1
|
Zhao Y, Ge Z, Guo T, Liu H, Zhou Y, Chen J, Xu H, Chen Z. Complement C3/C3aR Signaling Pathway Inhibition Ameliorates Retinal Damage in Experimental Retinal Vein Occlusion. Invest Ophthalmol Vis Sci 2025; 66:2. [PMID: 40310626 PMCID: PMC12054687 DOI: 10.1167/iovs.66.5.2] [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: 01/21/2025] [Accepted: 04/04/2025] [Indexed: 05/02/2025] Open
Abstract
Purpose Retinal vein occlusion (RVO) is a common retinal vascular disease that severely threatens visual function. This study aims to elucidate the role of the complement C3/C3aR signaling pathway in a laser-induced RVO mouse model and to explore its potential as a therapeutic target. Methods RVO was induced in C57BL/6J mice using laser photocoagulation combined with photosensitizer dye administration. Two days later, retinal tissues were collected for bulk RNA sequencing. The activation of the C3/C3aR signaling pathway was validated through RT-qPCR and Western blot. The C3aR antagonist SB290157 (C3aRA) was administered intravitreally and retinal morphological and functional changes were examined 1, 2, and 8 days later by optical coherence tomography (OCT), fundus photography (FP), and fluorescein angiography (FA), optomotor response (OKR) test, and electroretinogram (ERG). Results RVO mice exhibited marked increases in retinal thickness (P < 0.001) and fluorescence leakage (P < 0.01) compared to the sham-laser group. Bulk RNA-seq revealed significant upregulation of the complement pathway. Elevated expression of C3 and C3aR (P < 0.05) was confirmed by RT-qPCR and Western blot. Blocking C3aR with SB290157 significantly alleviated RVO-induced retinal edema, vascular leakage, and structural damage. Functional assessment showed that SB290157 treatment significantly improved contrast sensitivity (P < 0.05), increased b-wave (P < 0.001), and oscillatory potentials (Ops) amplitudes (P < 0.05) in RVO mice. RNA-seq analysis demonstrated that SB290157 significantly reduced the inflammatory mediator-related pathways and upregulated visual perception pathways (P < 0.05). Conclusions The complement C3/C3aR signaling pathway is critically involved in RVO-induced retinal damage and targeting this pathway may be a promising approach for RVO treatment.
Collapse
Affiliation(s)
- Yanying Zhao
- Aier Academy of Ophthalmology, Central South University, Changsha, Hunan Province, China
- Changsha Aier Eye Hospital, Changsha, Hunan Province, China
| | - Zhengwei Ge
- Changsha Aier Eye Hospital, Changsha, Hunan Province, China
- Aier Eye Hospital, Jinan University, Guangzhou, Guangdong Province, China
| | - Tingting Guo
- Changsha Aier Eye Hospital, Changsha, Hunan Province, China
- Aier Eye Hospital, Jinan University, Guangzhou, Guangdong Province, China
| | - Hengwei Liu
- Changsha Aier Eye Hospital, Changsha, Hunan Province, China
| | - Yufan Zhou
- Changsha Aier Eye Hospital, Changsha, Hunan Province, China
| | - Juan Chen
- Changsha Aier Eye Hospital, Changsha, Hunan Province, China
- Aier Eye Hospital, Jinan University, Guangzhou, Guangdong Province, China
| | - Heping Xu
- Aier Academy of Ophthalmology, Central South University, Changsha, Hunan Province, China
- Changsha Aier Eye Hospital, Changsha, Hunan Province, China
- Aier Eye Institute, Changsha, Hunan Province, China
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, United Kingdom
| | - Zhongping Chen
- Aier Academy of Ophthalmology, Central South University, Changsha, Hunan Province, China
- Changsha Aier Eye Hospital, Changsha, Hunan Province, China
- Aier Eye Hospital, Jinan University, Guangzhou, Guangdong Province, China
- School of Stomatology and Ophthalmology, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei Province, China
| |
Collapse
|
2
|
Nakanishi K, Takamura Y, Nakano Y, Inatani M, Oki M. The HAT Inhibitor ISOX-DUAL Diminishes Ischemic Areas in a Mouse Model of Oxygen-Induced Retinopathy. Genes Cells 2025; 30:e13196. [PMID: 39916601 PMCID: PMC11803434 DOI: 10.1111/gtc.13196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 11/22/2024] [Accepted: 01/07/2025] [Indexed: 02/11/2025]
Abstract
Retinal ischemic disease results in significant visual impairment due to the development of fragile and disorganized, pathologically running blood vessels in the eye. Currently, the mainstay treatment for this disease is the intravitreal administration of anti-VEGF drugs targeting vascular endothelial growth factor (VEGF), which induces angiogenesis. However, current anti-VEGF drugs do not diminish the ischemic areas that lead to angiogenesis, making fundamental treatment challenging. Since retinopathy is an acquired disease caused by hypoxic stimulation from ischemia, we paid particular attention to histone acetylases. We conducted a drug screening experiment using a mouse model of oxygen-induced retinopathy (OIR), which replicates retinal ischemic disease, through the intraperitoneal administration of 17 distinct inhibitors targeting histone acetyltransferases (HAT). The results indicated that, among the 17 inhibitors, only ISOX-DUAL decreased neovascularization and ischemic regions. Furthermore, microarray analysis was conducted on the drug-treated samples to refine genes altered by the administration of ISOX-DUAL. There were 21 genes associated with angiogenesis, including Angpt2, Hmox1, Edn1, and Serpine1, exhibited upregulation in OIR mice and downregulation following treatment with ISOX-DUAL. Furthermore, STRING analysis confirmed that the aforementioned four genes are downstream factors of hypoxia-inducible factors and are assumed to be important factors in retinal ischemic diseases.
Collapse
Affiliation(s)
- Kengo Nakanishi
- Department of Industrial Creation Engineering, Graduate School of EngineeringUniversity of FukuiFukuiJapan
| | - Yoshihiro Takamura
- Department of Ophthalmology, Faculty of Medical SciencesUniversity of FukuiFukuiJapan
| | - Yusei Nakano
- Department of Industrial Creation Engineering, Graduate School of EngineeringUniversity of FukuiFukuiJapan
| | - Masaru Inatani
- Department of Ophthalmology, Faculty of Medical SciencesUniversity of FukuiFukuiJapan
| | - Masaya Oki
- Department of Industrial Creation Engineering, Graduate School of EngineeringUniversity of FukuiFukuiJapan
- Life Science Innovation CenterUniversity of FukuiFukuiJapan
| |
Collapse
|
3
|
Darabuş DM, Dărăbuş RG, Munteanu M. The Diagnosis and Treatment of Branch Retinal Vein Occlusions: An Update. Biomedicines 2025; 13:105. [PMID: 39857689 PMCID: PMC11763247 DOI: 10.3390/biomedicines13010105] [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: 09/28/2024] [Revised: 12/26/2024] [Accepted: 01/03/2025] [Indexed: 01/27/2025] Open
Abstract
Branch retinal vein occlusion (BRVO) is a common retinal vascular condition and a significant contributor to vision loss worldwide, particularly in middle-aged and elderly populations. This review synthesizes current knowledge on the epidemiology, pathogenesis, and clinical features of BRVO, alongside recent advancements in diagnostic and therapeutic strategies. BRVO is approximately four times more prevalent than central retinal vein occlusion (CRVO) and often leads to significant vision impairment. By focusing on BRVO, this review aims to address the specific challenges and advancements in its diagnosis and management. The pathophysiology of BRVO is complex, involving factors such as venous compression, inflammation, and increased levels of vascular endothelial growth factor (VEGF). Diagnostic approaches such as optical coherence tomography (OCT) and fluorescein angiography are highlighted for their roles in assessing disease severity and guiding treatment decisions. Therapeutic interventions, including laser photocoagulation, anti-VEGF therapy, and intravitreal corticosteroids, are critically evaluated, emphasizing emerging treatments such as gene therapy, peptide-based agents, and small-molecule inhibitors. Despite advancements in management strategies, the recurrence of macular edema and treatment resistance remain significant challenges. Continued research is essential to refine therapeutic protocols and improve long-term visual outcomes for patients with BRVO.
Collapse
Affiliation(s)
- Diana-Maria Darabuş
- Department of Ophthalmology, “Victor Babeş” University of Medicine and Pharmacy, 300041 Timișoara, Romania; (D.-M.D.); (M.M.)
| | | | - Mihnea Munteanu
- Department of Ophthalmology, “Victor Babeş” University of Medicine and Pharmacy, 300041 Timișoara, Romania; (D.-M.D.); (M.M.)
| |
Collapse
|
4
|
Nakamura S, Hara H. [The potential of BCL6B as a therapeutic target for chorioretinal vascular lesions]. Nihon Yakurigaku Zasshi 2025; 160:26-30. [PMID: 39756899 DOI: 10.1254/fpj.24064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
Abstract
The ocular tissue is one of the most densely populated tissues in the body with extremely small blood vessels, and vascular lesions have been reported to be a factor in vision loss and visual field defects in many ocular diseases. Currently, vascular endothelial growth factor (VEGF)-targeted agents are the first line of treatment for intraocular vascular lesions, however, there are some cases in which they are not fully effective. Therefore, we explored pathogenic molecules other than VEGF, aiming to develop new molecular-targeted therapy. Using an experimental pathological model mimicking intraocular vascular lesions, we found that B-cell CLL/lymphoma 6 member B protein (BCL6B), which has been identified as a Bric-a-brac, Tramtrack, and Broad Complex protein, may play an important role in intraocular angiogenesis and vascular hyperpermeability. In this article, we introduce the usefulness of suppressing BCL6B expression and discuss the possibility of drug discovery by targeting Notch signaling in chorioretinal vascular lesions.
