retinopathy. Biomed Pharmacother 2023; 162:114714. [PMID: 37080089 DOI: 10.1016/j.biopha.2023.114714] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/11/2023] [Accepted: 04/14/2023] [Indexed: 04/22/2023] Open
Abstract
Proliferative retinopathies are the leading cause of irreversible blindness in all ages, and there is a critical need to identify novel therapies. We investigated the impact of triciribine (TCBN), a tricyclic nucleoside analog and a weak Akt inhibitor, on retinal neurovascular injury, vascular permeability, and inflammation in oxygen-induced retinopathy (OIR). Post-natal day 7 (P7) mouse pups were subjected to OIR, and treated (i.p.) with TCBN or vehicle from P14-P16 and compared with age-matched, normoxic, vehicle or TCBN-treated controls. P17 retinas were processed for flat mounts, immunostaining, Western blotting, and qRT-PCR studies. Fluorescein angiography, electroretinography, and spectral domain optical coherence tomography were performed on days P21, P26, and P30, respectively. TCBN treatment significantly reduced pathological neovascularization, vaso-obliteration, and inflammation marked by reduced TNFα, IL6, MCP-1, Iba1, and F4/80 (macrophage/microglia markers) expression compared to the vehicle-treated OIR mouse retinas. Pathological expression of VEGF (vascular endothelial growth factor), and claudin-5 compromised the blood-retinal barrier integrity in the OIR retinas correlating with increased vascular permeability and neovascular tuft formation, which were blunted by TCBN treatment. Of note, there were no changes in the retinal architecture or retinal cell function in response to TCBN in the normoxia or OIR mice. We conclude that TCBN protects against pathological neovascularization, restores blood-retinal barrier homeostasis, and reduces retinal inflammation without adversely affecting the retinal structure and neuronal function in a mouse model of OIR. Our data suggest that TCBN may provide a novel therapeutic option for proliferative retinopathy.
Collapse
Affiliation(s)
- Shengshuai Shan
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA, 30912, USA; Research Department, Charlie Norwood VA Medical Center, Augusta, GA, 30901, USA; Vascular Biology Center, Augusta University, Augusta, GA, 30912, USA; Culver Vision Discovery Institute, Augusta University, Augusta, GA, 30912, USA
| | - Fang Liu
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA, 30912, USA; Research Department, Charlie Norwood VA Medical Center, Augusta, GA, 30901, USA; Vascular Biology Center, Augusta University, Augusta, GA, 30912, USA; Culver Vision Discovery Institute, Augusta University, Augusta, GA, 30912, USA
| | - Edith Ford
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA, 30912, USA
| | - Ruth B Caldwell
- Vascular Biology Center, Augusta University, Augusta, GA, 30912, USA; Culver Vision Discovery Institute, Augusta University, Augusta, GA, 30912, USA
| | - S Priya Narayanan
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA, 30912, USA; Research Department, Charlie Norwood VA Medical Center, Augusta, GA, 30901, USA; Vascular Biology Center, Augusta University, Augusta, GA, 30912, USA; Culver Vision Discovery Institute, Augusta University, Augusta, GA, 30912, USA.
| | - Payaningal R Somanath
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA, 30912, USA; Research Department, Charlie Norwood VA Medical Center, Augusta, GA, 30901, USA; Culver Vision Discovery Institute, Augusta University, Augusta, GA, 30912, USA.
| |
Collapse
|
8
|
Deliyanti D, Figgett WA, Gebhardt T, Trapani JA, Mackay F, Wilkinson-Berka JL. CD8 + T Cells Promote Pathological Angiogenesis in Ocular Neovascular Disease. Arterioscler Thromb Vasc Biol 2023; 43:522-536. [PMID: 36794587 DOI: 10.1161/atvbaha.122.318079] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
BACKGROUND CD4+ (cluster of differentation) and CD8+ T cells are increased in the ocular fluids of patients with neovascular retinopathy, yet their role in the disease process is unknown. METHODS We describe how CD8+ T cells migrate into the retina and contribute to pathological angiogenesis by releasing cytokines and cytotoxic factors. RESULTS In oxygen-induced retinopathy, flow cytometry revealed the numbers of CD4+ and CD8+ T cells were increased in blood, lymphoid organs, and retina throughout the development of neovascular retinopathy. Interestingly, the depletion of CD8+ T cells but not CD4+ T cells reduced retinal neovascularization and vascular leakage. Using reporter mice expressing gfp (green fluorescence protein) in CD8+ T cells, these cells were localized near neovascular tufts in the retina, confirming that CD8+ T cells contribute to the disease. Furthermore, the adoptive transfer of CD8+ T cells deficient in TNF (tumor necrosis factor), IFNγ (interferon gamma), Prf (perforin), or GzmA/B (granzymes A/B) into immunocompetent Rag1-/- mice revealed that CD8+ T cells mediate retinal vascular disease via these factors, with TNF influencing all aspects of vascular pathology. The pathway by which CD8+ T cells migrate into the retina was identified as CXCR3 (C-X-C motif chemokine receptor 3) with the CXCR3 blockade reducing the number of CD8+ T cells within the retina and retinal vascular disease. CONCLUSIONS We discovered that CXCR3 is central to the migration of CD8+ T cells into the retina as the CXCR3 blockade reduced the number of CD8+ T cells within the retina and vasculopathy. This research identified an unappreciated role for CD8+ T cells in retinal inflammation and vascular disease. Reducing CD8+ T cells via their inflammatory and recruitment pathways is a potential treatment for neovascular retinopathies.
Collapse
Affiliation(s)
- Devy Deliyanti
- Department of Anatomy and Physiology, School of Biomedical Sciences (D.D., J.L.W.-B.), University of Melbourne, Parkville, Victoria, Australia
- Department of Diabetes, Monash University, Melbourne, Victoria, Australia (D.D., J.L.W.-B.)
| | - William A Figgett
- Department of Microbiology and Immunology, School of Biomedical Sciences, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Parkville, Victoria, Australia (W.A.F.)
- Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia (W.A.F., T.G.)
| | - Thomas Gebhardt
- Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia (W.A.F., T.G.)
| | - Joseph A Trapani
- Sir Peter MacCallum Department of Oncology (J.A.T.), University of Melbourne, Parkville, Victoria, Australia
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia (J.A.T.)
| | - Fabienne Mackay
- QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia (F.M.)
| | - Jennifer L Wilkinson-Berka
- Department of Anatomy and Physiology, School of Biomedical Sciences (D.D., J.L.W.-B.), University of Melbourne, Parkville, Victoria, Australia
- Department of Diabetes, Monash University, Melbourne, Victoria, Australia (D.D., J.L.W.-B.)
| |
Collapse
|
9
|
Systemic Cytokines in Retinopathy of Prematurity. J Pers Med 2023; 13:jpm13020291. [PMID: 36836525 PMCID: PMC9966226 DOI: 10.3390/jpm13020291] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 01/30/2023] [Accepted: 01/30/2023] [Indexed: 02/09/2023] Open
Abstract
Retinopathy of prematurity (ROP), a vasoproliferative vitreoretinal disorder, is the leading cause of childhood blindness worldwide. Although angiogenic pathways have been the main focus, cytokine-mediated inflammation is also involved in ROP etiology. Herein, we illustrate the characteristics and actions of all cytokines involved in ROP pathogenesis. The two-phase (vaso-obliteration followed by vasoproliferation) theory outlines the evaluation of cytokines in a time-dependent manner. Levels of cytokines may even differ between the blood and the vitreous. Data from animal models of oxygen-induced retinopathy are also valuable. Although conventional cryotherapy and laser photocoagulation are well established and anti-vascular endothelial growth factor agents are available, less destructive novel therapeutics that can precisely target the signaling pathways are required. Linking the cytokines involved in ROP to other maternal and neonatal diseases and conditions provides insights into the management of ROP. Suppressing disordered retinal angiogenesis via the modulation of hypoxia-inducible factor, supplementation of insulin-like growth factor (IGF)-1/IGF-binding protein 3 complex, erythropoietin, and its derivatives, polyunsaturated fatty acids, and inhibition of secretogranin III have attracted the attention of researchers. Recently, gut microbiota modulation, non-coding RNAs, and gene therapies have shown promise in regulating ROP. These emerging therapeutics can be used to treat preterm infants with ROP.
Collapse
|
10
|
Wu X, Qian L, Zhao H, Lei W, Liu Y, Xu X, Li J, Yang Z, Wang D, Zhang Y, Zhang Y, Tang R, Yang Y, Tian Y. CXCL12/CXCR4: An amazing challenge and opportunity in the fight against fibrosis. Ageing Res Rev 2023; 83:101809. [PMID: 36442720 DOI: 10.1016/j.arr.2022.101809] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 11/24/2022] [Accepted: 11/24/2022] [Indexed: 11/26/2022]
Abstract
Fibrosis is a pathological process caused by abnormal wound healing response, which often leads to excessive deposition of extracellular matrix, distortion of organ architecture, and loss of organ function. Aging is an important risk factor for the development of organ fibrosis. C-X-C receptor 4 (CXCR4) is the predominant chemokine receptor on fibrocytes, C-X-C motif ligand 12 (CXCL12) is the only ligand of CXCR4. Accumulated evidence have confirmed that CXCL12/CXCR4 can be involved in multiple pathological mechanisms in fibrosis, such as inflammation, immunity, epithelial-mesenchymal transition, and angiogenesis. In addition, CXCL12/CXCR4 have also been shown to improve fibrosis levels in many organs including the heart, liver, lung and kidney; thus, they are promising targets for anti-fibrotic therapy. Notably, inhibitors of CXCL12 or CXCR4 also play an important role in various fibrosis-related diseases. In summary, this review systematically summarizes the role of CXCL12/CXCR4 in fibrosis, and this information is of great significance for understanding CXCL12/CXCR4. This will also contribute to the design of further studies related to CXCL12/CXCR4 and fibrosis, and shed light on potential therapies for fibrosis.
Collapse
Affiliation(s)
- Xue Wu
- Deparment of Neurology, Xi'an No.3 Hospital, Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Lu Qian
- Deparment of Neurology, Xi'an No.3 Hospital, Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Huadong Zhao
- Department of General Surgery, Tangdu Hospital, The Airforce Medical University, Xi'an, China
| | - Wangrui Lei
- Deparment of Neurology, Xi'an No.3 Hospital, Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Yanqing Liu
- Deparment of Neurology, Xi'an No.3 Hospital, Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Xiaoling Xu
- Deparment of Neurology, Xi'an No.3 Hospital, Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Jiawen Li
- Deparment of Neurology, Xi'an No.3 Hospital, Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Zhi Yang
- Department of General Surgery, Tangdu Hospital, The Airforce Medical University, Xi'an, China
| | - Du Wang
- Deparment of Neurology, Xi'an No.3 Hospital, Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Yuchen Zhang
- Deparment of Neurology, Xi'an No.3 Hospital, Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Yan Zhang
- Deparment of Neurology, Xi'an No.3 Hospital, Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Ran Tang
- Deparment of Neurology, Xi'an No.3 Hospital, Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Yang Yang
- Deparment of Neurology, Xi'an No.3 Hospital, Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China.
| | - Ye Tian
- Deparment of Neurology, Xi'an No.3 Hospital, Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China.
| |
Collapse
|
11
|
Ma X, Wu W, Liang W, Takahashi Y, Cai J, Ma JX. Modulation of cGAS-STING signaling by PPARα in a mouse model of ischemia-induced retinopathy. Proc Natl Acad Sci U S A 2022; 119:e2208934119. [PMID: 36409895 PMCID: PMC9860285 DOI: 10.1073/pnas.2208934119] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 10/10/2022] [Indexed: 11/22/2022] Open
Abstract
In ischemic retinopathy, overactivated retinal myeloid cells are a crucial driving force of pathological angiogenesis and inflammation. The cyclic GMP-AMP synthase (cGAS) and stimulator of interferon genes (STING) signaling are key regulators of inflammation. This study aims to investigate the association of cGAS-STING signaling with ischemic retinopathy and the regulation of its activation. We found that protein levels of cGAS and STING were markedly up-regulated in retinal myeloid cells isolated from mice with oxygen-induced retinopathy (OIR). Knockout of Sting and pharmacological inhibition of STING both alleviated retinal neovascularization (NV) and reduced retinal vascular leakage in OIR. Further, Sting knockout and STING inhibitor also alleviated leukocyte adhesion to retinal vasculature and infiltration into the retina as well as microglial activation in OIR. These results suggest that cGAS-STING signaling played a pathogenic role in retinal myeloid cell activation and NV in ischemic retinopathy. To identify the regulation of cGAS-STING signaling in OIR, we evaluated the role of transcription factor peroxisome proliferator-activated receptor α (PPARα). The results demonstrated that PPARα was down-regulated in OIR retinas, primarily in myeloid cells. Furthermore, Pparα knockout significantly up-regulated cGAS and STING levels in retinal CD11b+ cells, while PPARα agonist inhibited cGAS-STING signaling and cytosolic mitochondrial DNA (mtDNA) release, a causative feature for cGAS activation. Knockout of Sting ameliorated retinal NV, hyperpermeability, and leukostasis in Pparα-/- mice with OIR. These observations suggest that PPARα regulates cGAS-STING signaling, likely through mtDNA release, and thus, is a potential therapeutic target for ischemic retinopathy.
Collapse
Affiliation(s)
- Xiang Ma
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
- Department of Biochemistry, Wake Forest University School of Medicine, Winston-Salem, NC27157
| | - Wenjing Wu
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
- Department of Biochemistry, Wake Forest University School of Medicine, Winston-Salem, NC27157
| | - Wentao Liang
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
- Department of Biochemistry, Wake Forest University School of Medicine, Winston-Salem, NC27157
| | - Yusuke Takahashi
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
- Department of Biochemistry, Wake Forest University School of Medicine, Winston-Salem, NC27157
| | - Jiyang Cai
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
| | - Jian-xing Ma
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
- Department of Biochemistry, Wake Forest University School of Medicine, Winston-Salem, NC27157
| |
Collapse
|
12
|
Chidlow G, Chan WO, Wood JPM, Casson RJ. Investigations into photoreceptor energy metabolism during experimental retinal detachment. Front Cell Neurosci 2022; 16:1036834. [PMID: 36467607 PMCID: PMC9716104 DOI: 10.3389/fncel.2022.1036834] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 11/01/2022] [Indexed: 08/27/2023] Open
Abstract
Retinal detachment is a sight-threatening disorder, which occurs when the photoreceptors are separated from their vascular supply. The aim of the present study was to shed light on photoreceptor energy metabolism during experimental detachment in rats. Retinal detachment was induced in the eyes of rats via subretinal injection of sodium hyaluronate. Initially, we investigated whether detachment caused hypoxia within photoreceptors, as evaluated by the exogenous and endogenous biomarkers pimonidazole and HIF-1α, as well as by qPCR analysis of HIF target genes. The results showed no unequivocal staining for pimonidazole or HIF-1α within any detached retina, nor upregulation of HIF target genes, suggesting that any reduction in pO2 is of insufficient magnitude to produce hypoxia-induced covalent protein adducts or HIF-1α stabilisation. Subsequently, we analysed expression of cellular bioenergetic enzymes in photoreceptors during detachment. We documented loss of mitochondrial, and downregulation of glycolytic enzymes during detachment, indicating that photoreceptors have reduced energetic requirements and/or capacity. Given that detachment did not cause widespread hypoxia, but did result in downregulated expression of bioenergetic enzymes, we hypothesised that substrate insufficiency may be critical in terms of pathogenesis, and that boosting metabolic inputs may preserve photoreceptor bioenergetic production and, protect against their degeneration. Thus, we tested whether supplementation with the bioavailable energy substrate pyruvate mitigated rod and cone injury and degeneration. Despite protecting photoreceptors in culture from nutrient deprivation, pyruvate failed to protect against apoptotic death of rods, loss of cone opsins, and loss of inner segment mitochondria, in situ, when evaluated at 3 days after detachment. The regimen was also ineffective against cumulative photoreceptor deconstruction and degeneration when evaluated after 4 weeks. Retinal metabolism, particularly the bioenergetic profiles and pathological responses of the various cellular subtypes still presents a considerable knowledge gap that has important clinical consequences. While our data do not support the use of pyruvate supplementation as a means of protecting detached photoreceptors, they do provide a foundation and motivation for future research in this area.
Collapse
Affiliation(s)
- Glyn Chidlow
- Ophthalmic Research Laboratories, Discipline of Ophthalmology and Visual Sciences, University of Adelaide, Adelaide, SA, Australia
| | | | | | | |
Collapse
|
13
|
Liu Z, Mao X, Yang Q, Zhang X, Xu J, Ma Q, Zhou Y, Da Q, Cai Y, Sopeyin A, Dong Z, Hong M, Caldwell RB, Sodhi A, Huo Y. Suppression of myeloid PFKFB3-driven glycolysis protects mice from choroidal neovascularization. Br J Pharmacol 2022; 179:5109-5131. [PMID: 35830274 DOI: 10.1111/bph.15925] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 06/09/2022] [Accepted: 07/05/2022] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND AND PURPOSE Pathological angiogenesis is a major cause of irreversible blindness in individuals with neovascular age-related macular degeneration (nAMD). Macrophages and microglia (MΦ) contribute to aberrant ocular angiogenesis. However, the role of glucose metabolism of MΦ in nAMD is still undefined. Here, we have investigated the involvement of glycolysis, driven by the kinase/phosphatase PFKFB3, in the development of choroidal neovascularization (CNV). EXPERIMENTAL APPROACH CNV was induced in mice with laser photocoagulation. Choroid/retinal pigment epithelium (RPE) complexes and MΦ were isolated for analysis by qRT-PCR, western blot, flow cytometry, immunostaining, metabolic measurements and angiogenesis assays. KEY RESULTS MΦ accumulated within the CNV of murine nAMD models and expressed high levels of glycolysis-related enzymes and M1/M2 polarization markers. This phenotype of hyper-glycolytic and activated MΦ was replicated in bone marrow-derived macrophages stimulated by necrotic RPE in vitro. Myeloid cell-specific knockout of PFKFB3, a key glycolytic activator, attenuated pathological neovascularization in laser-induced CNV, which was associated with decreased expression of MΦ polarization markers and pro-angiogenic factors, along with decreased sprouting of vessels in choroid/RPE complexes. Mechanistically, necrotic RPE increased PFKFB3-driven glycolysis in macrophages, leading to activation of HIF-1α/HIF-2α and NF-κB, and subsequent induction of M1/M2 markers and pro-angiogenic cytokines, finally promoting macrophage reprogramming towards an angiogenic phenotype to facilitate development of CNV. The PFKFB3 inhibitor AZ67 also inhibited activation of HIF-1α/HIF-2α and NF-κB signalling and almost completely prevented laser-induced CNV in mice. CONCLUSIONS AND IMPLICATIONS Modulation of PFKFB3-mediated macrophage glycolysis and activation is a promising strategy for the treatment of nAMD.
Collapse
Affiliation(s)
- Zhiping Liu
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Xiaoxiao Mao
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Qiuhua Yang
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Xiaoyu Zhang
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Jiean Xu
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Qian Ma
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Yaqi Zhou
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Qingen Da
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Yongfeng Cai
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Anu Sopeyin
- Wilmer Eye Institute, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Zheng Dong
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
- Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia, USA
| | - Mei Hong
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Ruth B Caldwell
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
- Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia, USA
- James and Jean Culver Vision Discovery Institute, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Akrit Sodhi
- Wilmer Eye Institute, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Yuqing Huo
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
- James and Jean Culver Vision Discovery Institute, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| |
Collapse
|
14
|
O’Hare M, Esquiva G, McGahon MK, Hombrebueno JMR, Augustine J, Canning P, Edgar KS, Barabas P, Friedel T, Cincolà P, Henry J, Mayne K, Ferrin H, Stitt AW, Lyons TJ, Brazil DP, Grieve DJ, McGeown JG, Curtis TM. Loss of TRPV2-mediated blood flow autoregulation recapitulates diabetic retinopathy in rats. JCI Insight 2022; 7:e155128. [PMID: 36134661 PMCID: PMC9675469 DOI: 10.1172/jci.insight.155128] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 08/10/2022] [Indexed: 11/17/2022] Open
Abstract
Loss of retinal blood flow autoregulation is an early feature of diabetes that precedes the development of clinically recognizable diabetic retinopathy (DR). Retinal blood flow autoregulation is mediated by the myogenic response of the retinal arterial vessels, a process that is initiated by the stretch‑dependent activation of TRPV2 channels on the retinal vascular smooth muscle cells (VSMCs). Here, we show that the impaired myogenic reaction of retinal arterioles from diabetic animals is associated with a complete loss of stretch‑dependent TRPV2 current activity on the retinal VSMCs. This effect could be attributed, in part, to TRPV2 channel downregulation, a phenomenon that was also evident in human retinal VSMCs from diabetic donors. We also demonstrate that TRPV2 heterozygous rats, a nondiabetic model of impaired myogenic reactivity and blood flow autoregulation in the retina, develop a range of microvascular, glial, and neuronal lesions resembling those observed in DR, including neovascular complexes. No overt kidney pathology was observed in these animals. Our data suggest that TRPV2 dysfunction underlies the loss of retinal blood flow autoregulation in diabetes and provide strong support for the hypothesis that autoregulatory deficits are involved in the pathogenesis of DR.
Collapse
Affiliation(s)
- Michael O’Hare
- Wellcome-Wolfson Institute for Experimental Medicine and
| | - Gema Esquiva
- Wellcome-Wolfson Institute for Experimental Medicine and
| | - Mary K. McGahon
- Wellcome-Wolfson Institute for Experimental Medicine and
- Centre for Biomedical Sciences Education, Queen’s University Belfast, Belfast, United Kingdom
| | | | - Josy Augustine
- Wellcome-Wolfson Institute for Experimental Medicine and
| | - Paul Canning
- Wellcome-Wolfson Institute for Experimental Medicine and
| | - Kevin S. Edgar
- Wellcome-Wolfson Institute for Experimental Medicine and
| | - Peter Barabas
- Wellcome-Wolfson Institute for Experimental Medicine and
| | - Thomas Friedel
- Wellcome-Wolfson Institute for Experimental Medicine and
| | | | - Jennifer Henry
- Wellcome-Wolfson Institute for Experimental Medicine and
- Centre for Biomedical Sciences Education, Queen’s University Belfast, Belfast, United Kingdom
| | - Katie Mayne
- Wellcome-Wolfson Institute for Experimental Medicine and
- Centre for Biomedical Sciences Education, Queen’s University Belfast, Belfast, United Kingdom
| | - Hannah Ferrin
- Wellcome-Wolfson Institute for Experimental Medicine and
- Centre for Biomedical Sciences Education, Queen’s University Belfast, Belfast, United Kingdom
| | - Alan W. Stitt
- Wellcome-Wolfson Institute for Experimental Medicine and
| | | | | | | | | | - Tim M. Curtis
- Wellcome-Wolfson Institute for Experimental Medicine and
| |
Collapse
|
15
|
Fouda AY, Xu Z, Suwanpradid J, Rojas M, Shosha E, Lemtalsi T, Patel C, Xing J, Zaidi SA, Zhi W, Stansfield BK, Cheng PNM, Narayanan SP, Caldwell RW, Caldwell RB. Targeting proliferative retinopathy: Arginase 1 limits vitreoretinal neovascularization and promotes angiogenic repair. Cell Death Dis 2022; 13:745. [PMID: 36038541 PMCID: PMC9424300 DOI: 10.1038/s41419-022-05196-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 08/15/2022] [Accepted: 08/17/2022] [Indexed: 01/21/2023]
Abstract
Current therapies for treatment of proliferative retinopathy focus on retinal neovascularization (RNV) during advanced disease and can trigger adverse side-effects. Here, we have tested a new strategy for limiting neurovascular injury and promoting repair during early-stage disease. We have recently shown that treatment with a stable, pegylated drug form of the ureohydrolase enzyme arginase 1 (A1) provides neuroprotection in acute models of ischemia/reperfusion injury, optic nerve crush, and ischemic stroke. Now, we have determined the effects of this treatment on RNV, vascular repair, and retinal function in the mouse oxygen-induced retinopathy (OIR) model of retinopathy of prematurity (ROP). Our studies in the OIR model show that treatment with pegylated A1 (PEG-A1), inhibits pathological RNV, promotes angiogenic repair, and improves retinal function by a mechanism involving decreased expression of TNF, iNOS, and VEGF and increased expression of FGF2 and A1. We further show that A1 is expressed in myeloid cells and areas of RNV in retinal sections from mice with OIR and human diabetic retinopathy (DR) patients and in blood samples from ROP patients. Moreover, studies using knockout mice with hemizygous deletion of A1 show worsened RNV and retinal injury, supporting the protective role of A1 in limiting the OIR-induced pathology. Collectively, A1 is critically involved in reparative angiogenesis and neuroprotection in OIR. Pegylated A1 may offer a novel therapy for limiting retinal injury and promoting repair during proliferative retinopathy.