Collapse
Affiliation(s)
- Shinsuke Nakamura
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University
| | - Hideaki Hara
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University
| |
Collapse
|
5
|
Kuriyama A, Nakamura S, Inokuchi Y, Abe H, Yasuda H, Hidaka Y, Nagaoka K, Soeda T, Shimazawa M, Hara H. The protective effect of anti-VEGF-A/Ang-2 bispecific antibody on retinal vein occlusion model mice. Eur J Pharmacol 2024; 976:176691. [PMID: 38821166 DOI: 10.1016/j.ejphar.2024.176691] [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: 12/13/2023] [Revised: 05/27/2024] [Accepted: 05/28/2024] [Indexed: 06/02/2024]
Abstract
(233/250) Retinal vein occlusion (RVO) causes macular edema and retinal ischemia resulting in visual field and vision loss. A bispecific antibody that blocks VEGF-A and angiopoietin-2 (Ang-2) has been recently launched and applied clinically to treat macular edema, but the role of Ang-2 in the pathogenesis of RVO is still unclear. In this study, we investigated the effects of the anti-VEGF-A/anti-Ang-2 bispecific antibody (BsAb) in a murine RVO model. By using RVO model mice, the expression of Ang-2 gene and protein was examined in the retina through real-time qPCR and Western blotting, respectively. A significant increase in Ang-2 was detected 1 day after occlusion. Immediately after occlusion, control IgG 400 μg/mL, anti-VEGF-A antibody 200 μg/mL, anti-Ang-2 antibody 200 μg/mL, and BsAb 400 μg/mL were intravitreally administered at 2 μL. Visual function was examined using electroretinograms, and apoptosis was examined using TUNEL staining. Interestingly, BsAb partially suppressed the decrease in amplitude of a and b waves compared to control IgG. Anti-Ang-2 antibody and BsAb reduced apoptosis-positive cells 1 day after occlusion. Comprehensive gene expression profiles were also examined using RNA sequencing analysis. RNA sequencing analysis of the retinal tissues showed that BsAb suppressed expression of gene groups associated with inflammatory response and vascular development compared to anti-VEGF-A antibody. Taken together, higher expression of Ang-2 contributes to the pathophysiology of RVO, providing a possible mechanism for the efficacy of BsAb in suppressing retinal dysfunction in RVO.
Collapse
Affiliation(s)
- Aika Kuriyama
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Shinsuke Nakamura
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Yuta Inokuchi
- Product Research Dept., Chugai Pharmaceutical Co., Ltd. Kanagawa, Japan
| | - Hiroto Abe
- Product Research Dept., Chugai Pharmaceutical Co., Ltd. Kanagawa, Japan
| | - Hiroto Yasuda
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Yae Hidaka
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Koki Nagaoka
- Product Research Dept., Chugai Pharmaceutical Co., Ltd. Kanagawa, Japan
| | - Tetsuhiro Soeda
- Product Research Dept., Chugai Pharmaceutical Co., Ltd. Kanagawa, Japan
| | - Masamitsu Shimazawa
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Hideaki Hara
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan.
| |
Collapse
|
6
|
Xu X, Li X, Tang Q, Zhang Y, Zhang L, Zhang M. Exploring laser-induced acute and chronic retinal vein occlusion mouse models: Development, temporal in vivo imaging, and application perspectives. PLoS One 2024; 19:e0305741. [PMID: 38885229 PMCID: PMC11182531 DOI: 10.1371/journal.pone.0305741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 06/03/2024] [Indexed: 06/20/2024] Open
Abstract
Photodynamic venous occlusion is a commonly accepted method for establishing mouse models of retinal vein occlusion (RVO). However, existing model parameters do not distinguish between acute and chronic RVO subtypes. Large variations in laser energy seem to correlate with fluctuating retinopathy severity and high rates of venous recanalization during the acute phase, along with the variable levels of retinal perfusion during the chronic phase. After optimizing the modeling procedure and defining success and exclusion criteria, laser energy groups of 80mW, 100mW, and 120mW were established. Multimodal imaging confirmed that higher energy levels increased the incidence of retinal cystoid edema and intraretinal hemorrhage, exacerbated the severity of exudative retinal detachment, and reduced the venous recanalization rate. For the acute model, 100mW was considered an appropriate parameter for balancing moderate retinopathy and venous recanalization. Continuous imaging follow-up revealed that day 1 after RVO was the optimal observation point for peaking of retinal thickness and intensive occurrence of retinal cystic edema and intraretinal hemorrhage. After excluding the influence of venous recanalization on retinal thickness, acute retinal edema demonstrated a positive response to standard anti-vascular endothelial growth factor therapy, validating the clinical relevance of the acute RVO model for further study in pathogenic mechanisms and therapeutic efficacy. For the chronic model, the 120mW parameter with the lowest venous recanalization rate was applied, accompanied by an increase in both photocoagulation shots and range to ensure sustained vein occlusion. Imaging follow-up clarified non-ischemic retinopathy characterized by tortuosity and dilation of the distal end, branches, and adjacent veins of the occluded vein. These morphological changes are quantifiable and could be combined with electrophysiological functional assessment for treatment effectiveness evaluation. Moreover, the stable state of venous occlusion may facilitate investigations into response and compensation mechanisms under conditions of chronic retinal hypoperfusion.
Collapse
Affiliation(s)
- Xiaowei Xu
- Department of Ophthalmology and Research Laboratory of Macular Disease, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xun Li
- Department of Ophthalmology and Research Laboratory of Macular Disease, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qingqing Tang
- Department of Ophthalmology and Research Laboratory of Macular Disease, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yi Zhang
- Department of Ophthalmology and Research Laboratory of Macular Disease, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Li Zhang
- Department of Ophthalmology and Research Laboratory of Macular Disease, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Meixia Zhang
- Department of Ophthalmology and Research Laboratory of Macular Disease, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
7
|
Nakamura S, Yamamoto R, Matsuda T, Yasuda H, Nishinaka A, Takahashi K, Inoue Y, Kuromitsu S, Shimazawa M, Goto M, Narumiya S, Hara H. Sphingosine-1-phosphate receptor 1/5 selective agonist alleviates ocular vascular pathologies. Sci Rep 2024; 14:9700. [PMID: 38678148 PMCID: PMC11055896 DOI: 10.1038/s41598-024-60540-6] [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: 10/12/2023] [Accepted: 04/24/2024] [Indexed: 04/29/2024] Open
Abstract
Ocular abnormal angiogenesis and edema are featured in several ocular diseases. S1P signaling via S1P1 likely is part of the negative feedback mechanism necessary to maintain vascular health. In this study, we conducted pharmacological experiments to determine whether ASP4058, a sphingosine 1-phosphate receptor 1/5 (S1P1/5) agonist, is useful in abnormal vascular pathology in the eye. First, human retinal microvascular endothelial cells (HRMECs) were examined using vascular endothelial growth factor (VEGF)-induced cell proliferation and hyperpermeability. ASP4058 showed high affinity and inhibited VEGF-induced proliferation and hyperpermeability of HRMECs. Furthermore, S1P1 expression and localization changes were examined in the murine laser-induced choroidal neovascularization (CNV) model, a mouse model of exudative age-related macular degeneration, and the efficacy of ASP4058 was verified. In the CNV model mice, S1P1 tended to decrease in expression immediately after laser irradiation and colocalized with endothelial cells and Müller glial cells. Oral administration of ASP4058 also suppressed vascular hyperpermeability and CNV, and the effect was comparable to that of the intravitreal administration of aflibercept, an anti-VEGF drug. Next, efficacy was also examined in a retinal vein occlusion (RVO) model in which retinal vascular permeability was increased. ASP4058 dose-dependently suppressed the intraretinal edema. In addition, it suppressed the expansion of the perfusion area observed in the RVO model. ASP4058 also suppressed the production of VEGF in the eye. Collectively, ASP4058 can be a potential therapeutic agent that normalizes abnormal vascular pathology, such as age-related macular degeneration and RVO, through its direct action on endothelial cells.
Collapse
Affiliation(s)
- Shinsuke Nakamura
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu, 501-1196, Japan
| | - Rie Yamamoto
- Discovery Accelerator, Astellas Pharma Inc., Tsukuba, Japan
- Alliance Laboratory for Advanced Medical Research, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takaya Matsuda
- Pharmaceutical Research and Technology Labs, Astellas Pharma Inc., Yaizu, Japan
| | - Hiroto Yasuda
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu, 501-1196, Japan
| | - Anri Nishinaka
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu, 501-1196, Japan
| | - Kei Takahashi
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu, 501-1196, Japan
| | - Yuki Inoue
- Astellas Institute for Regenerative Medicine, Marlborough, MA, USA
| | - Sadao Kuromitsu
- Discovery Accelerator, Astellas Pharma Inc., Tsukuba, Japan
- Alliance Laboratory for Advanced Medical Research, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Masamitsu Shimazawa
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu, 501-1196, Japan
| | - Masahide Goto
- Astellas Institute for Regenerative Medicine, Marlborough, MA, USA
| | - Shuh Narumiya
- Department of Drug Discovery Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hideaki Hara
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu, 501-1196, Japan.
| |
Collapse
|
8
|
Fuma S, Hidaka Y, Nishinaka A, Yasuda H, Aoshima K, Nakamura S, Hara H, Shimazawa M. Timolol maleate, a β blocker eye drop, improved edema in a retinal vein occlusion model. Mol Vis 2023; 29:188-196. [PMID: 38222457 PMCID: PMC10784217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 10/10/2023] [Indexed: 01/16/2024] Open
Abstract
Purpose To investigate the therapeutic effects of eye drops, namely, timolol maleate, a β-adrenergic receptor antagonist, and latanoprost, a prostaglandin F2α analog, on retinal edema in a murine retinal vein occlusion (RVO) model. Methods An RVO model was established using laser-induced RVO in mice, which were administered timolol maleate and latanoprost eye drops several times after venous occlusion. Subsequently, the thickness of the inner nuclear layer (INL) and the expression levels of such genes as Vegf and Atf4, which are stress markers of the endoplasmic reticulum, were examined. Primary human cultured retinal microvascular endothelial cells (HRMECs) were treated with timolol under hypoxic conditions, after which the gene expression pattern was investigated. Importantly, an integrated stress response inhibitor (ISRIB) was used in the RVO model, he known ISRIB, which suppresses the expression of ATF4 in retinal edema. Results Increased INL thickness was suppressed by timolol eye drops, as were the expressions of Vegf and Atf4, in the RVO model. However, latanoprost eye drops did not induce any change in INL thickness. In HRMECs, hypoxic stress and serum deprivation increased the Vegf and Atf4 expressions; in response, treatment with timolol suppressed the Vegf expression. Furthermore, the ISRIB decreased the Vegf expression pattern and edema formation, which are associated with RVO. Conclusions These results indicate that timolol eye drops may be a potential option for RVO treatment.