Collapse
Affiliation(s)
- Abdelrahman Y Fouda
- University of Arkansas for Medical Sciences, Little Rock, AR, USA.,Department of Clinical Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Zhimin Xu
- Vascular Biology Center, Augusta University, Augusta, GA, USA.,Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA
| | - Jutamas Suwanpradid
- Vascular Biology Center, Augusta University, Augusta, GA, USA.,Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA
| | - Modesto Rojas
- Vascular Biology Center, Augusta University, Augusta, GA, USA.,Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA.,Department of Pharmacology and Toxicology, Augusta University, Augusta, GA, USA
| | - Esraa Shosha
- University of Arkansas for Medical Sciences, Little Rock, AR, USA.,Department of Clinical Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Tahira Lemtalsi
- Vascular Biology Center, Augusta University, Augusta, GA, USA.,Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA
| | - Chintan Patel
- Vascular Biology Center, Augusta University, Augusta, GA, USA.,Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA
| | - Ji Xing
- Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA.,Department of Cellular Biology & Anatomy, Augusta University, Augusta, GA, USA
| | - Syed A Zaidi
- Vascular Biology Center, Augusta University, Augusta, GA, USA.,Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA
| | - Wenbo Zhi
- Center for Biotechnology and Genomic Medicine, Augusta University, Augusta, GA, USA
| | - Brain K Stansfield
- Vascular Biology Center, Augusta University, Augusta, GA, USA.,Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA.,Department of Pediatrics, Augusta University, Augusta, GA, USA
| | - Paul Ning-Man Cheng
- Bio-cancer Treatment International, 511-513, Bioinformatics Building, Hong Kong Science Park, Tai Po, Hong Kong SAR, China
| | - S Priya Narayanan
- Vascular Biology Center, Augusta University, Augusta, GA, USA.,Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA.,Department of Clinical and Administrative Pharmacy, University of Georgia, Augusta, GA, USA
| | - R William Caldwell
- Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA. .,Department of Pharmacology and Toxicology, Augusta University, Augusta, GA, USA.
| | - Ruth B Caldwell
- Vascular Biology Center, Augusta University, Augusta, GA, USA. .,Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA. .,Department of Cellular Biology & Anatomy, Augusta University, Augusta, GA, USA. .,Charlie Norwood VA Medical Center, Augusta, GA, USA.
| |
Collapse
|
16
|
Dysregulated genomic and coding-transcriptomic factors in retinopathy of prematurity. GENE REPORTS 2022. [DOI: 10.1016/j.genrep.2022.101558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
17
|
Klotzsche-von Ameln A, Sprott D. Harnessing retinal phagocytes to combat pathological neovascularization in ischemic retinopathies? Pflugers Arch 2022; 474:575-590. [PMID: 35524802 PMCID: PMC9117346 DOI: 10.1007/s00424-022-02695-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 04/28/2022] [Indexed: 12/12/2022]
Abstract
Ischemic retinopathies (IR) are vision-threatening diseases that affect a substantial amount of people across all age groups worldwide. The current treatment options of photocoagulation and anti-VEGF therapy have side effects and are occasionally unable to prevent disease progression. It is therefore worthwhile to consider other molecular targets for the development of novel treatment strategies that could be safer and more efficient. During the manifestation of IR, the retina, normally an immune privileged tissue, encounters enhanced levels of cellular stress and inflammation that attract mononuclear phagocytes (MPs) from the blood stream and activate resident MPs (microglia). Activated MPs have a multitude of effects within the retinal tissue and have the potential to both counter and exacerbate the harmful tissue microenvironment. The present review discusses the current knowledge about the role of inflammation and activated retinal MPs in the major IRs: retinopathy of prematurity and diabetic retinopathy. We focus particularly on MPs and their secreted factors and cell–cell-based interactions between MPs and endothelial cells. We conclude that activated MPs play a major role in the manifestation and progression of IRs and could therefore become a promising new target for novel pharmacological intervention strategies in these diseases.
Collapse
Affiliation(s)
| | - David Sprott
- Institute of Physiology, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
18
|
Gardiner TA, Branagh T, Tipping N, McDonald DM. Markers of Hypoxia and Metabolism Correlate With Cell Differentiation in Retina and Lens Development. FRONTIERS IN OPHTHALMOLOGY 2022; 2:867326. [PMID: 38983523 PMCID: PMC11182328 DOI: 10.3389/fopht.2022.867326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 03/24/2022] [Indexed: 07/11/2024]
Abstract
Recent studies have provided novel insights of co-development of the neural and vascular elements of the retina. Knowledge of these relationships are crucial to understand the impact of therapeutic measures in Retinopathy of Prematurity (ROP). ROP is imposed by therapeutic oxygen upon immature retinal blood vessels and neural cells causing delayed development and vascular regression. However, the impact of hyperoxia on developing retinal neurons is less understood because some aspects of normal development remain unknown. The metabolic changes during differentiation of retinal progenitor cells to functional neurons is one such aspect. We correlated immunomarkers of hypoxia with markers of metabolic change in developing retinal neurons during the early postnatal period in mice. The same marker proteins were studied in secondary lens fiber differentiation at postnatal day-3 (P3). Nuclear localization of the oxygen-sensitive subunits of hypoxia inducible factor, HIF-1α and HIF-2α was correlated with increasing mitochondrial content in differentiating neurons. Nuclear HIF was also correlated with AMP-dependent protein kinase (AMPK), and the AMPK phosphorylation target PPAR-gamma coactivator-1alpha (PGC-1α), the principal regulator of mitochondrial biogenesis. Expression of AMPK, PGC1α and HIF-2α in secondary fiber differentiation was visible in each profile of the lens equator. Strong nuclear localization for all markers was present at the onset of secondary fiber differentiation, and reflected changes in size, mitochondrial content, and metabolism. We speculate that the 'physiological hypoxia' that drives retinal vascular development is cell-specific and reliant upon neuronal differentiation and mitochondrial biogenesis. We suggest that the onset of differentiation increases energy consumption that is detected by AMPK. In turn AMPK increases mitochondrial biogenesis via PGC-1α. Mitochondrial oxygen consumption may then create intracellular hypoxia that activates HIF. This progression is congruent with the expression of these markers in secondary lens fiber differentiation and nuclear localization of HIF-2α. Nuclear localization of HIF-1α and HIF-2α in the postnatal retina is less defined than in the lens as it may involve the remnant of HIF expression from the embryonic period that is sustained and increased by intracellular hypoxia caused by increasing mitochondrial oxygen consumption. This the first report of the involvement of HIF-2α, AMPK and PGC-1α in lens development.
Collapse
Affiliation(s)
- Tom A Gardiner
- Centre for Biomedical Sciences Education, School of Medicine Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, United Kingdom
| | - Tiarnan Branagh
- Centre for Biomedical Sciences Education, School of Medicine Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, United Kingdom
| | - Nuala Tipping
- Centre for Biomedical Sciences Education, School of Medicine Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, United Kingdom
| | - Denise M McDonald
- Wellcome Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, United Kingdom
| |
Collapse
|
19
|
Fernandes AR, Dos Santos T, Granja PL, Sanchez-Lopez E, Garcia ML, Silva AM, Souto EB. Permeability, anti-inflammatory and anti-VEGF profiles of steroidal-loaded cationic nanoemulsions in retinal pigment epithelial cells under oxidative stress. Int J Pharm 2022; 617:121615. [PMID: 35217072 DOI: 10.1016/j.ijpharm.2022.121615] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 02/08/2022] [Accepted: 02/20/2022] [Indexed: 12/19/2022]
Abstract
Age-related macular degeneration (AMD) is defined as a degenerative, progressive and multifactorial disorder that affects the macula with a complex etiology. The retinal pigment epithelium is a monolayer of cells that has the function to separate the surface of the choroid from the neural retina that is involved in the signal transduction leading to vision. The blood-aqueous barrier and the blood retinal barrier limit the permeation of drugs into the retina and thereby reducing their efficacy. Triamcinolone acetonide (TA) is widely used as anti-inflammatory and immunomodulatory drug that promotes the inhibition of the inflammatory processes. The factors that stimulate or inhibit angiogenesis in AMD create a local balance that is responsible for the growth of sub-retinal neovascularization. In AMD, the main angiogenic stimulus is the vascular endothelial growth factor (VEGF). In this work, nanoemulsions with cationic surfactants (mono- and dicationic DABCO and quinuclidine) were produced to deliver TA, and were found to reduce the production of tumor necrosis factor alpha (TNF-α), which stimulates the choroidal neovascularization development by upregulating the VEGF production, and consequently decreased the VEGF levels. Our results support the potential use of mono- and dicationic DABCO and quinuclidine-based cationic nanoemulsions for the delivery of TA in the treatment of AMD.
Collapse
Affiliation(s)
- Ana R Fernandes
- i3S - Institute for Research & Innovation in Health, University of Porto, R. Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Biomedical Engineering National Institute, University of Porto, Alfredo Allen 208, 4200-135 Porto, Portugal; Faculty of Engineering, University of Porto, R. Dr. Roberto Frias, 4200-465 Porto, Portugal; Centre for Research and Technology of Agro-Environmental and Biological Sciences, CITAB, UTAD, Quinta de Prados, P-5001-801 Vila Real, Portugal; Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy, University of Barcelona, Barcelona, Spain
| | - Tiago Dos Santos
- i3S - Institute for Research & Innovation in Health, University of Porto, R. Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Biomedical Engineering National Institute, University of Porto, Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Pedro L Granja
- i3S - Institute for Research & Innovation in Health, University of Porto, R. Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Biomedical Engineering National Institute, University of Porto, Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Elena Sanchez-Lopez
- Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy, University of Barcelona, Barcelona, Spain; Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Barcelona, Spain
| | - Maria L Garcia
- Centre for Research and Technology of Agro-Environmental and Biological Sciences, CITAB, UTAD, Quinta de Prados, P-5001-801 Vila Real, Portugal; Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy, University of Barcelona, Barcelona, Spain
| | - Amelia M Silva
- Centre for Research and Technology of Agro-Environmental and Biological Sciences, CITAB, UTAD, Quinta de Prados, P-5001-801 Vila Real, Portugal; Department of Biology and Environment, University of Trás-os-Montes e Alto Douro, UTAD, Quinta de Prados, P-5001-801 Vila Real, Portugal.
| | - Eliana B Souto
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal; CEB - Centre of Biological Engineering, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; LABBELS - Associate Laboratory, Braga, Guimarães, Portugal.
| |
Collapse
|
20
|
Hui Q, Zheng F, Qin L, Pei C. Annexin A1 promotes reparative angiogenesis and ameliorates neuronal injury in ischemic retinopathy. Curr Eye Res 2022; 47:791-801. [PMID: 35179426 DOI: 10.1080/02713683.2022.2029904] [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] [Indexed: 11/03/2022]
Abstract
PURPOSE Retinal ischemia is the main reason for vision threatening. Inflammation and aberrant angiogenesis play an important role in the pathogenesis of ischemia. Annexin A1 is an endogenous protein modulating anti-inflammatory processes, and its therapeutic potential has been reported in a range of inflammatory diseases. However, the effect of annexin A1 on ischemic retinal injury has not been examined. METHODS Expression of annexin A1 was assessed by real time PCR and western blotting, and location of annexin A1 was evaluated by immunofluorescence staining in retina of OIR. The activation of annexin A1 were assayed in HRECs after hypoxia stimuli. The effect of annexin A1 on vascularization of OIR mouse through quantification vaso-obliteration and neovascularization, as well as expression of relevant angiogenic factors and inflammatory cytokines was compared between wild type and annexin A1 deficiency mice. We also investigated the effect of annexin A1 on retinal neuronal degeneration as measured by ERG and OCT. RESULTS In retinas of OIR, the expression of annexin A1 significantly increased and located in inner retinal layers. Annexin A1 was induced in HRECs after hypoxic stimuli. Furthermore, annexin A1 deficiency increased pro-angiogenic and pro-inflammatory cytokines. Ablation of annexin A1 suppressed aortic outgrowth and retinal reparative revascularization and promoted pathological neovascularization to exacerbate retinal dysfunction after ischemia injury. CONCLUSION Annexin A1 inhibits angiogenic and inhibits pro-inflammatory cytokines and promotes reparative angiogenesis, thus exhibits neuronal protective function in ischemic retinopathy.
Collapse
Affiliation(s)
- Qiaoyan Hui
- Department of Ophthalmology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.,Department of Ophthalmology, Xi'an Fourth Hospital, Affiliated Xi'an Fourth Hospital, Northwestern Polytechnical University, Affiliated Guangren Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Fengwei Zheng
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Li Qin
- Department of Ophthalmology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Cheng Pei
- Department of Ophthalmology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
21
|
Zhou L, Xu Z, Oh Y, Gamuyao R, Lee G, Xie Y, Cho H, Lee S, Duh EJ. Myeloid cell modulation by a GLP-1 receptor agonist regulates retinal angiogenesis in ischemic retinopathy. JCI Insight 2021; 6:93382. [PMID: 34673570 PMCID: PMC8675187 DOI: 10.1172/jci.insight.93382] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 10/20/2021] [Indexed: 01/04/2023] Open
Abstract
Ischemic retinopathies including diabetic retinopathy are major causes of blindness. Although neurons and Müller glia are recognized as important regulators of reparative and pathologic angiogenesis, the role of mononuclear phagocytes (MPs) — particularly microglia, the resident retinal immune cells — is unclear. Here, we found MP activation in human diabetic retinopathy, especially in neovessels from human neovascular membranes in proliferative retinopathy, including TNF-α expression. There was similar activation in the mouse oxygen-induced retinopathy (OIR) model of ischemia-induced neovascularization. Glucagon-like peptide-1 receptor (GLP-1R) agonists are in clinical use for glycemic control in diabetes and are also known to modulate microglia. Herein, we investigated the effect of a long-acting GLP-1R agonist, NLY01. Following intravitreal administration, NLY01 selectively localized to MPs in retina with OIR. NLY01 modulated MPs but not retinal endothelial cell viability, apoptosis, and tube formation in vitro. In OIR, NLY01 treatment inhibited MP infiltration and activation, including MP expression of cytokines in vivo. NLY01 significantly suppressed global induction of retinal inflammatory cytokines, promoted reparative angiogenesis, and suppressed pathologic retinal neovascularization. Collectively, these findings indicate the important role of mononuclear phagocytes in regulation of retinal vascularization in ischemia and suggest modulation of MPs as a potentially new treatment strategy for ischemic retinopathies.
Collapse
Affiliation(s)
| | | | - Yumin Oh
- Wilmer Eye Institute and.,The Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | | | | | | | - Seulki Lee
- Wilmer Eye Institute and.,The Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | |
Collapse
|
22
|
Sheemar A, Soni D, Takkar B, Basu S, Venkatesh P. Inflammatory mediators in diabetic retinopathy: Deriving clinicopathological correlations for potential targeted therapy. Indian J Ophthalmol 2021; 69:3035-3049. [PMID: 34708739 PMCID: PMC8725076 DOI: 10.4103/ijo.ijo_1326_21] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 08/10/2021] [Accepted: 09/27/2021] [Indexed: 11/22/2022] Open
Abstract
The role of inflammation in diabetic retinopathy (DR) is well-established and dysregulation of a large number of inflammatory mediators is known. These include cytokines, chemokines, growth factors, mediators of proteogenesis, and pro-apoptotic molecules. This para-inflammation as a response is not directed to a particular pathogen or antigen but is rather directed toward the by-products of the diabetic milieu. The inflammatory mediators take part in cascades that result in cellular level responses like neurodegeneration, pericyte loss, leakage, capillary drop out, neovascularization, etc. There are multiple overlaps between the inflammatory pathways occurring within the diabetic retina due to a large number of mediators, their varied sources, and cross-interactions. This makes understanding the role of inflammation in clinical manifestations of DR difficult. Currently, mediator-based therapy for DR is being evaluated for interventions that target a specific step of the inflammatory cascade. We reviewed the role of inflammation in DR and derived a simplified clinicopathological correlation between the sources and stimuli of inflammation, the inflammatory mediators and pathways, and the clinical manifestations of DR. By doing so, we deliberate mediator-specific therapy for DR. The cross-interactions between inflammatory mediators and the molecular cycles influencing the inflammatory cascades are crucial challenges to such an approach. Future research should be directed to assess the feasibility of the pathology-based therapy for DR.
Collapse
Affiliation(s)
- Abhishek Sheemar
- Department of Ophthalmology, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Deepak Soni
- Department of Ophthalmology, All India Institute of Medical Sciences, Bhopal, Madhya Pradesh, India
| | - Brijesh Takkar
- Smt. Kanuri Santhamma Center for Vitreoretinal Diseases, L V Prasad Eye Institute, Hyderabad, India
- Indian Health Outcomes, Public Health and Economics Research (IHOPE) Centre, L V Prasad Eye Institute, Hyderabad, India
| | - Soumyava Basu
- Uveitis Service, L V Prasad Eye Institute, Hyderabad, Telangana, India
| | - Pradeep Venkatesh
- Dr.R.P.Centre for Ophthalmic Sciences, All India Institute of Medical Science, New Delhi, India
| |
Collapse
|
23
|
Wickramasinghe LC, van Wijngaarden P, Tsantikos E, Hibbs ML. The immunological link between neonatal lung and eye disease. Clin Transl Immunology 2021; 10:e1322. [PMID: 34466225 PMCID: PMC8387470 DOI: 10.1002/cti2.1322] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 06/02/2021] [Accepted: 07/13/2021] [Indexed: 01/02/2023] Open
Abstract
Bronchopulmonary dysplasia (BPD) and retinopathy of prematurity (ROP) are two neonatal diseases of major clinical importance, arising in large part as a consequence of supplemental oxygen therapy used to promote the survival of preterm infants. The presence of coincident inflammation in the lungs and eyes of neonates receiving oxygen therapy indicates that a dysregulated immune response serves as a potential common pathogenic factor for both diseases. This review examines the current state of knowledge of immunological dysregulation in BPD and ROP, identifying similarities in the cellular subsets and inflammatory cytokines that are found in the alveoli and retina during the active phase of these diseases, indicating possible mechanistic overlap. In addition, we highlight gaps in the understanding of whether these responses emerge independently in the lung and retina as a consequence of oxygen exposure or arise because of inflammatory spill-over from the lung. As BPD and ROP are anatomically distinct, they are often considered discreet disease entities and are therefore treated separately. We propose that an improved understanding of the relationship between BPD and ROP is key to the identification of novel therapeutic targets to treat or prevent both conditions simultaneously.
Collapse
Affiliation(s)
- Lakshanie C Wickramasinghe
- Leukocyte Signalling LaboratoryDepartment of Immunology and PathologyCentral Clinical SchoolMonash UniversityMelbourneVICAustralia
| | - Peter van Wijngaarden
- OphthalmologyDepartment of SurgeryUniversity of MelbourneMelbourneVICAustralia
- Centre for Eye Research AustraliaRoyal Victorian Eye and Ear HospitalEast MelbourneVICAustralia
| | - Evelyn Tsantikos
- Leukocyte Signalling LaboratoryDepartment of Immunology and PathologyCentral Clinical SchoolMonash UniversityMelbourneVICAustralia
| | - Margaret L Hibbs
- Leukocyte Signalling LaboratoryDepartment of Immunology and PathologyCentral Clinical SchoolMonash UniversityMelbourneVICAustralia
| |
Collapse
|
24
|
Taniguchi H, Yoshida I, Sakamoto M, Maeno T. Epiretinal membrane appearance or progression after intravitreal injection in age-related macular degeneration. BMC Ophthalmol 2021; 21:190. [PMID: 33906612 PMCID: PMC8080384 DOI: 10.1186/s12886-021-01944-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 04/09/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The purpose of this study is to evaluate the influence of anti-vascular endothelial growth factor (VEGF) in the appearance or progression of epiretinal membranes (ERMs) in age-related macular degeneration (ARMD) and investigate confounding factors causing ERMs. METHODS Seventy-six eyes that were treated for more than 36 months from the first anti-VEGF injection were assessed. Binary logistic regression analysis was performed between smoking, lens status, subretinal hemorrhage, posterior vitreous detachment (PVD) status, peripheral retinal degeneration, type of AMD, conditions of contralateral eye, and the number of injections as independent variables and appearance or progression of ERMs during 36 months as dependent variables. RESULTS The presence of vitreomacular adhesion (VMA) or development of PVD during the observation period was significantly associated (Odds ratio [OR]: 5.77; 95% confidence interval [CI], 1.72-19.4; p = 0.005) with the appearance or progression of ERMs. Moreover, peripheral retinal degeneration was significantly associated (OR: 3.87; 95% CI, 1.15-13.0; p = 0.029). Injection number of anti-VEGF was not significantly associated (OR: 1.02; 95% CI, 0.90-1.16; p = 0.72). CONCLUSION This study suggests possibilities that anti-VEGF injections alone are unable to cause the development of ERMs, that VMA or developing PVD has a prior impact on the developing ERMs in ARMD similar to that of idiopathic ERMs, and that peripheral retinal degenerations and vitreomacular adhesion were both related to ERMs development and pathogenesis of ARMD.
Collapse
Affiliation(s)
- Hikari Taniguchi
- Sakura Medical Center, Toho University, 285-8741 Sakura-shi, Chiba, Japan
| | - Izumi Yoshida
- Sakura Medical Center, Toho University, 285-8741 Sakura-shi, Chiba, Japan. .,Toho-Kamagaya Hospital, 273-0132 Kamagaya-shi, Chiba, Japan.
| | - Masashi Sakamoto
- Sakura Medical Center, Toho University, 285-8741 Sakura-shi, Chiba, Japan
| | - Takatoshi Maeno
- Sakura Medical Center, Toho University, 285-8741 Sakura-shi, Chiba, Japan
| |
Collapse
|
25
|
Zhang Z, Song C, Wang T, Sun L, Qin L, Ju J. miR-139-5p promotes neovascularization in diabetic retinopathy by regulating the phosphatase and tensin homolog. Arch Pharm Res 2021; 44:205-218. [PMID: 33609236 DOI: 10.1007/s12272-021-01308-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 01/13/2021] [Indexed: 10/22/2022]
Abstract
Pathological retinal neovascularization is a driver of the progression of diabetic retinopathy (DR). The present study sought to identify the microRNAs (miRNAs) that are differentially expressed during the progression of DR as well as to explore the specific regulatory mechanism of those miRNAs in retinal neovascularization. Using a microarray data set and a diabetic mouse model, it was determined that miR-139-5p was significantly upregulated during the progression of DR. The in vitro investigation revealed an elevation in the miR-139-5p level in both the high glucose (HG)-treated mouse retinal microvascular endothelial cells (mRMECs) and the HG-treated human RMECs (hRMECs). The miR-139-5p overexpression elevated cell migration, facilitated tube formation, and increased vascular endothelial growth factor (VEGF) protein level in the hRMECs. While the angiogenic effect of miR-139-5p overexpression was halted by an anti-VEGF antibody. Meanwhile, the miR-139-5p knockdown eliminated the VEGF-induced cell migration and tube formation in the hRMECs. The phosphatase and tensin homolog (PTEN) was the target gene of the miR-139-5p. PTEN overexpression removed the angiogenic effect of miR-139-5p overexpression, which led to reduced cell migration and tube formation. In the diabetic mice, the miR-139-5p antagomir effectively decreased the acellular capillaries and suppressed the formation of aberrant blood vessels in the retinal tissues. Taken together, miR-139-5p promotes retinal neovascularization by repressing PTEN expression.
Collapse
Affiliation(s)
- Zhongwei Zhang
- Department of Ophthalmology, Weihai Municipal Hospital, Weihai, 264200, Shandong, China
| | - Caiping Song
- Department of Ophthalmology, Weihai Municipal Hospital, Weihai, 264200, Shandong, China
| | - Tao Wang
- Department of Ophthalmology, Weihai Municipal Hospital, Weihai, 264200, Shandong, China
| | - Lei Sun
- Department of Ophthalmology, Weihai Municipal Hospital, Weihai, 264200, Shandong, China
| | - Ling Qin
- Department of Ophthalmology, Weihai Municipal Hospital, Weihai, 264200, Shandong, China
| | - Jianghua Ju
- Department of Endocrinology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, 266035, Shandong, People's Republic of China.
| |
Collapse
|
26
|
Cho H, Kambhampati SP, Lai MJ, Zhou L, Lee G, Xie Y, Hui Q, Kannan RM, Duh EJ. Dendrimer-Triamcinolone Acetonide Reduces Neuroinflammation, Pathological Angiogenesis, and Neuroretinal Dysfunction in Ischemic Retinopathy. ADVANCED THERAPEUTICS 2021; 4:2000181. [PMID: 34527806 PMCID: PMC8436818 DOI: 10.1002/adtp.202000181] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Indexed: 12/11/2022]
Abstract
Diabetic retinopathy (DR) is the leading cause of blindness in working-age adults. Severe visual loss in DR is primarily due to proliferative diabetic retinopathy, characterized by pathologic preretinal angiogenesis driven by retinal ischemia. Microglia, the resident immune cells of the retina, have emerged as a potentially important regulator of pathologic retinal angiogenesis. Corticosteroids including triamcinolone acetonide (TA), known for their antiangiogenic effects, are used in treating retinal diseases, but their use is significantly limited by side effects including cataracts and glaucoma. Generation-4 hydroxyl polyamidoamine dendrimer nanoparticles are utilized to deliver TA to activated microglia in the ischemic retina in a mouse model of oxygen-induced retinopathy (OIR). Following intravitreal injection, dendrimer-conjugated TA (D-TA) exhibits selective localization and sustained retention in activated microglia in disease-associated areas of the retina. D-TA, but not free TA, suppresses inflammatory cytokine production, microglial activation, and preretinal neovascularization in OIR. In addition, D-TA, but not free TA, ameliorates OIR-induced neuroretinal and visual dysfunction. These results indicate that activated microglia are a promising therapeutic target for retinal angiogenesis and neuroprotection in ischemic retinal diseases. Furthermore, dendrimer-based targeted therapy and specifically D-TA constitute a promising treatment approach for DR, offering increased and sustained drug efficacy with reduced side effects.
Collapse
Affiliation(s)
- Hongkwan Cho
- Department of Ophthalmology, School of Medicine Johns Hopkins University, Baltimore, MD 21231, USA
| | - Siva P Kambhampati
- Department of Ophthalmology, School of Medicine Johns Hopkins University, Baltimore, MD 21231, USA; Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Michael J Lai
- Department of Ophthalmology, School of Medicine Johns Hopkins University, Baltimore, MD 21231, USA
| | - Lingli Zhou
- Department of Ophthalmology, School of Medicine Johns Hopkins University, Baltimore, MD 21231, USA
| | - Grace Lee
- Department of Ophthalmology, School of Medicine Johns Hopkins University, Baltimore, MD 21231, USA
| | - Yangyiran Xie
- Department of Ophthalmology, School of Medicine Johns Hopkins University, Baltimore, MD 21231, USA
| | - Qiaoyan Hui
- Department of Ophthalmology, School of Medicine Johns Hopkins University, Baltimore, MD 21231, USA
| | - Rangaramanujam M Kannan
- Department of Ophthalmology, School of Medicine Johns Hopkins University, Baltimore, MD 21231, USA; Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University Baltimore, MD 21218, USA
| | - Elia J Duh
- Department of Ophthalmology, School of Medicine Johns Hopkins University, Baltimore, MD 21231, USA; Center for Nanomedicine at the Wilmer Eye Institute Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| |
Collapse
|
27
|
The innate immune system in diabetic retinopathy. Prog Retin Eye Res 2021; 84:100940. [PMID: 33429059 DOI: 10.1016/j.preteyeres.2021.100940] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 12/24/2020] [Accepted: 01/03/2021] [Indexed: 12/20/2022]
Abstract
The prevalence of diabetes has been rising steadily in the past half-century, along with the burden of its associated complications, including diabetic retinopathy (DR). DR is currently the most common cause of vision loss in working-age adults in the United States. Historically, DR has been diagnosed and classified clinically based on what is visible by fundoscopy; that is vasculature alterations. However, recent technological advances have confirmed pathology of the neuroretina prior to any detectable vascular changes. These, coupled with molecular studies, and the positive impact of anti-inflammatory therapeutics in DR patients have highlighted the central involvement of the innate immune system. Reminiscent of the systemic impact of diabetes, immune dysregulation has become increasingly identified as a key element of the pathophysiology of DR by interfering with normal homeostatic systems. This review uses the growing body of literature across various model systems to demonstrate the clear involvement of all three pillars of the immune system: immune-competent cells, mediators, and the complement system. It also demonstrates how the relative contribution of each of these requires more extensive analysis, including in human tissues over the continuum of disease progression. Finally, although this review demonstrates how the complex interactions of the immune system pose many more questions than answers, the intimately connected nature of the three pillars of the immune system may also point to possible new targets to reverse or even halt reverse retinopathy.