Collapse
Affiliation(s)
- Shinichiro Fuma
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Yae Hidaka
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Anri Nishinaka
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Hiroto Yasuda
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Kota Aoshima
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Shinsuke Nakamura
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Hideaki Hara
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
- Laboratory of Collaborative research for Innovative Drug Discovery, Gifu Pharmaceutical University, Gifu, Japan
| | - Masamitsu Shimazawa
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
- Laboratory of Collaborative research for Innovative Drug Discovery, Gifu Pharmaceutical University, Gifu, Japan
| |
Collapse
|
9
|
Bora K, Kushwah N, Maurya M, Pavlovich MC, Wang Z, Chen J. Assessment of Inner Blood-Retinal Barrier: Animal Models and Methods. Cells 2023; 12:2443. [PMID: 37887287 PMCID: PMC10605292 DOI: 10.3390/cells12202443] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 10/07/2023] [Accepted: 10/08/2023] [Indexed: 10/28/2023] Open
Abstract
Proper functioning of the neural retina relies on the unique retinal environment regulated by the blood-retinal barrier (BRB), which restricts the passage of solutes, fluids, and toxic substances. BRB impairment occurs in many retinal vascular diseases and the breakdown of BRB significantly contributes to disease pathology. Understanding the different molecular constituents and signaling pathways involved in BRB development and maintenance is therefore crucial in developing treatment modalities. This review summarizes the major molecular signaling pathways involved in inner BRB (iBRB) formation and maintenance, and representative animal models of eye diseases with retinal vascular leakage. Studies on Wnt/β-catenin signaling are highlighted, which is critical for retinal and brain vascular angiogenesis and barriergenesis. Moreover, multiple in vivo and in vitro methods for the detection and analysis of vascular leakage are described, along with their advantages and limitations. These pre-clinical animal models and methods for assessing iBRB provide valuable experimental tools in delineating the molecular mechanisms of retinal vascular diseases and evaluating therapeutic drugs.
Collapse
Affiliation(s)
| | | | | | | | | | - Jing Chen
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| |
Collapse
|
10
|
Avrutsky MI, Chen CW, Lawson JM, Snipas SJ, Salvesen GS, Troy CM. Caspase-9 inhibition confers stronger neuronal and vascular protection compared to VEGF neutralization in a mouse model of retinal vein occlusion. Front Neurosci 2023; 17:1209527. [PMID: 37449272 PMCID: PMC10336837 DOI: 10.3389/fnins.2023.1209527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 06/05/2023] [Indexed: 07/18/2023] Open
Abstract
Purpose Retinal vein occlusion (RVO) is a sight-threatening condition typically treated with intravitreal injection of vascular endothelial growth factor (VEGF) antagonists. Treatment response to anti-VEGF therapies is highly variable, with poor visual outcomes and treatment response in patients with significant retinal nonperfusion following RVO. Recently, caspase-9 has been identified as a potent regulator of edema, gliosis, and neuronal dysfunction during acute retinal hypoxia. The purpose of this study was to compare the therapeutic effect of caspase-9 inhibition against VEGF-neutralization in an established mouse model of RVO. Methods Adult male C57Bl/6 J mice were randomized to induction of RVO and treatment with either vehicle, intravitreal injection of anti-VEGF antibody, topical administration of a selective caspase-9 inhibitor (Pen1-XBir3), or a combination therapy. Animals were followed on days 1, 2, and 8 after RVO with fundus retinal imaging, and with optical coherence tomography (OCT) to capture retinal swelling, capillary nonperfusion (measured by disorganization of retinal inner layers, DRIL), hyperreflective foci (HRF), and retinal atrophy. Focal electroretinography (ERG) measurements were performed on day 7. Histology was performed on retinal sections from day 8. Results Both VEGF neutralization and caspase-9 inhibition showed significant retinal protection from RVO compared to vehicle treatment arm. Retinal reperfusion of occluded veins was accelerated in eyes receiving caspase-9 inhibitor, but not significantly different from vehicle in the anti-VEGF group. Retinal edema was suppressed in all treatment groups, with approximately 2-fold greater edema reduction with caspase-9 inhibition compared to VEGF neutralization. HRF were reduced similarly across all treatment groups compared to vehicle. Retinal detachment was reduced only in eyes treated with caspase-9 inhibitor monotherapy. Caspase-9 inhibition reduced retinal atrophy and preserved ERG response; VEGF neutralization did not prevent neurodegeneration following RVO. Conclusion Caspase-9 inhibition confers stronger neuronal and vascular protection compared to VEGF neutralization in the mouse laser-induced model of RVO.
Collapse
Affiliation(s)
- Maria I. Avrutsky
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, United States
| | - Claire W. Chen
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, United States
| | - Jacqueline M. Lawson
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, United States
| | - Scott J. Snipas
- NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
| | - Guy S. Salvesen
- NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
| | - Carol M. Troy
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, United States
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, United States
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, United States
| |
Collapse
|
11
|
TRPV4 channels promote vascular permeability in retinal vascular disease. Exp Eye Res 2023; 228:109405. [PMID: 36773739 DOI: 10.1016/j.exer.2023.109405] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 01/06/2023] [Accepted: 02/02/2023] [Indexed: 02/11/2023]
Abstract
This study aimed to determine the role of transient receptor potential vanilloid 4 (TRPV4), a calcium (Ca2+)-permeable cation channel, in the pathophysiology of retinal vascular disease. The retinal vein occlusion (RVO) murine model was created by irradiating retinal veins using lasers. TRPV4 expression and localization were evaluated in RVO mice retinas. In addition, we examined the effects of TRPV4 antagonists (RQ-00317310, HC-067047, GSK2193874, and GSK2798745) on retinal edema, blood flow, and ischemic areas in RVO mice. Furthermore, changes in the retinal expression of tumor necrosis factor (TNF)-α and aquaporin4 (AQP4) by RQ-00317310 were analyzed using Western blot. We also assessed the barrier integrity of epithelial cell monolayers using trans-endothelial electrical resistance (TEER) in Human Retinal Microvascular Endothelial Cells (HRMECs). The expression of TRPV4 was significantly increased and co-localized with glutamine synthetase (GS), a Müller glial marker, in the ganglion cell layer (GCL) of the RVO mice. Moreover, RQ-00317310 administration ameliorated the development of retinal edema and ischemia in RVO mice. In addition, the up regulation of TNF-α and down-regulation of AQP4 were lessened by the treatment with RQ-00317310. Treatment with GSK1016790A, a TRPV4 agonist, increased vascular permeability, while RQ-00317310 treatment decreased vascular endothelial growth factor (VEGF)- or TRPV4-induced retinal vascular hyperpermeability in HRMECs. These findings suggest that TRPV4 plays a role in the development of retinal edema and ischemia. Thus, TRPV4 could be a new therapeutic target against the pathological symptoms of retinal vascular diseases.
Collapse
|
12
|
Neurovascular injury associated non-apoptotic endothelial caspase-9 and astroglial caspase-9 mediate inflammation and contrast sensitivity decline. Cell Death Dis 2022; 13:937. [PMID: 36347836 PMCID: PMC9643361 DOI: 10.1038/s41419-022-05387-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/25/2022] [Accepted: 10/27/2022] [Indexed: 11/11/2022]
Abstract
Retinal neurovascular injuries are a leading cause of vision loss in young adults presenting unmet therapeutic needs. Neurovascular injuries damage homeostatic communication between endothelial, pericyte, glial, and neuronal cells through signaling pathways that remain to be established. To understand the mechanisms that contribute to neuronal death, we use a mouse model of retinal vein occlusion (RVO). Using this model, we previously discovered that after vascular damage, there was non-apoptotic activation of endothelial caspase-9 (EC Casp9); knock-out of EC Casp9 led to a decrease in retinal edema, capillary ischemia, and neuronal death. In this study, we aimed to explore the role of EC Casp9 in vision loss and inflammation. We found that EC Casp9 is implicated in contrast sensitivity decline, induction of inflammatory cytokines, and glial reactivity. One of the noted glial changes was increased levels of astroglial cl-caspase-6, which we found to be activated cell intrinsically by astroglial caspase-9 (Astro Casp9). Lastly, we discovered that Astro Casp9 contributes to capillary ischemia and contrast sensitivity decline after RVO (P-RVO). These findings reveal specific endothelial and astroglial non-apoptotic caspase-9 roles in inflammation and neurovascular injury respectively; and concomitant relevancy to contrast sensitivity decline.
Collapse
|
13
|
Nakamura S, Nishinaka A, Hidaka Y, Shimazawa M, Thomas L, Bakker RA, Hara H. Efficacy of an Anti-Semaphorin 3A Neutralizing Antibody in a Male Experimental Retinal Vein Occlusion Mouse Model. Invest Ophthalmol Vis Sci 2022; 63:14. [PMID: 35822950 PMCID: PMC9288153 DOI: 10.1167/iovs.63.8.14] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose Semaphorin 3A (Sema3A) is a promising therapeutic target for macular edema in age-related macular degeneration, diabetic retinopathy, and retinal vein occlusion (RVO). Anti-vascular endothelial growth factors (anti-VEGFs) are the current standard of care for many retinal diseases. This study investigated the Sema3A neutralizing antibody BI-X and/or anti-VEGF therapy (aflibercept) in an RVO mouse model. Treatment efficacy was examined and grouped by timing subsequent to the RVO mouse model induction: efficacy against the onset of intraretinal edema 1 day postinduction and protective effects at 7 days postinduction. Methods We examined the changes in expression of Sema3A in the retina of an RVO mouse model. In addition, changes in expression of tumor necrosis factor (TNF)-α and semaphorin-related proteins (neuropilin-1 and plexin A1) in the retina upon treatment were analyzed by Western blotting. The effects of BI-X and/or aflibercept were evaluated using measures of retinal edema, blood flow, and thinning of the inner nuclear layer. Results Induction of vein occlusion in the RVO mouse model significantly increased Sema3A expression in the retina, particularly in the inner nuclear layer. BI-X was effective as a monotherapy and in combination with anti-VEGF therapy, demonstrating a beneficial effect on intraretinal edema and retinal blood flow. Moreover, in the RVO mouse model, BI-X monotherapy normalized the changes in expression of TNF-α and semaphorin-related proteins. Conclusions These findings support targeting Sema3A to treat intraretinal edema and retinal ischemia.