Collapse
|
28
|
Synergistic interactions of PlGF and VEGF contribute to blood-retinal barrier breakdown through canonical NFκB activation. Exp Cell Res 2020; 397:112347. [PMID: 33130176 DOI: 10.1016/j.yexcr.2020.112347] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 10/26/2020] [Accepted: 10/29/2020] [Indexed: 12/22/2022]
Abstract
To investigate the role of placental growth factor/vascular endothelial growth factor (PlGF-VEGF) heterodimers are involved in the blood-retinal barrier (BRB) breakdown and the associated mechanism, human retinal endothelial cells (HRECs) were treated with recombinant human (rh)PlGF-VEGF heterodimers and rhPlGF and studied in normal and high-glucose conditions. HREC barrier function was evaluated by the measurement of trans-endothelial electrical resistance (TEER). Adeno-Associated Virus Type 5 (AAV5) vectors overexpressed PlGF in the retina by intravitreal injection into the C57BL6 mouse eye. AAV5-GFP vector and naïve animals were used as controls. Immunofluorescence (IF) and western blots examined the protein expression of PlGF-VEGF heterodimers, VEGF, PlGF, NFκB, p-IκBα, ZO-1, and VE-cadherin in HREC and mouse retina. PlGF-VEGF heterodimers were detected predominantly in the HREC cell nuclei based on IF and cytoplasmic and nuclear fractionation experiments. High glucose treatment increased PlGF-VEGF nuclear abundance. Dot immunoblotting demonstrated a strong affinity of the 5D11D4 antibody to PlGF-VEGF heterodimers. rhPlGF-VEGF disrupted the barrier function of HREC, which was prevented by the neutralization of PlGF-VEGF by the 5D11D4 antibody. Stimulation of HRECs with rhPlGF also led to an increase in the nuclear signals for PlGF-VEGF, p-IκBα, and colocalization of NFκB p65 and PlGF-VEGF in the nuclei. The selective IKK2 inhibitor IMD0354 disrupted the nuclear colocalization. Treatment with IMD0354 restored the barrier function of HREC, as indicated by the ZO-1 and VE-cadherin expression. In the mouse retinas, PlGF overexpression by AAV5 vector reduced ZO-1 expression and increased abundance of pIκBα. PIGF/VEGF heterodimers mediate BRB breakdown potentially through the canonical NFκB activation.
Collapse
|
29
|
Suzumura A, Terao R, Kaneko H. Protective Effects and Molecular Signaling of n-3 Fatty Acids on Oxidative Stress and Inflammation in Retinal Diseases. Antioxidants (Basel) 2020; 9:E920. [PMID: 32993153 PMCID: PMC7600094 DOI: 10.3390/antiox9100920] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 09/23/2020] [Accepted: 09/24/2020] [Indexed: 02/06/2023] Open
Abstract
Oxidative stress and inflammation play crucial roles in the development and progression of retinal diseases. Retinal damage by various etiologies can result in retinopathy of prematurity (ROP), diabetic retinopathy (DR), and age-related macular degeneration (AMD). n-3 fatty acids are essential fatty acids and are necessary for homeostasis. They are important retinal membrane components and are involved in energy storage. n-3 fatty acids also have antioxidant and anti-inflammatory properties, and their suppressive effects against ROP, DR, and AMD have been previously evaluated. α-linolenic acid (ALA), eicosapentaenoic acid (EPA), docosahexaenoic acid (DHA), and their metabolites have been shown to alleviate retinal oxidative stress and inflammation involving various biological signaling pathways. In this review, we summarize the current understanding of the n-3 fatty acids effects on the mechanisms of these retinal diseases and how they exert their therapeutic effects, focusing on ALA, EPA, DHA, and their metabolites. This knowledge may provide new remedial strategies for n-3 fatty acids in the prevention and treatment of retinal diseases associated with oxidative stress and inflammation.
Collapse
Affiliation(s)
- Ayana Suzumura
- Department of Ophthalmology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan;
| | - Ryo Terao
- Department of Ophthalmology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan;
| | - Hiroki Kaneko
- Department of Ophthalmology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan;
| |
Collapse
|
30
|
Luo Y, Tan Y, Wang X. Influence of polymorphisms in VEGF, TNF-α, and GSTP1 genes on retinopathy of prematurity risk: a Meta-analysis. J Matern Fetal Neonatal Med 2020; 35:1248-1257. [PMID: 32264735 DOI: 10.1080/14767058.2020.1745179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Purpose: To investigate the influence of polymorphisms in vascular endothelial growth factor (VEGF), tumor necrosis factor-α (TNF-α), and glutathione S-transferase Pi isoform (GSTP1) genes on retinopathy of prematurity (ROP) risk, we performed a Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA)-compliant meta-analysis.Methods: An exhaustive search was conducted in PubMed, Web of Science, and CNKI for genetic studies evaluating the relationship between VEGF (-460 T/C, +936 C/T, -634 G/C, and -2578 C/A), TNF-α (-308 G/A) and GSTP1 (Ile/Val) polymorphisms and ROP risk from inception until November 2019. Odds ratio (OR) with the 95% confidence interval (CI) were used for estimating combined effect size. The quality of the included studies was evaluated using the Newcastle-Ottawa Scale (NOS).Results: A total of 14 studies met the inclusion criteria. The meta-analyses revealed that VEGF - 460 T/C was associated with ROP risk in the allele model (C vs. T, OR = 0.83, 95% CI: 0.74-0.94, POR=0.004), homozygous gene model (CC vs. TT, OR = 0.70, 95% CI: 0.54-0.91, POR=0.008), dominant gene model (CC + TC vs. TT, OR = 0.80, 95% CI: 0.67-0.95, POR = 0.012), and recessive gene model (CC vs. TC + TT, OR = 0.74, 95% CI: 0.59-0.94, POR = 0.014). However, we did not find significant differences in the genotype and allele distribution of VEGF + 936 C/T, -634 G/C, -2578 C/A, TNF-α - 308 G/A and GSTP1 Ile/Val polymorphisms, between ROP and control group (p > .05).Conclusions: VEGF polymorphism -460 T/C was associated with a lower ROP risk. Further research is warranted to investigate haplotype effects of VEGF polymorphisms on the risk of ROP.
Collapse
Affiliation(s)
- Yulin Luo
- Department of Ophthalmology, Hunan Children's Hospital, Changsha, China
| | - Yilan Tan
- Department of Ophthalmology, Hunan Children's Hospital, Changsha, China
| | - Xilang Wang
- Department of Ophthalmology, Hunan Children's Hospital, Changsha, China
| |
Collapse
|
31
|
Wang T, Tsirukis DI, Sun Y. Targeting Neuroinflammation in Neovascular Retinal Diseases. Front Pharmacol 2020; 11:234. [PMID: 32210818 PMCID: PMC7076162 DOI: 10.3389/fphar.2020.00234] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 02/20/2020] [Indexed: 12/12/2022] Open
Abstract
Retinal blood vessels provide the necessary energy, nutrients and oxygen in order to support visual function and remove harmful particles from blood, thus acting to protect neuronal cells. The homeostasis of the retinal vessels is important for the maintenance of retinal visual function. Neovascularization is the most common cause of blindness in patients with retinopathy. Previous studies have shown that inflammatory mediators are known key regulators in retinopathy, but their causal link has been elusive. Although inflammation is often thought to arise from inflammatory cells like macrophages, neutrophils, and resident microglia, retinal neurons have also been reported to contribute to inflammation, through inflammatory signals, which mediate blood vessel growth. Therefore, it is important to explore the detailed mechanisms of neuroinflammation’s effects on retinal neovascularization. This review looks to summarize current research on the relationship between retinal angiogenesis and neuroinflammation in retinopathy, as well as the potential effects of neuroinflammation on retinal neovascularization in different animal models.
Collapse
Affiliation(s)
- Tianxi Wang
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Demetrios I Tsirukis
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Ye Sun
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
32
|
Fu Z, Sun Y, Cakir B, Tomita Y, Huang S, Wang Z, Liu CH, S. Cho S, Britton W, S. Kern T, Antonetti DA, Hellström A, E.H. Smith L. Targeting Neurovascular Interaction in Retinal Disorders. Int J Mol Sci 2020; 21:E1503. [PMID: 32098361 PMCID: PMC7073081 DOI: 10.3390/ijms21041503] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 02/13/2020] [Accepted: 02/21/2020] [Indexed: 02/07/2023] Open
Abstract
The tightly structured neural retina has a unique vascular network comprised of three interconnected plexuses in the inner retina (and choroid for outer retina), which provide oxygen and nutrients to neurons to maintain normal function. Clinical and experimental evidence suggests that neuronal metabolic needs control both normal retinal vascular development and pathological aberrant vascular growth. Particularly, photoreceptors, with the highest density of mitochondria in the body, regulate retinal vascular development by modulating angiogenic and inflammatory factors. Photoreceptor metabolic dysfunction, oxidative stress, and inflammation may cause adaptive but ultimately pathological retinal vascular responses, leading to blindness. Here we focus on the factors involved in neurovascular interactions, which are potential therapeutic targets to decrease energy demand and/or to increase energy production for neovascular retinal disorders.
Collapse
Affiliation(s)
- Zhongjie Fu
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Z.F.); (Y.S.); (B.C.); (Y.T.); (S.H.); (Z.W.); (C.-H.L.); (S.S.C.); (W.B.)
- Manton Center for Orphan Disease, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Ye Sun
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Z.F.); (Y.S.); (B.C.); (Y.T.); (S.H.); (Z.W.); (C.-H.L.); (S.S.C.); (W.B.)
| | - Bertan Cakir
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Z.F.); (Y.S.); (B.C.); (Y.T.); (S.H.); (Z.W.); (C.-H.L.); (S.S.C.); (W.B.)
| | - Yohei Tomita
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Z.F.); (Y.S.); (B.C.); (Y.T.); (S.H.); (Z.W.); (C.-H.L.); (S.S.C.); (W.B.)
| | - Shuo Huang
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Z.F.); (Y.S.); (B.C.); (Y.T.); (S.H.); (Z.W.); (C.-H.L.); (S.S.C.); (W.B.)
| | - Zhongxiao Wang
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Z.F.); (Y.S.); (B.C.); (Y.T.); (S.H.); (Z.W.); (C.-H.L.); (S.S.C.); (W.B.)
| | - Chi-Hsiu Liu
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Z.F.); (Y.S.); (B.C.); (Y.T.); (S.H.); (Z.W.); (C.-H.L.); (S.S.C.); (W.B.)
| | - Steve S. Cho
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Z.F.); (Y.S.); (B.C.); (Y.T.); (S.H.); (Z.W.); (C.-H.L.); (S.S.C.); (W.B.)
| | - William Britton
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Z.F.); (Y.S.); (B.C.); (Y.T.); (S.H.); (Z.W.); (C.-H.L.); (S.S.C.); (W.B.)
| | - Timothy S. Kern
- Center for Translational Vision Research, Gavin Herbert Eye Institute, Irvine, CA 92697, USA;
| | - David A. Antonetti
- Kellogg Eye Center, Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105, USA;
| | - Ann Hellström
- Section for Ophthalmology, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, 405 30 Göteborg, Sweden;
| | - Lois E.H. Smith
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Z.F.); (Y.S.); (B.C.); (Y.T.); (S.H.); (Z.W.); (C.-H.L.); (S.S.C.); (W.B.)
| |
Collapse
|
33
|
O'Leary C, McGahon MK, Ashraf S, McNaughten J, Friedel T, Cincolà P, Barabas P, Fernandez JA, Stitt AW, McGeown JG, Curtis TM. Involvement of TRPV1 and TRPV4 Channels in Retinal Angiogenesis. Invest Ophthalmol Vis Sci 2019; 60:3297-3309. [PMID: 31369032 DOI: 10.1167/iovs.18-26344] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose We investigate the contribution of TRPV1 and TRPV4 channels to retinal angiogenesis. Methods Primary retinal microvascular endothelial cells (RMECs) were used for RT-PCR, Western blotting, immunolabeling, Ca2+ signaling, and whole-cell patch-clamp studies while localization of TRPV1 also was assessed in retinal endothelial cells using whole mount preparations. The effects of pharmacologic blockers of TRPV1 and TRPV4 on retinal angiogenic activity was evaluated in vitro using sprout formation, cell migration, proliferation, and tubulogenesis assays, and in vivo using the mouse model of oxygen-induced retinopathy (OIR). Heteromultimerization of TRPV1 and TRPV4 channels in RMECs was assessed using proximity ligation assays (PLA) and electrophysiologic recording. Results TRPV1 mRNA and protein expression were identified in RMECs. TRPV1 labelling was found to be mainly localized to the cytoplasm with some areas of staining colocalizing with the plasma membrane. Staining patterns for TRPV1 were broadly similar in endothelial cells of intact vessels within retinal flat mounts. Functional expression of TRPV1 and TRPV4 in RMECs was confirmed by patch-clamp recording. Pharmacologic inhibition of TRPV1 or TRPV4 channels suppressed in vitro retinal angiogenesis through a mechanism involving the modulation of tubulogenesis. Blockade of these channels had no effect on VEGF-stimulated angiogenesis or Ca2+ signals in vitro. PLA and patch-clamp studies revealed that TRPV1 and TRPV4 form functional heteromeric channel complexes in RMECs. Inhibition of either channel reduced retinal neovascularization and promoted physiologic revascularization of the ischemic retina in the OIR mouse model. Conclusions TRPV1 and TRPV4 channels represent promising targets for therapeutic intervention in vasoproliferative diseases of the retina.
Collapse
Affiliation(s)
- Caitriona O'Leary
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University of Belfast, Belfast, United Kingdom
| | - Mary K McGahon
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University of Belfast, Belfast, United Kingdom
| | - Sadaf Ashraf
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University of Belfast, Belfast, United Kingdom
| | - Jennifer McNaughten
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University of Belfast, Belfast, United Kingdom
| | - Thomas Friedel
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University of Belfast, Belfast, United Kingdom
| | - Patrizia Cincolà
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University of Belfast, Belfast, United Kingdom
| | - Peter Barabas
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University of Belfast, Belfast, United Kingdom
| | - Jose A Fernandez
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University of Belfast, Belfast, United Kingdom
| | - Alan W Stitt
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University of Belfast, Belfast, United Kingdom
| | - J Graham McGeown
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University of Belfast, Belfast, United Kingdom
| | - Tim M Curtis
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University of Belfast, Belfast, United Kingdom
| |
Collapse
|
34
|
Desjarlais M, Dussault S, Rivard F, Harel S, Sanchez V, Hussain SNA, Rivard A. Forced expression of microRNA-146b reduces TRAF6-dependent inflammation and improves ischemia-induced neovascularization in hypercholesterolemic conditions. Atherosclerosis 2019; 289:73-84. [PMID: 31479774 DOI: 10.1016/j.atherosclerosis.2019.08.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 07/29/2019] [Accepted: 08/22/2019] [Indexed: 01/30/2023]
Abstract
BACKGROUND AND AIMS MicroRNA (miR)-146 is a key regulator of inflammation, endothelial activation and atherosclerosis. This study sought to define its potential role for the modulation of ischemia-induced neovascularization in atherosclerotic conditions. METHODS Next generation sequencing and qRT-PCR analyses were used to compare microRNA expression in the ischemic muscles of hypercholesterolemic ApoE-deficient (ApoE-/-) mice vs. wild type mice, and in HUVECs exposed or not to oxLDL. Neovascularization was investigated in a mouse model of hindlimb ischemia and the functional activities of HUVECs and pro-angiogenic cells (PACs) were assessed in vitro. RESULTS We found that miR-146b (but not miR-146a) is significantly reduced in the ischemic muscles of ApoE-/- mice, and in HUVECs exposed to oxLDL. Inhibition of miR-146b reduces angiogenesis in vitro, whereas forced expression of miR-146b rescues oxLDL-mediated impairment of endothelial cell proliferation and tube formation. Mechanistically, miR146b directly targets tumor necrosis factor-alpha (TNFa) Receptor Associated Factor 6 (TRAF6) to inhibit inflammation. We found that hypercholesterolemia and oxLDL exposure are associated with higher levels of TRAF6, and increased expression of TNFa. However, forced expression of miR-146b in high cholesterol conditions reduces the expression of these inflammatory factors. In vivo, intramuscular injection of miR-146b mimic reduces ischemic damages and restores blood flow recuperation and capillary density in the ischemic muscles of ApoE-/- mice. Treatment with miR-146b also increases the number and functional activities of pro-angiogenic cells (PACs). CONCLUSIONS Hypercholesterolemia is associated with reduced expression of miR-146b, which increases TRAF6-dependent inflammation and is associated with poor neovascularization in response to ischemia. Forced expression of miR-146b using a miR mimic could constitute a novel therapeutic strategy to improve ischemia-induced neovascularization in atherosclerotic conditions.
Collapse
Affiliation(s)
- Michel Desjarlais
- Department of Medicine, Centre Hospitalier de l'Université de Montréal (CHUM) Research Center, Montréal, Québec, Canada
| | - Sylvie Dussault
- Department of Medicine, Centre Hospitalier de l'Université de Montréal (CHUM) Research Center, Montréal, Québec, Canada
| | - François Rivard
- Department of Medicine, Centre Hospitalier de l'Université de Montréal (CHUM) Research Center, Montréal, Québec, Canada
| | - Sharon Harel
- Department of Medicine, McGill University Health Center, Montréal, Québec, Canada
| | - Veronica Sanchez
- Department of Medicine, McGill University Health Center, Montréal, Québec, Canada
| | - Sabah N A Hussain
- Department of Medicine, McGill University Health Center, Montréal, Québec, Canada
| | - Alain Rivard
- Department of Medicine, Centre Hospitalier de l'Université de Montréal (CHUM) Research Center, Montréal, Québec, Canada.
| |
Collapse
|
35
|
Prenatal and postnatal inflammation-related risk factors for retinopathy of prematurity. J Perinatol 2019; 39:964-973. [PMID: 30932029 PMCID: PMC6592770 DOI: 10.1038/s41372-019-0357-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 02/15/2019] [Accepted: 02/21/2019] [Indexed: 12/29/2022]
Abstract
OBJECTIVE To evaluate the relationship between prenatal and postnatal inflammation-related risk factors and severe retinopathy of prematurity (ROP). STUDY DESIGN The study included infants born <30 weeks in California from 2007 to 2011. Multivariable log-binomial regression was used to assess the association between prenatal and postnatal inflammation-related exposures and severe ROP, defined as stage 3-5 or surgery for ROP. RESULTS Of 14,816 infants, 10.8% developed severe ROP. Though prenatal inflammation-related risk factors were initially associated with severe ROP, after accounting for the effect of these risk factors on gestational age at birth through mediation analysis, the association was non-significant (P = 0.6). Postnatal factors associated with severe ROP included prolonged oxygen exposure, sepsis, intraventricular hemorrhage, and necrotizing enterocolitis. CONCLUSION Postnatal inflammation-related factors were associated with severe ROP more strongly than prenatal factors. The association between prenatal inflammation-related factors and ROP was explained by earlier gestational age in infants exposed to prenatal inflammation.
Collapse
|
36
|
Abstract
The retina is one of the most metabolically active tissues in the body, consuming high levels of oxygen and nutrients. A well-organized ocular vascular system adapts to meet the metabolic requirements of the retina to ensure visual function. Pathological conditions affect growth of the blood vessels in the eye. Understanding the neuronal biological processes that govern retinal vascular development is of interest for translational researchers and clinicians to develop preventive and interventional therapeutics for vascular eye diseases that address early drivers of abnormal vascular growth. This review summarizes the current knowledge of the cellular and molecular processes governing both physiological and pathological retinal vascular development, which is dependent on the interaction among retinal cell populations, including neurons, glia, immune cells, and vascular endothelial cells. We also review animal models currently used for studying retinal vascular development.
Collapse
Affiliation(s)
- Ye Sun
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, Massachusetts 02115, USA;
| | - Lois E H Smith
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, Massachusetts 02115, USA;
| |
Collapse
|
37
|
Ashraf S, Bell S, O'Leary C, Canning P, Micu I, Fernandez JA, O'Hare M, Barabas P, McCauley H, Brazil DP, Stitt AW, McGeown JG, Curtis TM. CAMKII as a therapeutic target for growth factor-induced retinal and choroidal neovascularization. JCI Insight 2019; 4:122442. [PMID: 30721154 PMCID: PMC6482993 DOI: 10.1172/jci.insight.122442] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 01/31/2019] [Indexed: 12/14/2022] Open
Abstract
While anti-VEGF drugs are commonly used to inhibit pathological retinal and choroidal neovascularization, not all patients respond in an optimal manner. Mechanisms underpinning resistance to anti‑VEGF therapy include the upregulation of other proangiogenic factors. Therefore, therapeutic strategies that simultaneously target multiple growth factor signaling pathways would have significant value. Here, we show that Ca2+/calmodulin-dependent kinase II (CAMKII) mediates the angiogenic actions of a range of growth factors in human retinal endothelial cells and that this kinase acts as a key nodal point for the activation of several signal transduction cascades that are known to play a critical role in growth factor-induced angiogenesis. We also demonstrate that endothelial CAMKIIγ and -δ isoforms differentially regulate the angiogenic effects of different growth factors and that genetic deletion of these isoforms suppresses pathological retinal and choroidal neovascularization in vivo. Our studies suggest that CAMKII could provide a novel and efficacious target to inhibit multiple angiogenic signaling pathways for the treatment of vasoproliferative diseases of the eye. CAMKIIγ represents a particularly promising target, as deletion of this isoform inhibited pathological neovascularization, while enhancing reparative angiogenesis in the ischemic retina.
Collapse
Affiliation(s)
- Sadaf Ashraf
- Wellcome-Wolfson Institute for Experimental Medicine and
| | - Samuel Bell
- Wellcome-Wolfson Institute for Experimental Medicine and
| | | | - Paul Canning
- Wellcome-Wolfson Institute for Experimental Medicine and
| | - Ileana Micu
- Advanced Imaging Core Technology Unit, Faculty of Medicine, Health and Life Sciences, Queen's University of Belfast, Belfast, United Kingdom
| | | | - Michael O'Hare
- Wellcome-Wolfson Institute for Experimental Medicine and
| | - Peter Barabas
- Wellcome-Wolfson Institute for Experimental Medicine and
| | | | - Derek P Brazil
- Wellcome-Wolfson Institute for Experimental Medicine and
| | - Alan W Stitt
- Wellcome-Wolfson Institute for Experimental Medicine and
| | | | - Tim M Curtis
- Wellcome-Wolfson Institute for Experimental Medicine and
| |
Collapse
|
38
|
Mezu-Ndubuisi OJ, Wang Y, Schoephoerster J, Falero-Perez J, Zaitoun IS, Sheibani N, Gong S. Intravitreal Delivery of VEGF-A 165-loaded PLGA Microparticles Reduces Retinal Vaso-Obliteration in an In Vivo Mouse Model of Retinopathy of Prematurity. Curr Eye Res 2018; 44:275-286. [PMID: 30383455 DOI: 10.1080/02713683.2018.1542736] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
PURPOSE Retinopathy of prematurity (ROP) is a condition of abnormal retinal vascularization with reduced levels of vascular endothelial growth factor (VEGF) causing vaso-obliteration (Phase I), followed by abnormal neovascularization from increased VEGF (Phase II). We hypothesized that intravitreal pro-angiogenic VEGF-A in microparticle form would promote earlier retinal revascularization in an oxygen-induced ischemic retinopathy (OIR) mouse model. MATERIALS AND METHODS Wildtype mice (39) were exposed to 77% oxygen from postnatal day 7 (P7) to P12. VEGF-A165-loaded poly(lactic-co-glycolic acid) (PLGA) (n = 15) or empty PLGA (n = 14) microparticles were fabricated using a water-in-oil-in-water double emulsion method, and injected intravitreally at P13 into mice right eyes (RE). Left eyes (LE) were untreated. At P20, after retinal fluorescein angiography, vascular parameters were quantified. Retinal VEGF levels at P13 and flatmounts at P20 were performed separately. RESULTS VEGF-A165-loaded microparticles had a mean diameter of 4.2 μm. with a loading level of 8.6 weight.%. Retinal avascular area was reduced in VEGF-treated RE (39.5 ± 9.0%) compared to untreated LE (52.6 ± 6.1%, p < 0.0001) or empty microparticle-treated RE (p < 0.001) and untreated LEs (p = 0.001). Retinal arteries in VEGF-treated RE were less tortuous than untreated LE (1.08 ± 0.05 vs. 1.18 ± 0.08, p < 0.001) or empty-microparticles-treated RE (p = 0.02). Retinal arterial tortuosity was similar in the LE of VEGF and empty microparticle-treated mice (P > 0.05). Retinal vein width was similar in VEGF-treated and empty microparticle-treated RE (P > 0.9), which were each less dilated than their contralateral LE (p < 0.01). VEGF levels were higher in P13 OIR mice than RA mice (p < 0.0001). Retinal flatmounts showed vaso-obliteration and neovascularization. CONCLUSIONS Endogenous retinal VEGF is suppressed in OIR mice. Exogenous intravitreal VEGF-A165-loaded microparticles in OIR mice reduced retinal vaso-obliteration and accelerated recovery from vein dilation and arterial tortuosity. This may be beneficial in preventing Phase II ROP without systemic effects.