Collapse
Affiliation(s)
- Shinsuke Nakamura
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Anri Nishinaka
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Yae Hidaka
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Masamitsu Shimazawa
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Leo Thomas
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach and der Riß, Germany
| | - Remko A Bakker
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach and der Riß, Germany
| | - Hideaki Hara
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| |
Collapse
|
14
|
Nishinaka A, Tanaka M, Aoshima K, Kuriyama A, Sasaki T, Otsu W, Yasuda H, Nakamura S, Shimazawa M, Hara H. The pathological association between the anterior eye segment and the retina in a murine model of neovascular glaucoma. FASEB J 2022; 36:e22323. [PMID: 35485981 DOI: 10.1096/fj.202101917r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 03/30/2022] [Accepted: 04/07/2022] [Indexed: 11/11/2022]
Abstract
Neovascular glaucoma (NVG) is caused by the formation of new blood vessels in the angle, iris, and cornea in retinal ischemic disease, such as proliferative diabetic retinopathy (PDR) and retinal vein occlusion (RVO), which can reduce the visual acuity. However, the pathophysiological symptoms of NVG are still not well understood because there is no model for the formation of NVG in the angle, iris, and cornea. The aim of this study was to investigate the involvement of NVG during ischemic disease, in a murine model of retinal ischemia. We evaluated the changes of the intraocular pressure (IOP) and pathological symptoms in the anterior eye segment and retina in this model, and the changes in the RNA or protein expression of vascular endothelial growth factor (VEGF) and fibrosis-related factors were analyzed in the retina and cornea by quantitative real-time polymerase chain reaction or western blot, respectively. Furthermore, we examined the changes in IOP after intravitreal injection of an anti-VEGF antibody. First, NVG formed in the retinal ischemic murine model, and the IOP was elevated in mice with NVG formation. Interestingly, VEGF expression was decreased in the retina but increased in the cornea in the murine model of NVG. On the other hand, fibrosis-related factors were increased in the retina and also significantly increased in the cornea in NVG. Moreover, the administration of anti-VEGF antibody immediately after vessel occlusion suppressed the increase in IOP, but administration at 7 days after vessel occlusion accelerated the increase in IOP. These findings suggest that the formation of NVG may be correlated with the pathological symptoms of retinal ischemic disease, via changes in VEGF and fibrosis-related factor expression.
Collapse
Affiliation(s)
- Anri Nishinaka
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Miruto Tanaka
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Kota Aoshima
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Aika Kuriyama
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Takahiro Sasaki
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Wataru Otsu
- Department of Biomedical Research Laboratory, Gifu Pharmaceutical University, Gifu, Japan
| | - Hiroto Yasuda
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Shinsuke Nakamura
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Masamitsu Shimazawa
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan.,Department of Biomedical Research Laboratory, Gifu Pharmaceutical University, Gifu, Japan
| | - Hideaki Hara
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| |
Collapse
|
15
|
Shi R, Guo Z, Yang X, Che X. Aggravation of retinal hard exudates after intravitreal anti-vascular endothelial growth factor therapy for cystoid macular edema and the risk factors: a retrospective study. BMC Ophthalmol 2022; 22:92. [PMID: 35197011 PMCID: PMC8867860 DOI: 10.1186/s12886-022-02315-z] [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: 03/26/2021] [Accepted: 01/27/2022] [Indexed: 12/03/2022] Open
Abstract
Background/aims To evaluate retinal hard exudates (HEs) progression in patients with cystoid macular edema (CME) secondary to diabetic retinopathy (DR) or branch retinal vascular occlusion (BRVO) after intravitreal injections of ranibizumab (IVR) treatment and identify the risk factors for the deterioration of HEs. Methods This retrospective study enrolled 288 eyes with center-involving CME secondary to DR or BRVO from 288 patients (one eye per patient). All patients were treated with three loading doses of ranibizumab intravitreally at a monthly interval. The morphologic features of HEs were observed, and the HEs areas were quantified using a semi-automatic method at baseline, 1 month after the first dose of IVR and 1 month after the third dose of IVR therapy. HEs progression was defined as having a > =2-grade increase in the HEs severity scale. The best-corrected vision acuity (BCVA) and alterations in HEs areas were compared between DR and BRVO groups. And logistic regression analyses were used to identify the risk factors for HEs exacerbation. Results Morphological changes of retinal HEs occurred in all eyes after IVR therapy, although HEs area was not significantly changed in some eyes. DR group has a higher percentage of eyes with progressed HEs area than the BRVO groups (34.9% vs. 21.8%, P = 0.019) 1 month after the first dose of IVR. Both DR and BRVO groups had a decreased percentage of enlarged HEs 1 month after the third injection, but the DR group is still higher than the BRVO group (17.1% vs. 8.4%, P = 0.027). At baseline, there was no correlation between VA and HEs areas. After the first and third doses of IVR, there still was no consistent correlation between HEs severity and change in VA over time. Furthermore, CME with subretinal fluid (SRF) is associated with a higher risk of HEs progression (P = 0.001). Long CME duration and high serum low-density lipoprotein cholesterol (LDL-C) level were identified as risk factors for HEs progression following IVR treatment in both univariable and multivariable regression analyses (Odds ratio (OR) = 1.88, P = 0.012 and OR = 1.14, P = 0.021, respectively). Conclusions Alterations in the area of retinal HEs are widely observed after IVR treatment for CME. The eyes with CME secondary to DR have a higher percentage of progressed HEs than the BRVO eyes. DME with SRF, extended duration of CME, and high LDL-C level are potential risk factors of deteriorated HEs after IVR treatment.
Collapse
Affiliation(s)
- Rui Shi
- Department of Ophthalmology, Shaanxi Provincial People's Hospital, Xi'an, 710068, China
| | - Zhonglan Guo
- Department of Ophthalmology, Shaanxi Provincial People's Hospital, Xi'an, 710068, China
| | - Xiangxiang Yang
- Department of Ophthalmology, Shaanxi Provincial People's Hospital, Xi'an, 710068, China
| | - Xuanyi Che
- Department of Ophthalmology, Shaanxi Provincial People's Hospital, Xi'an, 710068, China.
| |
Collapse
|
16
|
Nakamura S, Hara H. [Prospects and Challenges of Anti-VEGF Drug Treatment for Pathological Angiogenesis of the Retina]. YAKUGAKU ZASSHI 2021; 141:1307-1317. [PMID: 34853203 DOI: 10.1248/yakushi.21-00158-1] [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: 11/22/2022]
Abstract
The number of patients with exudative age-related macular degeneration, diabetic retinopathy and retinal vein occlusion is expected to rise in proportion with the aging of the population and increasing diabetes patients. Also, they are the most common diseases caused by intraocular neovascularization and are often difficult to treat. Currently, anti-vascular endothelial growth factor (VEGF) therapy has been developed and has demonstrated excellent results in treating macular edema, and many patients have avoided blindness. Unfortunately, there are problems with cases that do not respond to the anti-VEGF drugs and complications of administration. It is necessary to deepen the understanding of the physiological and pathological retinal roles of VEGF and to optimize the anti-VEGF therapy. There are also no drugs indicated for the regression of neovascularization itself. The solution to this problem is to develop novel therapies targeting other than VEGF. In this symposium review, we introduce the roles of VEGF in the ischemic retina and anti-angiogenic factors as promising therapeutic targets.
Collapse
Affiliation(s)
- Shinsuke Nakamura
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University
| | - Hideaki Hara
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University
| |
Collapse
|
17
|
Nakazawa Y, Nagai N. [Eye Diseases with Aging -for Keeping High QOV (Quality of Vision)]. YAKUGAKU ZASSHI 2021; 141:1305-1306. [PMID: 34853202 DOI: 10.1248/yakushi.21-00158-f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
18
|
Nishinaka A, Nakamura S, Tanaka M, Masuda T, Inoue Y, Yamamoto T, Imai T, Hidaka Y, Shimazawa M, Hara H. Excess adiponectin in eyes with progressive ocular vascular diseases. FASEB J 2021; 35:e21313. [PMID: 33484194 DOI: 10.1096/fj.202001740rr] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 12/11/2020] [Accepted: 12/14/2020] [Indexed: 11/11/2022]
Abstract
Anti-vascular endothelial growth factor (VEGF) therapies are now the first-line treatment for many ocular diseases, but some patients are non-responders to these therapies. The purpose of this study was to determine whether the level of adiponectin increased the pathogenesis of retinal edema and neovascularization in the retina of progressive ocular vascular diseases. We examined the role played by adiponectin in two types of cells and animal models which are retinal vein occlusion (RVO) and oxygen-induced retinopathy (OIR) mice. Our results showed that an injection of anti-adiponectin antibody ameliorated the retinal edema and ischemia through the depression of the expression level of VEGF-related factors and tight junction-related proteins in the retina of RVO mice. The intravitreal injection of anti-adiponectin antibody also decreased the degree of retinal neovascularization in an OIR mice. In addition, exposure of human retinal microvascular endothelial cells and human brain microvascular pericytes in culture to adiponectin increased both the vascular permeability and neovascularization through the increase of inflammatory factor and the dropout of the pericytes. These findings indicate that adiponectin plays a critical role in retinal edema and neovascularization, and adiponectin is a potential therapeutic target for the treatment of diabetic macular edema, proliferative diabetic retinopathy, and RVO.
Collapse
Affiliation(s)
- Anri Nishinaka
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Shinsuke Nakamura
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Miruto Tanaka
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Tomomi Masuda
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Yuki Inoue
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Takumi Yamamoto
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Takahiko Imai
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Yae Hidaka
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Masamitsu Shimazawa
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Hideaki Hara
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| |
Collapse
|
19
|
Yamamoto T, Nitta K, Nishinaka A, Tanaka M, Nakamura S, Shimazawa M, Hara H. Oral administration of NSP-116, a free radical scavenger, suppresses the symptoms of retinal vein occlusion in the murine model. Exp Eye Res 2021; 204:108453. [PMID: 33503450 DOI: 10.1016/j.exer.2021.108453] [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: 09/15/2020] [Revised: 01/09/2021] [Accepted: 01/15/2021] [Indexed: 11/16/2022]
Abstract
Retinal vein occlusion (RVO) is an intractable eye disease that results in reduced visual acuity, associated with retinal ischemia, hemorrhage, and edema. RVO results in excessive ROS production in the retina, causing inflammation and retinal edema. A free radical scavenger, 4-(4-acetylpiperazin-1-yl)-2-(1H-imidazole-1-yl) aniline (NSP-116), has been reported to demonstrate antioxidative effects and prevent ROS production in the retina. Therefore, NSP-116 may represent a useful drug for treating the pathological symptoms of RVO, such as retinal edema and ischemic symptoms. This study aimed to investigate the effects of NSP-116 in a murine model of RVO. We evaluated the thickness of the retinal layer and the size of the non-perfused area following the oral administration of NSP-116. Moreover, we used western blot analysis to examine the expression levels of vascular endothelial growth factor (VEGF) and tumor necrosis factor (TNF)-α, after NSP-116 administration, and examined the localization of 8-hydroxy-2'-deoxyguanosine (8-OHdG), by immunostaining. The findings indicate that NSP-116 suppressed retinal edema and expansion the non-perfused area by suppressing the increased expression of VEGF, TNF-α, and 8-OHdG in the murine RVO model. In conclusion, the oral administration of NSP-116 may serve as an effective pharmacological treatment for the pathological symptoms of RVO.
Collapse
Affiliation(s)
- Takumi Yamamoto
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Koki Nitta
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Anri Nishinaka
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Miruto Tanaka
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Shinsuke Nakamura
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan.
| | - Masamitsu Shimazawa
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Hideaki Hara
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| |
Collapse
|
20
|
Establishment of a pigmented murine model abundant with characteristics of retinal vein occlusion. Exp Eye Res 2021; 204:108441. [PMID: 33453278 DOI: 10.1016/j.exer.2021.108441] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 12/21/2020] [Accepted: 01/06/2021] [Indexed: 02/07/2023]
Abstract
Retinal vein occlusion (RVO) is a vascular disease that represents characteristic retinal hemorrhage and dilated retinal veins. Despite its clinical importance, its pathogenesis remains largely unknown because of limited opportunities to acquire human retinal samples. Therefore, an animal model that reproduces the clinical features of RVO patients is required for further investigation. In this study, we established a pigmented murine RVO model that reproduced characteristic fundus appearances similar to human RVO findings. Retinal edema in this model was observed in both optical coherence tomography and histological analysis, which is a clinically important outcome. With quantitative real-time PCR analysis on retinal samples, we revealed that the mRNA level of vascular endothelial growth factor (VEGF) increased in the retina induced RVO. Moreover, this retinal edema was reduced by intravitreal injection of anti-VEGF antibody. These results were consistent with human clinical knowledge and suggested that this model could be a useful tool for research into new therapeutic approaches.
Collapse
|
21
|
Takahashi H, Nakagawa K, Yamada H, Mori H, Oba S, Toyama K, Takahashi K. Time course of collateral vessel formation after retinal vein occlusion visualized by OCTA and elucidation of factors in their formation. Heliyon 2021; 7:e05902. [PMID: 33474512 PMCID: PMC7803649 DOI: 10.1016/j.heliyon.2021.e05902] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 12/01/2020] [Accepted: 12/31/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND It is clinically recognized that collateral vessels can form after retinal vein occlusion (RVO) in some cases and these vessels can lead to spontaneous recovery of the pathological condition. In recent years, optical coherence tomography angiography (OCTA) has become a decisive clinical instrument. Unlike previous angiography tests, OCTA enables the non-invasive visualization of fundus vasculature without the need for administration of a contrast agent. However, it remains to be determined if OCTA depicts the 'true' histological status as several studies have reported artifacts in OCTA imaging. METHODS We generated a laser-induced mouse RVO model, and evaluated the subsequent formation of collateral vessels in order to understand the mechanisms by which collateral vessels form using OCTA imaging, as well as molecular and histological assessments. RESULTS We succeeded in visualizing the time course of collateral vessel formation in a mouse RVO model and confirmed the similarity in formation of collateral vessels only within the deep layer of the retina in both human and mouse. We hypothesized that sphingosine 1-phosphate receptor-1 (S1PR1) may play important roles via vascular shear stress linking vein occlusion and collateral vessel formation. Results from OCTA revealed that collateral vessels are increased in response to administration of a S1PR1 agonist in a mouse RVO model. Based on quantitative reverse transcription polymerase chain reaction (qRT-PCR), S1PR1 messenger ribonucleic acid (mRNA) levels in the whole retina peaked 6 h after photocoagulation in this model. Immunohistochemical staining of retinal flat mounts revealed that S1PR1 staining occurred along the laser-occluded blood vessels. CONCLUSION We observed the temporal process of collateral vessel formation in a mouse RVO model and identified the relationship between S1PR1 and shear stress as one of the factors in collateral vessel formation in RVO.
Collapse
Affiliation(s)
- Hajime Takahashi
- Department of Ophthalmology, Kansai Medical University, Hirakata, Osaka, Japan
| | - Kazuki Nakagawa
- Department of Ophthalmology, Kansai Medical University, Hirakata, Osaka, Japan
| | - Haruhiko Yamada
- Department of Ophthalmology, Kansai Medical University, Hirakata, Osaka, Japan
| | - Hidetsugu Mori
- Department of Ophthalmology, Kansai Medical University, Hirakata, Osaka, Japan
| | - Shimpei Oba
- Department of Ophthalmology, Kansai Medical University, Hirakata, Osaka, Japan
| | - Keiko Toyama
- Department of Ophthalmology, Kansai Medical University, Hirakata, Osaka, Japan
| | - Kanji Takahashi
- Department of Ophthalmology, Kansai Medical University, Hirakata, Osaka, Japan
| |
Collapse
|
22
|
Luo Y, Wan J, Luo C, Liu H, Zhou Y, Chen J, Duan Y, Ning X, Zhou Z, Wang K, Li J, Xu H, Chen Z. Higher Aqueous Levels of Resistin and Lipocalin-2 Indicated Worse Visual Improvement following anti-VEGF Therapy in Patients with Retinal Vein Occlusion. Curr Eye Res 2020; 46:845-854. [PMID: 33103481 DOI: 10.1080/02713683.2020.1842462] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
PURPOSE The aim of this study was to investigate the aqueous humor levels of elastase-2, lactoferrin, lipocalin-2 (LCN-2), resistin, and thrombospondin-1 (TSP-1) in patients with retinal vein occlusion (RVO) and their relationship with visual prognosis following intravitreal anti-vascular endothelial growth factor (VEGF) therapy. MATERIALS AND METHODS 52 RVO patients (23 cases of central retinal vein occlusion (CRVO) and 29 cases of branch retinal vein occlusion (BRVO)) and 20 cases of senile cataract were enrolled in this study. All RVO patients underwent fundus examinations before and 6-8 months after intravitreal anti-VEGF treatment. Five milliliters of blood were collected from RVO patients before treatment for the measurement of lipids and coagulation factors. Sixty microliters of aqueous humor were collected during intravitreal injection of anti-VEGF or during cataract surgery. The levels of elastase-2, lactoferrin, LCN-2, resistin, and TSP-1 in aqueous humor were determined by Luminex xMAP multiple analysis. RESULTS The aqueous levels of resistin and LCN-2 were significantly higher but the level of TSP-1 was significantly lower in RVO patients compared to controls. Further, sub-group analysis showed that CRVO patients had significantly higher levels of resistin and LCN-2 than controls. The aqueous levels of resistin and LCN-2 were negatively correlated with visual improvement following anti-VEGF therapy in CRVO but not in BRVO patients. Visual improvement in RVO patients was not associated with blood lipid levels or any of the coagulation factors. CONCLUSION CRVO patients had significantly higher aqueous levels of resistin and LCN-2, which negatively impacted on visual improvement after anti-VEGF therapy.
Collapse
Affiliation(s)
- Yong Luo
- Aier School of Ophthalmology, Central South University, Changsha, Hunan Province, China.,Aier Eye Hospital of Changsha, Changsha, Hunan Province, China
| | - Jianbo Wan
- Aier Eye Hospital of Changsha, Changsha, Hunan Province, China
| | - Chang Luo
- Aier School of Ophthalmology, Central South University, Changsha, Hunan Province, China.,Aier Eye Institute, Changsha, Hunan, China
| | - HengWei Liu
- Aier School of Ophthalmology, Central South University, Changsha, Hunan Province, China
| | - YuFan Zhou
- Aier School of Ophthalmology, Central South University, Changsha, Hunan Province, China
| | - Juan Chen
- Aier Eye Hospital of Changsha, Changsha, Hunan Province, China
| | - Yiqin Duan
- Aier Eye Hospital of Changsha, Changsha, Hunan Province, China
| | - Xiangyan Ning
- Aier Eye Hospital of Changsha, Changsha, Hunan Province, China
| | - Zengchao Zhou
- Aier Eye Hospital of Changsha, Changsha, Hunan Province, China
| | - Kehua Wang
- Aier Eye Hospital of Changsha, Changsha, Hunan Province, China
| | - Jun Li
- Aier Eye Hospital of Changsha, Changsha, Hunan Province, China
| | - Heping Xu
- Aier School of Ophthalmology, Central South University, Changsha, Hunan Province, China.,Aier Institute of Optometry and Vision Science, Changsha, Hunan, China.,Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, UK
| | - ZhongPing Chen
- Aier School of Ophthalmology, Central South University, Changsha, Hunan Province, China.,Aier Eye Hospital of Changsha, Changsha, Hunan Province, China
| |
Collapse
|
23
|
Avrutsky MI, Ortiz CC, Johnson KV, Potenski AM, Chen CW, Lawson JM, White AJ, Yuen SK, Morales FN, Canepa E, Snipas S, Salvesen GS, Jean YY, Troy CM. Endothelial activation of caspase-9 promotes neurovascular injury in retinal vein occlusion. Nat Commun 2020; 11:3173. [PMID: 32576823 PMCID: PMC7311551 DOI: 10.1038/s41467-020-16902-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 06/02/2020] [Indexed: 12/27/2022] Open
Abstract
Central nervous system ischemic injury features neuronal dysfunction, inflammation and breakdown of vascular integrity. Here we show that activation of endothelial caspase-9 after hypoxia-ischemia is a critical event in subsequent dysfunction of the blood-retina barrier, using a panel of interrelated ophthalmic in vivo imaging measures in a mouse model of retinal vein occlusion (RVO). Rapid nonapoptotic activation of caspase-9 and its downstream effector caspase-7 in endothelial cells promotes capillary ischemia and retinal neurodegeneration. Topical eye-drop delivery of a highly selective caspase-9 inhibitor provides morphological and functional retinal protection. Inducible endothelial-specific caspase-9 deletion phenocopies this protection, with attenuated retinal edema, reduced inflammation and preserved neuroretinal morphology and function following RVO. These results reveal a non-apoptotic function of endothelial caspase-9 which regulates blood-retina barrier integrity and neuronal survival, and identify caspase-9 as a therapeutic target in neurovascular disease. Retinal vein occlusion can cause blindness, and features neuronal dysfunction, inflammation and breakdown of vascular integrity. Here the authors report a non-apoptotic role of endothelial caspase-9 in regulating blood-retina barrier integrity and neuronal survival, which can be therapeutically targeted in a mouse model of retinal vein occlusion.