Collapse
Affiliation(s)
- Olachi J Mezu-Ndubuisi
- a Department of Pediatrics , University of Wisconsin-Madison , Madison , WI , USA.,b Department of Ophthalmology and Visual Sciences , University of Wisconsin-Madison , Madison , WI , USA
| | - Yuyuan Wang
- c Department of Materials Science and Engineering and Wisconsin Institute for Discovery , University of Wisconsin-Madison , Madison , WI , USA
| | - Jamee Schoephoerster
- a Department of Pediatrics , University of Wisconsin-Madison , Madison , WI , USA
| | - Juliana Falero-Perez
- b Department of Ophthalmology and Visual Sciences , University of Wisconsin-Madison , Madison , WI , USA
| | - Ismail S Zaitoun
- b Department of Ophthalmology and Visual Sciences , University of Wisconsin-Madison , Madison , WI , USA
| | - Nader Sheibani
- b Department of Ophthalmology and Visual Sciences , University of Wisconsin-Madison , Madison , WI , USA.,d Department of Biomedical Engineering , University of Wisconsin-Madison , Madison , WI , USA
| | - Shaoqin Gong
- c Department of Materials Science and Engineering and Wisconsin Institute for Discovery , University of Wisconsin-Madison , Madison , WI , USA.,d Department of Biomedical Engineering , University of Wisconsin-Madison , Madison , WI , USA
| |
Collapse
|
39
|
Lei XJ, Zhao YX, Qiao T. Influence of polymorphisms in VEGF, ACE, TNF and GST genes on the susceptibility to retinopathy of prematurity among Chinese infants. Int J Ophthalmol 2018; 11:1451-1457. [PMID: 30225217 DOI: 10.18240/ijo.2018.09.04] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Accepted: 03/28/2018] [Indexed: 01/02/2023] Open
Abstract
AIM To investigate common polymorphisms in VEGF, ACE, TNF and GST genes with retinopathy of prematurity (ROP) risk among Chinese infants. METHODS Nine polymorphisms in the above genes were genotyped on 724 advanced cases of ROP and 878 prematurely-born infants of low birth weight who were without any ophthalmologic disease. The frequencies of the polymorphisms were compared between cases and controls to identify the association present, if any. RESULTS Of the nine polymorphisms, only two showed significant associations: ACE insertion deletion (ID) polymorphism (P=0.031) and TNF -308G/A polymorphism (P<0.001). The former was associated with a reduced ROP risk [ID genotype, adjusted OR (aOR): 0.603, 95%CI: 0.427-0.893, P=0.034; DD genotype, aOR: 0.468, 95%CI: 0.229-0.626, P=0.002], while the latter showed an increased risk (GA genotype, aOR: 1.956, 95%CI: 1.396-2.465, P<0.001; AA genotype, aOR: 2.809, 95%CI: 1.802-4.484, P<0.001). The association was also noted at the allele level (ACE D allele aOR: 0.698, 95%CI: 0.294-0.883, P<0.001; TNF -308A allele aOR: 1.776, 95%CI: 1.446-2.561, P<0.001). CONCLUSION The ACE ID polymorphism can protect against ROP development while the TNF -308G/A can increase the risk of the disease among Chinese infants.
Collapse
Affiliation(s)
- Xiao-Jun Lei
- Department of Ophthalmology, Friendship Hospital of Dalian, Dalian 116000, Liaoning Province, China
| | - Yong-Xia Zhao
- Department of Ophthalmology, Center Hospital in Cangzhou, Cangzhou 061000, Hebei Province, China
| | - Tong Qiao
- Department of Ophthalmology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai 200000, China
| |
Collapse
|
40
|
Suppression of connexin 43 phosphorylation promotes astrocyte survival and vascular regeneration in proliferative retinopathy. Proc Natl Acad Sci U S A 2018; 115:E5934-E5943. [PMID: 29891713 DOI: 10.1073/pnas.1803907115] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Degeneration of retinal astrocytes precedes hypoxia-driven pathologic neovascularization and vascular leakage in ischemic retinopathies. However, the molecular events that underlie astrocyte loss remain unclear. Astrocytes abundantly express connexin 43 (Cx43), a transmembrane protein that forms gap junction (GJ) channels and hemichannels. Cx channels can transfer toxic signals from dying cells to healthy neighbors under pathologic conditions. Here we show that Cx43 plays a critical role in astrocyte apoptosis and the resulting preretinal neovascularization in a mouse model of oxygen-induced retinopathy. Opening of Cx43 hemichannels was not observed following hypoxia. In contrast, GJ coupling between astrocytes increased, which could lead to amplification of injury. Accordingly, conditional deletion of Cx43 maintained a higher density of astrocytes in the hypoxic retina. We also identify a role for Cx43 phosphorylation in mediating these processes. Increased coupling in response to hypoxia is due to phosphorylation of Cx43 by casein kinase 1δ (CK1δ). Suppression of this phosphorylation using an inhibitor of CK1δ or in site-specific phosphorylation-deficient mice similarly protected astrocytes from hypoxic damage. Rescue of astrocytes led to restoration of a functional retinal vasculature and lowered the hypoxic burden, thereby curtailing neovascularization and neuroretinal dysfunction. We also find that absence of astrocytic Cx43 does not affect developmental angiogenesis or neuronal function in normoxic retinas. Our in vivo work directly links phosphorylation of Cx43 to astrocytic coupling and apoptosis and ultimately to vascular regeneration in retinal ischemia. This study reveals that targeting Cx43 phosphorylation in astrocytes is a potential direction for the treatment of proliferative retinopathies.
Collapse
|
41
|
Rathnasamy G, Foulds WS, Ling EA, Kaur C. Retinal microglia - A key player in healthy and diseased retina. Prog Neurobiol 2018; 173:18-40. [PMID: 29864456 DOI: 10.1016/j.pneurobio.2018.05.006] [Citation(s) in RCA: 139] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 03/09/2018] [Accepted: 05/29/2018] [Indexed: 01/04/2023]
Abstract
Microglia, the resident immune cells of the brain and retina, are constantly engaged in the surveillance of their surrounding neural tissue. During embryonic development they infiltrate the retinal tissues and participate in the phagocytosis of redundant neurons. The contribution of microglia in maintaining the purposeful and functional histo-architecture of the adult retina is indispensable. Within the retinal microenvironment, robust microglial activation is elicited by subtle changes caused by extrinsic and intrinsic factors. When there is a disturbance in the cell-cell communication between microglia and other retinal cells, for example in retinal injury, the activated microglia can manifest actions that can be detrimental. This is evidenced by activated microglia secreting inflammatory mediators that can further aggravate the retinal injury. Microglial activation as a harbinger of a variety of retinal diseases is well documented by many studies. In addition, a change in the microglial phenotype which may be associated with aging, may predispose the retina to age-related diseases. In light of the above, the focus of this review is to highlight the role played by microglia in the healthy and diseased retina, based on findings of our own work and from that of others.
Collapse
Affiliation(s)
- Gurugirijha Rathnasamy
- Department of Anatomy, Yong Loo Lin School of Medicine, Blk MD10, 4 Medical Drive, National University of Singapore, 117594, Singapore; Department of Ophthalmology and Visual Sciences, School of Medicine and Public Health, University of Wisconsin, Madison, WI, 53706, United States
| | - Wallace S Foulds
- Singapore Eye Research Institute Level 6, The Academia, Discovery Tower, 20 College Road, 169856, Singapore; University of Glasgow, Glasgow, Scotland, G12 8QQ, United Kingdom
| | - Eng-Ang Ling
- Department of Anatomy, Yong Loo Lin School of Medicine, Blk MD10, 4 Medical Drive, National University of Singapore, 117594, Singapore
| | - Charanjit Kaur
- Department of Anatomy, Yong Loo Lin School of Medicine, Blk MD10, 4 Medical Drive, National University of Singapore, 117594, Singapore.
| |
Collapse
|
42
|
Kim SJ, Port AD, Swan R, Campbell JP, Chan RVP, Chiang MF. Retinopathy of prematurity: a review of risk factors and their clinical significance. Surv Ophthalmol 2018; 63:618-637. [PMID: 29679617 DOI: 10.1016/j.survophthal.2018.04.002] [Citation(s) in RCA: 292] [Impact Index Per Article: 48.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 04/05/2018] [Accepted: 04/09/2018] [Indexed: 01/09/2023]
Abstract
Retinopathy of prematurity (ROP) is a retinal vasoproliferative disease that affects premature infants. Despite improvements in neonatal care and management guidelines, ROP remains a leading cause of childhood blindness worldwide. Current screening guidelines are primarily based on two risk factors: birth weight and gestational age; however, many investigators have suggested other risk factors, including maternal factors, prenatal and perinatal factors, demographics, medical interventions, comorbidities of prematurity, nutrition, and genetic factors. We review the existing literature addressing various possible ROP risk factors. Although there have been contradictory reports, and the risk may vary between different populations, understanding ROP risk factors is essential to develop predictive models, to gain insights into pathophysiology of retinal vascular diseases and diseases of prematurity, and to determine future directions in management of and research in ROP.
Collapse
Affiliation(s)
- Sang Jin Kim
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, USA; Department of Ophthalmology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Alexander D Port
- Department of Ophthalmology, Weill Cornell Medical College, New York, New York, USA
| | - Ryan Swan
- Department of Medical Informatics and Clinical Epidemiology, Oregon Health and Science University, Portland, Oregon, USA
| | - J Peter Campbell
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - R V Paul Chan
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, Illinois, USA; Center for Global Health, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Michael F Chiang
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, USA; Department of Medical Informatics and Clinical Epidemiology, Oregon Health and Science University, Portland, Oregon, USA.
| |
Collapse
|
43
|
Hellgren G, Löfqvist C, Hansen-Pupp I, Gram M, Smith LE, Ley D, Hellström A. Increased postnatal concentrations of pro-inflammatory cytokines are associated with reduced IGF-I levels and retinopathy of prematurity. Growth Horm IGF Res 2018; 39:19-24. [PMID: 29274846 PMCID: PMC5858996 DOI: 10.1016/j.ghir.2017.11.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 10/26/2017] [Accepted: 11/26/2017] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Retinopathy of prematurity (ROP) is a multifactorial disease linked to low insulin-like growth factor (IGF)-I levels and perhaps to postnatal inflammation. Here, we investigated the longitudinal postnatal serum concentrations of pro-inflammatory cytokines interleukin (IL)-6 and tumor necrosis factor (TNF)-α in relation to IGF-I levels and ROP. DESIGN The study cohort included 52 infants born before 31 gestational weeks. The infants were screened for ROP and classified as non-ROP (n=33), non-proliferative ROP (stages 1 and 2; n=10), or proliferative ROP (stage 3, all treated for ROP; n=9). Blood samples were collected at birth, 24h after birth, and then weekly until at least 36weeks postmenstrual age (PMA) (i.e., up to 13weeks after birth). Circulating levels of IL-6 and TNF-α were evaluated in relation to circulating IGF-I levels and ROP. RESULTS IL-6 levels negatively correlated with IGF-I levels between 5 and 8weeks after birth, (p<0.01 to p<0.05). At birth, the IL-6 and TNF-α levels were similar independent of later ROP. Twenty-four hours after birth, both IL-6 and TNF-α levels had increased in infants later treated for ROP (p<0.05). Postnatal, infants treated for ROP had higher IL-6 levels than infants without ROP. CONCLUSIONS The pro-inflammatory response is associated with low IGF-I levels and the development of ROP.
Collapse
Affiliation(s)
- Gunnel Hellgren
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Department of Pediatrics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - Chatarina Löfqvist
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ingrid Hansen-Pupp
- Department of Clinical Sciences, Lund University, Lund, Sweden; Skåne University Hospital, Sweden
| | - Magnus Gram
- Department of Clinical Sciences, Lund University, Lund, Sweden; Skåne University Hospital, Sweden
| | - Lois E Smith
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, USA
| | - David Ley
- Department of Clinical Sciences, Lund University, Lund, Sweden; Skåne University Hospital, Sweden
| | - Ann Hellström
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
44
|
Becker S, Wang H, Stoddard GJ, Hartnett ME. Effect of subretinal injection on retinal structure and function in a rat oxygen-induced retinopathy model. Mol Vis 2017; 23:832-843. [PMID: 29259390 PMCID: PMC5723151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 11/27/2017] [Indexed: 11/29/2022] Open
Abstract
PURPOSE Subretinal injections are used to deliver agents in experimental studies of retinal diseases, often through viral vectors. However, few studies have investigated the effects of subretinal injections alone on the structure and function of the healthy or diseased retina, particularly in models of oxygen-induced retinopathy (OIR). We report on the effects of subretinal injections in a rat OIR model, which is used to study mechanisms of retinopathy of prematurity. METHODS Within 6 h of birth, neonatal rat pups were exposed to repeated cycles of oxygen between 50% and 10% O2 every 24 h for 14 days and subsequently moved to room air. On postnatal day 8 (P8), animals were treated in both eyes with advancement of the injection needle into the vitreous (pilot-treated) or with a subretinal PBS injection (sPBS-treated) or were left untreated (untreated). Additional control animals were exposed to microscope light after eyelid opening only (light-treated). Retinal fundus images were recorded on P26. Areas of the avascular retina and intravitreal neovascularization were determined in flat mounted retinas stained with isolectin B4 on P32. Retinal function of the respective eyes was assessed with the Ganzfeld electroretinogram (ERG) on P31 or P32 and with focal ERG in the central retina on P28 or P29. The thickness of the retinal layers was measured with spectral domain optical coherence tomography (OCT) on P30 and in opsin- and TO-PRO 3-stained retinal cryosections from pups euthanized on P32. Two sections were analyzed in each pup. For each section, three images of three different locations were analyzed accounting for 18 thickness measurements per pup. RESULTS Compared to untreated animals, the avascular area of the retina was greater in the pilot-treated (p<0.05) and sPBS-treated eyes (p<0.01), and the sPBS-treated eyes had a greater avascular retinal area compared to the pilot-treated eyes (p<0.01). The intravitreal neovascular area was larger in the sPBS-treated eyes compared to the untreated eyes (p<0.01). The outer nuclear and outer segment layers were thinner in the pilot- (p<0.01) and sPBS-treated eyes (p<0.05) compared to the untreated eyes as measured with OCT and immunohistochemical staining of the retinal cryosections. Compared to the untreated eyes, the amplitudes of the scotopic a- and b-waves in the Ganzfeld ERG were reduced in the pilot-treated eyes (p<0.001 and p<0.01, respectively), but only the a-wave was reduced in the sPBS-treated eyes (p<0.001). The a-wave amplitude in the focal ERG was reduced in the pilot- and sPBS-treated eyes, and no difference was seen in the b-wave amplitude between any of the groups. There was no difference between the light-treated and untreated eyes in the areas of the avascular retina or intravitreal neovascularization or Ganzfeld or focal ERG. CONCLUSIONS Pilot injections alone without injection into the subretinal space resulted in an increased avascular retinal area, reduced thickness of the photoreceptors, and reduced ERG function compared to the untreated animals. Although subretinal PBS injections further increased the areas of avascular retina and intravitreal neovascularization and resulted in similar retinal thinning compared to the pilot treatment, inner retinal function was improved, as evidenced by higher Ganzfeld b-wave amplitudes. Differences in the Ganzfeld and focal ERGs may indicate that the peripheral retina is more susceptible to remote beneficial effects from potential protective mechanisms induced by subretinal injection. This study stresses the importance of appropriate controls in experiments with subretinal delivery of agents.