Collapse
Affiliation(s)
- Maria I Avrutsky
- Department of Pathology & Cell Biology; Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| | - Crystal Colón Ortiz
- Department of Pathology & Cell Biology; Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| | - Kendra V Johnson
- Department of Pathology & Cell Biology; Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| | - Anna M Potenski
- Department of Pharmacology; Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| | - Claire W Chen
- Department of Pathology & Cell Biology; Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| | - Jacqueline M Lawson
- Department of Pathology & Cell Biology; Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| | - Alexandra J White
- Department of Pathology & Cell Biology; Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| | - Stephanie K Yuen
- Department of Pathology & Cell Biology; Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| | - Fatima N Morales
- Department of Pathology & Cell Biology; Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| | - Elisa Canepa
- Department of Pathology & Cell Biology; Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| | - Scott Snipas
- NCI-designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute La Jolla, La Jolla, CA, 92037, USA
| | - Guy S Salvesen
- NCI-designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute La Jolla, La Jolla, CA, 92037, USA
| | - Ying Y Jean
- Department of Pathology & Cell Biology; Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| | - Carol M Troy
- Department of Pathology & Cell Biology; Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA. .,Department of Neurology; Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA. .,The Taub Institute for Research on Alzheimer's Disease and the Aging Brain; Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA.
| |
Collapse
|
24
|
The Changes in Blood Flow Seen in the Eye after Foot Acupuncture Treatment in Mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:6405471. [PMID: 32328137 PMCID: PMC7165330 DOI: 10.1155/2020/6405471] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 02/27/2020] [Indexed: 11/26/2022]
Abstract
Acupuncture is used to treat a wide variety of eye diseases, although there is little evidence about the effects of acupuncture treatment and the mechanisms responsible for them. Foot acupuncture treatment has effects in both mice and humans. The purpose of this study was to investigate the effects of acupuncture treatment on ocular blood flow in mice. We evaluated ocular blood flow in C57BL/6J mice after foot acupuncture treatment using laser speckle flowgraphy. The mean blur rate, which is an index of blood flow velocity, was increased in the foot acupuncture group. Our results showed that, after 3 minutes' foot acupuncture, ocular blood flow was significantly increased in both the blood vessels and tissue of the eye in C57BL/6J mice. Thus, performing acupuncture in mice might help to determine its effects. Furthermore, acupuncture is considered to be a possible treatment for ocular disease.
Collapse
|
25
|
Wada T, Song Y, Oomae T, Sogawa K, Yoshioka T, Nakabayashi S, Takahashi K, Tani T, Ishibazawa A, Ishiko S, Yoshida A. Longitudinal Changes in Retinal Blood Flow in a Feline Retinal Vein Occlusion Model as Measured by Doppler Optical Coherence Tomography and Optical Coherence Tomography Angiography. Invest Ophthalmol Vis Sci 2020; 61:34. [PMID: 32084269 PMCID: PMC7326600 DOI: 10.1167/iovs.61.2.34] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose We aimed to observe longitudinal changes in retinal blood flow (RBF) and structural transformations in capillaries using Doppler optical coherence tomography (DOCT) and optical coherence tomography angiography (OCTA) in a feline retinal blood occlusion (RVO) model. Methods RVO was induced by argon green laser photocoagulation (PC) in six eyes of six cats. RBF was measured at a first-order retinal artery and vein by a DOCT flowmeter, and structural changes in the capillaries around the occluded vessels (12 × 12 and 3 × 3 mm) were assessed by OCTA before (at baseline); immediately after PC; and on days 1, 4, 7, and 14 thereafter. Systemic and ocular parameters were monitored during the observation period. Results There were no significant differences in any systemic or ocular parameters before and after PC. Arterial RBF increased significantly on day 1 (160.6 ± 8.6% vs. baseline, P < 0.001) and decreased below baseline after day 1 through 14. Venous RBF decreased immediately after PC (17.4 ± 9.6% vs. baseline, P = 0.001) and then gradually increased afterwards, but did not return to baseline. OCTA showed dilatation of retinal venules immediately after PC to day 1. Collateral vessels began to form on day 4, had matured by day 7, and were pruned on day 14, which formed as mature as normal retinal venule diameters. Conclusions With increasing arterial RBF within 1 day after inducing RVO, venules gradually expanded to form collateral vessels, suggesting that collateral vessels originate from existing capillary networks, not neovascularization.
Collapse
|
26
|
Darche M, Cossutta M, Caruana L, Houppe C, Gilles ME, Habert D, Guilloneau X, Vignaud L, Paques M, Courty J, Cascone I. Antagonist of nucleolin, N6L, inhibits neovascularization in mouse models of retinopathies. FASEB J 2020; 34:5851-5862. [PMID: 32141122 DOI: 10.1096/fj.201901876r] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 02/11/2020] [Accepted: 02/21/2020] [Indexed: 12/18/2022]
Abstract
Retinal vascular diseases (RVD) have been identified as a major cause of blindness worldwide. These pathologies, including the wet form of age-related macular degeneration, retinopathy of prematurity, and diabetic retinopathy are currently treated by intravitreal delivery of anti-vascular endothelial growth factor (VEGF) agents. However, repeated intravitreal injections can lead to ocular complications and resistance to these treatments. Thus, there is a need to find new targeted therapies. Nucleolin regulates the endothelial cell (EC) activation and angiogenesis. In previous studies, we designed a pseudopeptide, N6L, that binds the nucleolin and blocks the tumor angiogenesis. In this study, the effect of N6L was investigated in two experimental models of retinopathies including oxygen-induced retinopathy (OIR) and choroidal neovascularization (CNV). We found that in mouse OIR, intraperitoneal injection of N6L is delivered to activated ECs and induced a 50% reduction of pathological neovascularization. The anti-angiogenic effect of N6L has been tested in CNV model in which the systemic injection of N6L induced a 33% reduction of angiogenesis. This effect is comparable to those obtained with VEGF-trap, a standard of care drug for RVD. Interestingly, with preventive and curative treatments, neoangiogenesis is inhibited by 59%. Our results have potential interest in the development of new therapies targeting other molecules than VEGF for RVD.
Collapse
Affiliation(s)
- Marie Darche
- CRRET Laboratory, CNRS ERL 9215, University of Paris-Est Créteil, Créteil, France
- Clinical Investigation Center 1423, Centre Hospitalier National des Quinze-Vingts, Institut Hospitalo-Universitaire ForeSight, Sorbonne Université, Paris, France
| | - Mélissande Cossutta
- CRRET Laboratory, CNRS ERL 9215, University of Paris-Est Créteil, Créteil, France
| | - Laure Caruana
- CRRET Laboratory, CNRS ERL 9215, University of Paris-Est Créteil, Créteil, France
| | - Claire Houppe
- CRRET Laboratory, CNRS ERL 9215, University of Paris-Est Créteil, Créteil, France
| | | | - Damien Habert
- CRRET Laboratory, CNRS ERL 9215, University of Paris-Est Créteil, Créteil, France
| | - Xavier Guilloneau
- INSERM, CNRS, Institut de la Vision, Sorbonne Université, Paris, France
| | - Lucile Vignaud
- INSERM, CNRS, Institut de la Vision, Sorbonne Université, Paris, France
| | - Michel Paques
- Clinical Investigation Center 1423, Centre Hospitalier National des Quinze-Vingts, Institut Hospitalo-Universitaire ForeSight, Sorbonne Université, Paris, France
| | - José Courty
- CRRET Laboratory, CNRS ERL 9215, University of Paris-Est Créteil, Créteil, France
| | - Ilaria Cascone
- CRRET Laboratory, CNRS ERL 9215, University of Paris-Est Créteil, Créteil, France
| |
Collapse
|
27
|
Smith JR, Ashander LM, Ma Y, Rochet E, Furtado JM. Model Systems for Studying Mechanisms of Ocular Toxoplasmosis. Methods Mol Biol 2020; 2071:297-321. [PMID: 31758460 DOI: 10.1007/978-1-4939-9857-9_17] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The most common human disease caused by infection with Toxoplasma gondii is ocular toxoplasmosis, which typically is manifest as recurrent attacks of necrotizing retinal inflammation with subsequent scarring. The multilayered retina contains specialized cell populations, including endothelial cells, epithelial cells, neurons and supporting cells, all of which may be involved in this condition. In vitro investigations of basic mechanisms operating in human ocular toxoplasmosis use cellular and molecular methods that are common to the study of many pathological processes, and the novel aspect of this research is the use of human retinal cell subsets. Most in vivo research on ocular toxoplasmosis is conducted in the laboratory mouse. Experimental models involve local or systemic inoculation of parasites to induce acute disease, or sequential systemic and local parasite inoculations to trigger recurrent disease. We present methods for in vitro and in vivo studies of ocular toxoplasmosis, including dissection of the human eye, and culture and infection of differentiated cell populations from the retina, as well as induction of mouse ocular toxoplasmosis by intraocular, or sequential systemic and intraocular, inoculations, and imaging of toxoplasmic retinal lesions.
Collapse
Affiliation(s)
- Justine R Smith
- College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia.
| | - Liam M Ashander
- College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia
| | - Yuefang Ma
- College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia
| | - Elise Rochet
- College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia
| | - João M Furtado
- Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
28
|
Nitta K, Nishinaka A, Hida Y, Nakamura S, Shimazawa M, Hara H. Oral and ocular administration of crocetin prevents retinal edema in a murine retinal vein occlusion model. Mol Vis 2019; 25:859-868. [PMID: 31908404 PMCID: PMC6937220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 12/23/2019] [Indexed: 11/18/2022] Open
Abstract
Purpose We investigated the effects of oral and ocular administration of crocetin in a murine retinal vein occlusion (RVO) model. Crocetin is a type of carotenoid contained in the fruit of gardenia (Gardenia jasminoides Ellis) and the stigma of saffron (Crocus staruts L). Methods This study was performed on a murine RVO model, which was created by laser irradiation of retinal veins. We evaluated the retinal thickness after the oral administration of crocetin (100 mg/kg) 1 and 6 h before laser irradiation, and immediately, 6 h, 12 h, and 18 h after laser irradiation in the murine RVO model. In addition, we measured the retinal layer thickness after administration of crocetin eye drops (0.03% or 0.10%) immediately, 6 h, and 12 h after laser irradiation. Western blotting of retinal tissue was used to determine the expression levels of matrix metalloproteinase (MMP-9), tumor nuclear factor (TNF-α), and occludin after oral administration of crocetin. Results Oral and ocular administration of crocetin improved retinal edema in the murine RVO model. Crocetin administration statistically significantly suppressed overexpression of MMP-9 and TNF-α, and reversed the reduction of occludin. Conclusions These findings indicate that crocetin can protect retinal tight junctions by suppressing retinal edema through an anti-inflammatory effect, which suggests that crocetin may be useful for RVO disease.
Collapse
|
29
|
Spooner K, Fraser-Bell S, Hong T, Chang A. Prospective study of aflibercept for the treatment of persistent macular oedema secondary to retinal vein occlusions in eyes not responsive to long-term treatment with bevacizumab or ranibizumab. Clin Exp Ophthalmol 2019; 48:53-60. [PMID: 31498950 DOI: 10.1111/ceo.13636] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 08/28/2019] [Accepted: 09/02/2019] [Indexed: 11/27/2022]
Abstract
IMPORTANCE To examine the effect of switching from intravitreal bevacizumab or ranibizumab to aflibercept in eyes with persistent macular oedema due to retinal vein occlusion (RVO). BACKGROUND We report the results of a prospective interventional study on the effect of aflibercept 2 mg in eyes with persistent macular oedema after long-term treatment with bevacizumab or ranibizumab. DESIGN Non-randomized, prospective clinical trial. PARTICIPANTS Eighteen eyes of eighteen patients were included. METHODS Eyes with persistent macular oedema despite a minimum of four previous intravitreal bevacizumab/ranibizumab injections were recruited into this 48-week trial. Three loading doses of intravitreal aflibercept were administered every 4-weeks, thereafter every 8-weeks until week 48. MAIN OUTCOME MEASURES Mean change from baseline in best corrected visual acuity (BCVA) as measured by early treatment diabetic retinopathy score (ETDRS) and central macular thickness (CMT) as measured by spectral domain optical coherence tomography (SD-OCT) at 48 weeks. RESULTS Patients had received a mean of 40.0 ± 17.8 bevacizumab/ranibizumab intravitreal injections prior to switching to aflibercept. The mean number of previous injections administered in the 12-months preceding entry into the study was 10.2 ± 2.4. Mean vision change at week 48 was +21.1 ± 5.1 ETDRS letters in the BRVO group and +18.8 ± 5.9 letters at in the CRVO group (P < .001 for both groups). Mean decrease in CMT was 87.6 ± 48.8 μm and 191.0 ± 128.3 μm, in the BRVO and CRVO groups, respectively (P < .001). Using linear regression analyses, a higher number of previous intravitreal ranibizumab/bevacizumab injections and thicker pre-switch CMT were correlated with greater visual gains. CONCLUSION AND RELEVANCE Switching to aflibercept from bevacizumab or ranibizumab in eyes with persistent macular oedema due to RVO can lead to functional and anatomical improvement. This effect was more obvious in eyes with a greater CMT prior to the switch.
Collapse
Affiliation(s)
- Kimberly Spooner
- Sydney Retina, Sydney, New South Wales, Australia.,Sydney Institute of Vision Science, Sydney, New South Wales, Australia.,Save Sight Institute, The University of Sydney, Sydney, New South Wales, Australia
| | - Samantha Fraser-Bell
- Sydney Retina, Sydney, New South Wales, Australia.,Save Sight Institute, The University of Sydney, Sydney, New South Wales, Australia
| | - Thomas Hong
- Sydney Retina, Sydney, New South Wales, Australia.,Sydney Institute of Vision Science, Sydney, New South Wales, Australia
| | - Andrew Chang
- Sydney Retina, Sydney, New South Wales, Australia.,Sydney Institute of Vision Science, Sydney, New South Wales, Australia.,Save Sight Institute, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
30
|
Allingham MJ, Tserentsoodol N, Saloupis P, Mettu PS, Cousins SW. Aldosterone Exposure Causes Increased Retinal Edema and Severe Retinopathy Following Laser-Induced Retinal Vein Occlusion in Mice. Invest Ophthalmol Vis Sci 2019; 59:3355-3365. [PMID: 30025072 DOI: 10.1167/iovs.17-23073] [Citation(s) in RCA: 25] [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 To determine the effects of aldosterone exposure on retinal edema and retinopathy in a mouse model of retinal vein occlusion (RVO). Methods RVO was induced immediately following intravenous injection of Rose bengal (66 mg/kg) using a 532-nm wavelength laser to place three to seven applications at 80 mW and 50-μm spot size directed at the superior retinal vein one disc diameter away from the nerve. Negative control consisted of placing an equal number of laser spots without targeting the vein. Male and female C57BL/6J mice aged 7 to 9 months with confirmed absence of Crb1rd8 were used. Aldosterone pellets releasing a daily dose of 0.83 μg/day were implanted subcutaneously 4 weeks prior to RVO. Retinal imaging by optical coherence tomography (OCT) was performed using a Micron IV rodent imaging system. Retinas were analyzed by immunohistochemistry using standard techniques. Retinal imaging and tissue analysis were performed 2, 4, and 7 days following RVO. Comparisons were made using Student's t-test, ANOVA, and Pearson's χ2. Results RVO caused retinal edema in the form of cystic spaces and retinal thickening detectable by both OCT and histology. RVO also caused Müller glia (MG) dysfunction manifest as upregulated glial fibrillary acidic protein (GFAP) and altered localization of aquaporin 4 (AQP4) and Kir4.1. Treatment with aldosterone caused a significant increase in retinal edema and more severe retinopathy manifest as retinal whitening and extensive intraretinal hemorrhage. MG dysfunction was more severe and persistent in aldosterone-treated mice. Finally, aldosterone greatly increased the number of infiltrating mononuclear phagocytes following RVO. Conclusions Systemic aldosterone exposure causes a more severe RVO phenotype manifest as increased severity and duration of retinal edema and more severe retinopathy. The effects of aldosterone may be mediated by MG dysfunction and increased infiltration of mononuclear phagocytes. This suggests that small increases in aldosterone levels may be a risk factor for severe RVO.
Collapse
Affiliation(s)
- Michael J Allingham
- Department of Ophthalmology, Duke University Medical Center, Durham, North Carolina, United States
| | - Nomingerel Tserentsoodol
- Department of Ophthalmology, Duke University Medical Center, Durham, North Carolina, United States
| | - Peter Saloupis
- Department of Ophthalmology, Duke University Medical Center, Durham, North Carolina, United States
| | - Priyatham S Mettu
- Department of Ophthalmology, Duke University Medical Center, Durham, North Carolina, United States
| | - Scott W Cousins
- Department of Ophthalmology, Duke University Medical Center, Durham, North Carolina, United States
| |
Collapse
|
31
|
Moriguchi M, Nakamura S, Inoue Y, Nishinaka A, Nakamura M, Shimazawa M, Hara H. Irreversible Photoreceptors and RPE Cells Damage by Intravenous Sodium Iodate in Mice Is Related to Macrophage Accumulation. Invest Ophthalmol Vis Sci 2019; 59:3476-3487. [PMID: 30025075 DOI: 10.1167/iovs.17-23532] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose To determine the mechanism causing degeneration of the retinal pigment epithelium (RPE) and photoreceptors in mice after an intravenous injection of sodium iodate (NaIO3). Methods The time-dependent changes in NaIO3-induced retinal degeneration were determined by analyzing the retinal morphology by optical coherence tomographic (OCT) images, histological sections of the retina, physiology of the retina by electroretinography (ERG), and retinal blood flow by laser speckle flowgraphy. In addition, the expression of the genes associated with age-related macular degeneration in humans was assessed in the NaIO3-treated mice by RT-PCR. We also investigated whether macrophages were involved in the NaIO3-induced retinal degeneration. Results The intravenous injection of 20 mg/kg NaIO3 altered the morphology of the RPE cells and the ERGs transiently. With 40 mg/kg of NaIO3, the degeneration of the RPE cells was still present at 28 days. Aggregated melanin granules were surrounded by zonula occludens protein 1 (ZO-1)-positive cells. In addition, 40 mg/kg of NaIO3 led to a reduction in the amplitudes of the a- and b-waves of the dark-adapted ERGs. Histological studies showed that macrophages had infiltrated the retina and were present around the altered RPE cells. Depletion of the macrophages by a prior injection of clodronate liposomes prevented the damage of the outer retina after the NaIO3 injection but not the RPE. Conclusions The NaIO3-induced retinal damage was reversible at low concentrations but permanent at high concentrations of NaIO3. The accumulation of macrophages around the RPE cells caused the photoreceptor cell death.