Collapse
Affiliation(s)
- Silke Becker
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT
| | - Haibo Wang
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT
| | | | | |
Collapse
|
45
|
Deng G, Moran EP, Cheng R, Matlock G, Zhou K, Moran D, Chen D, Yu Q, Ma JX. Therapeutic Effects of a Novel Agonist of Peroxisome Proliferator-Activated Receptor Alpha for the Treatment of Diabetic Retinopathy. Invest Ophthalmol Vis Sci 2017; 58:5030-5042. [PMID: 28979999 PMCID: PMC5633008 DOI: 10.1167/iovs.16-21402] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Purpose Clinical studies have shown that peroxisome proliferator-activated receptor alpha (PPARα) agonist fenofibrate has therapeutic effects on diabetic retinopathy (DR). The purpose of this study was to identify a novel PPARα agonist and to evaluate its beneficial effects on DR. Methods The transcriptional activity of PPARα was measured by a luciferase-based promoter assay. TUNEL was used to evaluate apoptosis in retinal precursor cells (R28). Diabetes was induced in rats by injection of streptozotocin. Retinal inflammation was examined using leukostasis assay, and retinal vascular leakage was measured using permeability assay. Retinal function was measured using electroretinogram (ERG) recording, and retinal apoptosis was quantified using the cell death ELISA. The anti-angiogenic effect was evaluated in the oxygen-induced retinopathy (OIR) model. Results A compound, 7-chloro-8-methyl-2-phenylquinoline-4-carboxylic acid (Y-0452), with a chemical structure distinct from existing PPARα agonists, activated PPARα transcriptional activity and upregulated PPARα expression. Y-0452 significantly inhibited human retinal capillary endothelial cell migration and tube formation. The compound also protected R28 cells against apoptosis and inhibited NF-κB signaling in R28 cells exposed to palmitate. In diabetic rats, Y-0452 ameliorated leukostasis and vascular leakage in the retina. In addition, Y-0452 preserved the retinal function and reduced retinal cell death in diabetic rats. Y-0452 also alleviated retinal neovascularization in the OIR model. Conclusions Y-0452 is a novel PPARα agonist and has therapeutic potential for DR.
Collapse
Affiliation(s)
- Guotao Deng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China.,Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Elizabeth P Moran
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Rui Cheng
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Greg Matlock
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Kelu Zhou
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - David Moran
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Danyang Chen
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Qiang Yu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Jian-Xing Ma
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| |
Collapse
|
46
|
Davidson CT, Dover AR, McVicar CM, Megaw R, Glenn JV, Hadoke PWF, Stitt AW, Walker BR. Inhibition or deletion of 11β-HSD1 does not increase angiogenesis in ischemic retinopathy. DIABETES & METABOLISM 2017; 43:480-483. [PMID: 28089372 DOI: 10.1016/j.diabet.2016.12.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 11/24/2016] [Accepted: 12/01/2016] [Indexed: 11/27/2022]
Affiliation(s)
- C T Davidson
- University/BHF Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, 47, Little France Crescent, Edinburgh EH16 4TJ, Scotland, UK
| | - A R Dover
- University/BHF Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, 47, Little France Crescent, Edinburgh EH16 4TJ, Scotland, UK
| | - C M McVicar
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - R Megaw
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, Scotland, UK
| | - J V Glenn
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - P W F Hadoke
- University/BHF Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, 47, Little France Crescent, Edinburgh EH16 4TJ, Scotland, UK
| | - A W Stitt
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - B R Walker
- University/BHF Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, 47, Little France Crescent, Edinburgh EH16 4TJ, Scotland, UK.
| |
Collapse
|
47
|
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
|
48
|
Chidlow G, Wood JPM, Casson RJ. Investigations into Hypoxia and Oxidative Stress at the Optic Nerve Head in a Rat Model of Glaucoma. Front Neurosci 2017; 11:478. [PMID: 28883787 PMCID: PMC5573812 DOI: 10.3389/fnins.2017.00478] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Accepted: 08/11/2017] [Indexed: 01/03/2023] Open
Abstract
The vascular hypothesis of glaucoma proposes that retinal ganglion cell axons traversing the optic nerve head (ONH) undergo oxygen and nutrient insufficiency as a result of compromised local blood flow, ultimately leading to their degeneration. To date, evidence for the hypothesis is largely circumstantial. Herein, we made use of an induced rat model of glaucoma that features reproducible and widespread axonal transport disruption at the ONH following chronic elevation of intraocular pressure. If vascular insufficiency plays a role in the observed axonal transport failure, there should exist a physical signature at this time point. Using a range of immunohistochemical and molecular tools, we looked for cellular events indicative of vascular insufficiency, including the presence of hypoxia, upregulation of hypoxia-inducible, or antioxidant-response genes, alterations to antioxidant enzymes, increased formation of superoxide, and the presence of oxidative stress. Our data show that ocular hypertension caused selective hypoxia within the laminar ONH in 11/13 eyes graded as either medium or high for axonal transport disruption. Hypoxia was always present in areas featuring injured axons, and, the greater the abundance of axonal transport disruption, the greater the likelihood of a larger hypoxic region. Nevertheless, hypoxic regions were typically focal and were not necessarily evident in sections taken deeper within the same ONH, while disrupted axonal transport was frequently encountered without any discernible hypoxia. Ocular hypertension caused upregulation of heme oxygenase-1—an hypoxia-inducible and redox-sensitive enzyme—in ONH astrocytes. The distribution and abundance of heme oxygenase-1 closely matched that of axonal transport disruption, and encompassed hypoxic regions and their immediate penumbra. Ocular hypertension also caused upregulations in the iron-regulating protein ceruloplasmin, the anaerobic glycolytic enzyme lactate dehydrogenase, and the transcription factors cFos and p-cJun. Moreover, ocular hypertension increased the generation of superoxide radicals in the retina and ONH, as well as upregulating the active subunit of the superoxide-generating enzyme NADPH oxidase, and invoking modest alterations to antioxidant-response enzymes. The results of this study provide further indirect support for the hypothesis that reduced blood flow to the ONH contributes to axonal injury in glaucoma.
Collapse
Affiliation(s)
- Glyn Chidlow
- Ophthalmic Research Laboratories, Discipline of Ophthalmology and Visual Sciences, University of AdelaideAdelaide, SA, Australia
| | - John P M Wood
- Ophthalmic Research Laboratories, Discipline of Ophthalmology and Visual Sciences, University of AdelaideAdelaide, SA, Australia
| | - Robert J Casson
- Ophthalmic Research Laboratories, Discipline of Ophthalmology and Visual Sciences, University of AdelaideAdelaide, SA, Australia
| |
Collapse
|
49
|
Wang J, Lin J, Kaiser U, Wohlfart P, Hammes HP. Absence of macrophage migration inhibitory factor reduces proliferative retinopathy in a mouse model. Acta Diabetol 2017; 54:383-392. [PMID: 28070752 DOI: 10.1007/s00592-016-0956-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 12/19/2016] [Indexed: 01/06/2023]
Abstract
AIMS Ischemia-induced neovascularization is the key feature of proliferative diabetic retinopathy. Macrophage migration inhibitory factor (MIF) is a pleiotropic proinflammatory and proangiogenic cytokine, and its levels are elevated in the vitreous of patients with proliferative diabetic retinopathy. In this study, we aimed at investigating the relative potential of MIF in the ischemia-induced retinal neovascularization. METHODS Both WT and MIF-knockout mice were subjected to the retinopathy of prematurity (ROP) model. Intraretinal vessel regrowth was assessed by whole-mount immunofluorescence, and preretinal neovascularization was analyzed in retinal vertical sections after periodic acid-Schiff staining in the hypoxic stage of the ROP model. Gene expression of selected proangiogenic and proinflammatory factors at postnatal day 13 (p13) was measured by real-time PCR. Vascular endothelial growth factor (VEGF) expression, recruitment of endothelial progenitor cells (EPCs) and microglial activation were analyzed with immunofluorescence. RESULTS MIF deficiency increased areas of vascular obliteration by 49%, reduced sprouting tips by 27% and inhibited preretinal angiogenesis by 35%. VEGF expression was reduced in Müller cells of MIF-knockout mice. MIF absence reduced gene expression of erythropoietin, tumor necrosis factor alpha and intercellular adhesion molecule-1 by 30, 70 and 50%, respectively, decreased the number of retinal EPCs by 37.5% and inhibited microglial activation in the hypoxic condition. CONCLUSIONS In conclusion, we found that MIF has proangiogenic and proinflammatory properties in retinal neovascularization. The proangiogenic role of MIF in ischemia-induced retinal neovascularization is associated with the expression of VEGF and erythropoietin, EPC recruitment and inflammation. Therefore, MIF has a potential role in the pathological angiogenesis of proliferative retinopathy.
Collapse
Affiliation(s)
- Jing Wang
- 5th Medical Department, Medical Faculty Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Jihong Lin
- 5th Medical Department, Medical Faculty Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Ulrike Kaiser
- 5th Medical Department, Medical Faculty Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Paulus Wohlfart
- R&D Diabetes Division, Research and Translational Medicine, Sanofi, Industriepark Höchst, 65926, Frankfurt, Germany
| | - Hans-Peter Hammes
- 5th Medical Department, Medical Faculty Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany.
| |
Collapse
|
50
|
Qing L, Lei P, Tang J, Wu P, Wang L, Xie J, Hu Y. Inflammatory response associated with choke vessel remodeling in the extended perforator flap model. Exp Ther Med 2017; 13:2012-2018. [PMID: 28565801 PMCID: PMC5443226 DOI: 10.3892/etm.2017.4205] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 12/23/2016] [Indexed: 01/06/2023] Open
Abstract
Ischemic necrosis of the surgical flap is a common complication. The behavior of choke vessels has an important role in skin flap survival. However, the mechanism of choke vessel remodeling has remained elusive. The purpose of the present study was to investigate the possible association between inflammatory responses and choke vessel remodeling in the extended perforator flap model in rats. After flap elevation, the animals were randomly divided into three groups (n=6 in each) for tissue analysis at three, five or seven days after flap surgery. Six additional rats served as a control group (no flap elevation). Tissue samples were collected from the choke zone for histological, western blot and PCR analyses. Monocyte chemoattractant protein-1 (MCP-1) and tumor necrosis factor-α (TNF-α) as inflammatory cytokines were examined in the present study. Histopathological analysis showed that dilation of choke vessels and increased vessel wall thickness was obvious after flap elevation. It also showed edema, inflammation cell aggregation after the operation. Compared with the control group, the protein and mRNA expression levels of MCP-1 and TNF-α were significantly increased at days 3, 5 and 7 after flap elevation, while reaching a maximum at day 5. These findings indicated that inflammatory responses may have an important role in choke vessel remodeling. MCP-1 and TNF-α may be considered as potential targets for modulating the behavior of choke vessels.
Collapse
Affiliation(s)
- Liming Qing
- Department of Orthopaedics, Xiangya Hospital of Central South University, Changsha, Hunan 410008, P.R. China
| | - Pengfei Lei
- Department of Orthopaedics, Xiangya Hospital of Central South University, Changsha, Hunan 410008, P.R. China
| | - Juyu Tang
- Department of Orthopaedics, Xiangya Hospital of Central South University, Changsha, Hunan 410008, P.R. China
| | - Pangfeng Wu
- Department of Orthopaedics, Xiangya Hospital of Central South University, Changsha, Hunan 410008, P.R. China
| | - Long Wang
- Department of Orthopaedics, Xiangya Hospital of Central South University, Changsha, Hunan 410008, P.R. China
| | - Jie Xie
- Department of Orthopaedics, Xiangya Hospital of Central South University, Changsha, Hunan 410008, P.R. China
| | - Yihe Hu
- Department of Orthopaedics, Xiangya Hospital of Central South University, Changsha, Hunan 410008, P.R. China
| |
Collapse
|