Collapse
Affiliation(s)
- Mayu Moriguchi
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Shinsuke Nakamura
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Yuki Inoue
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Anri Nishinaka
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Maho Nakamura
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Masamitsu Shimazawa
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Hideaki Hara
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| |
Collapse
|
32
|
Long P, Yan W, Liu J, Li M, Chen T, Zhang Z, An J. Therapeutic Effect of Traditional Chinese Medicine on a Rat Model of Branch Retinal Vein Occlusion. J Ophthalmol 2019; 2019:9521379. [PMID: 30906588 PMCID: PMC6398022 DOI: 10.1155/2019/9521379] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Accepted: 01/14/2019] [Indexed: 02/06/2023] Open
Abstract
Branch retinal vein occlusion (BRVO) is a common retinal vascular disorder leading to visual impairment. Currently, the general strategies for BRVO are symptomatic therapies. Cardiovascular aspects are essential risk factors for BRVO. The traditional Chinese medicine hexuemingmu (HXMM), consisting of tanshinol and baicalin, dilates the vasculature and accelerates microcirculation. Therefore, the aim of this study was to determine the efficacy and possible mechanism of HXMM in a BRVO rat model established by laser photocoagulation. Successful BRVO rat models were treated with different doses of HXMM. Fundus photography and fluorescein fundus angiography (FFA) of the animals were applied. The retinal layers were measured by optical coherence tomography (OCT). Full-field electroretinography (ffERG) was applied to evaluate the retinal function. The ear vein flow velocity was measured via a microcirculation detector. The expression of the vascular endothelial growth factor (VEGF-α) was measured via western blotting and immunofluorescent staining. Our study found that retinal edema predominantly occurred in the inner nuclear layer (INL) and outer nuclear layer (ONL). The retinal edema of the treated groups was significantly relieved in the early stage of BRVO as visualized via OCT detection and HE staining. The amplitudes of the b wave and oscillatory potentials (OPs) waves of ffERG in the treated groups were increased compared with those of the control group at several detection points (3, 5, 7, 10, 14, and 21 d postocclusion). The expression of VEGF-α was reduced in the treated groups at an early stage of BRVO. Furthermore, the ear vein flow velocity of the HXMM treatment groups was faster than that of the control group. Thus, our study indicates that the traditional Chinese medicine HXMM could ameliorate retinal edema and rescue the retinal structure and function in BRVO models through promoting occluded vein recanalization, improving microcirculation, and regulating the expression of VEGF-α.
Collapse
Affiliation(s)
- Pan Long
- Center of Clinical Aerospace Medicine, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Weiming Yan
- Department of Ophthalmology, The 900th Hospital of the Logistic Team of Chinese PLA, Fuzhou, Fujian 350025, China
| | - Jianwen Liu
- Center of Clinical Aerospace Medicine, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Manhong Li
- Department of Ophthalmology of Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Tao Chen
- Center of Clinical Aerospace Medicine, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Zuoming Zhang
- Center of Clinical Aerospace Medicine, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Jing An
- Institute of Neurobiology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| |
Collapse
|
33
|
Středová M, Stepanov A, Studnička J, Nekolová J, Jirásková N. Ranibizumab in macular oedema secondary to branch retinal vein occlusion - 24 months of treatment. CESKÁ A SLOVENSKÁ OFTALMOLOGIE : CASOPIS CESKÉ OFTALMOLOGICKÉ SPOLECNOSTI A SLOVENSKÉ OFTALMOLOGICKÉ SPOLECNOSTI 2019; 75:190-198. [PMID: 32397721 DOI: 10.31348/2019/4/3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
PURPOSE To retrospectively evaluate the efficacy and safety of ranibizumab treatment for macular edema (ME) secondary to branch retina vein occlusion (BRVO) after 24 months. MATERIALS AND METHODS This study included 39 eyes of 39 patients with ME associated with BRVO treated at the Ophthalmology Department of Faculty Hospital in Hradec Kralove. The average age of included patiens was 69,3 years, the mean duration of symptoms before treament was 5,4 months, the mean baseline visual acuity (VA) was 54,6 ETDRS (Early Treatment Diabetic Retinopathy Study) letters, the mean baseline central retinal thickness (CRT) was 544,9 µm. At 64,1% patients a retinal laserphotocoagulation was performed before intravitreal ranibizumab. After one year, the study was discontinued by 17 patiens, the remaining 22 patients were observed for 24 months. Initially, there were 3 doses of intravitreal ranibizumab administered in monthly intervals, further injections were applied according to PRN (pro re nata) regiment. Patients were examined at baseline and then at 3, 6, 9, 12 and 24 months from initiation of the treatment. In this study, the effect of ranibizumab on functional and morphological parameters of the affected eye was monitored, the safety of this treatment was also evaluated. During the follow-up, a statistically significant improvement in VA was achieved in every visit in comparison to baseline parameters, the mean VA gain at the 3 month visit was 12,1 ETDRS letters (p.
Collapse
|
34
|
Xia JP, Wang S, Zhang JS. The anti-inflammatory and anti-oxidative effects of conbercept in treatment of macular edema secondary to retinal vein occlusion. Biochem Biophys Res Commun 2018; 508:1264-1270. [PMID: 30558792 DOI: 10.1016/j.bbrc.2018.12.049] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 12/06/2018] [Indexed: 12/15/2022]
Abstract
To investigate the effects of conbercept on inflammatory and oxidative response in macular edema secondary to retinal vein occlusion (RVO-ME). Retinal microvasculature were detected by optical coherence tomographic angiography (OCTA). The inflammation related factors including prostaglandin E1 (PGE1), prostaglandin E2 (PGE2), prostaglandin F2a (PGF2a), intercellular cell adhesion molecule-1 (ICAM-1) and macrophage inflammatory protein-1 (MIP-1) were determined in human and mice with RVO-ME. OCTA images showed that capillary non-perfusion, enlargement of the foveal avascular zone, telangiectatic vessels and some forms of intraretinal edema in RVO-ME and all these were alleviated by conbercept treatment. PGE1, PGE2, PGF2a, ICAM-1 and MIP-1 in aqueous fluid extracted from RVO-ME patients was significantly increased compared with non-RVO subjects, intravitreal injection of conbercept partly reduced ICAM-1 and MIP-1 levels but not PGE1, PGE2 and PGF2a. The glutathione level was reduced in aqueous fluid extracted from RVO-ME patients but was restored after conbercept treatment. The inflammation, angiogenesis and ROS generation was increased in RVO-ME mice, conbercept partly inhibited these effects. Mechanistically, conbercept inhibited vascular endothelial growth factor (VEGF), ICAM-1, MIP-1, NOX-1 and NOX-4 protein expressions, but not PGE1, PGE2 and PGF2a expressions. Conbercept alleviates RVO-ME through inhibiting inflammation, angiogenesis and oxidative responses. These findings further reveals the molecular mechanism of conbercept for treatment of RVO-ME.
Collapse
Affiliation(s)
- Jian-Ping Xia
- Department of Ophthalmology, The Fourth Affiliated Hospital of China Medical University, Eye Hospital of China Medical University, Shenyang, 110005, China; Department of Ophthalmology, Shenyang Aier Eye Hospital, Shenyang, 110043, China
| | - Shuai Wang
- Department of Ophthalmology, Second Affiliated Hospital of Dalian Medical University, Dalian, 116023, China.
| | - Jin-Song Zhang
- Department of Ophthalmology, The Fourth Affiliated Hospital of China Medical University, Eye Hospital of China Medical University, Shenyang, 110005, China.
| |
Collapse
|
35
|
Elamaa H, Kihlström M, Kapiainen E, Kaakinen M, Miinalainen I, Ragauskas S, Cerrada-Gimenez M, Mering S, Nätynki M, Eklund L. Angiopoietin-4-dependent venous maturation and fluid drainage in the peripheral retina. eLife 2018; 7:37776. [PMID: 30444491 PMCID: PMC6239434 DOI: 10.7554/elife.37776] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 10/22/2018] [Indexed: 01/09/2023] Open
Abstract
The maintenance of fluid homeostasis is necessary for function of the neural retina; however, little is known about the significance of potential fluid management mechanisms. Here, we investigated angiopoietin-4 (Angpt4, also known as Ang3), a poorly characterized ligand for endothelial receptor tyrosine kinase Tie2, in mouse retina model. By using genetic reporter, fate mapping, and in situ hybridization, we found Angpt4 expression in a specific sub-population of astrocytes at the site where venous morphogenesis occurs and that lower oxygen tension, which distinguishes peripheral and venous locations, enhances Angpt4 expression. Correlating with its spatiotemporal expression, deletion of Angpt4 resulted in defective venous development causing impaired venous drainage and defects in neuronal cells. In vitro characterization of angiopoietin-4 proteins revealed both ligand-specific and redundant functions among the angiopoietins. Our study identifies Angpt4 as the first growth factor for venous-specific development and its importance in venous remodeling, retinal fluid clearance and neuronal function.
Collapse
Affiliation(s)
- Harri Elamaa
- Oulu Center for Cell-Matrix Research, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland.,Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Minna Kihlström
- Oulu Center for Cell-Matrix Research, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland.,Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Emmi Kapiainen
- Oulu Center for Cell-Matrix Research, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland.,Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Mika Kaakinen
- Oulu Center for Cell-Matrix Research, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland.,Biocenter Oulu, University of Oulu, Oulu, Finland
| | | | | | | | - Satu Mering
- R&D Department, Experimentica Ltd, Kuopio, Finland
| | - Marjut Nätynki
- Oulu Center for Cell-Matrix Research, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland.,Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Lauri Eklund
- Oulu Center for Cell-Matrix Research, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland.,Biocenter Oulu, University of Oulu, Oulu, Finland
| |
Collapse
|
36
|
Nishinaka A, Inoue Y, Fuma S, Hida Y, Nakamura S, Shimazawa M, Hara H. Pathophysiological Role of VEGF on Retinal Edema and Nonperfused Areas in Mouse Eyes With Retinal Vein Occlusion. ACTA ACUST UNITED AC 2018; 59:4701-4713. [DOI: 10.1167/iovs.18-23994] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Affiliation(s)
- Anri Nishinaka
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Yuki Inoue
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Shinichiro Fuma
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Yoshifumi Hida
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Shinsuke Nakamura
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Masamitsu Shimazawa
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Hideaki Hara
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| |
Collapse
|
37
|
Effects of ripasudil, a ROCK inhibitor, on retinal edema and nonperfusion area in a retinal vein occlusion murine model. J Pharmacol Sci 2018; 137:129-136. [PMID: 29983234 DOI: 10.1016/j.jphs.2018.06.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 04/26/2018] [Accepted: 05/08/2018] [Indexed: 12/23/2022] Open
Abstract
Rho-associated coiled-coil containing protein kinase (ROCK) inhibitors are used to treat glaucoma patients and have protective effects on ischemic states. However, it is poorly understood how the ROCK pathway affects the pathological signs of retinal vein occlusion (RVO). In this study, we evaluated the effects of ripasudil, a ROCK inhibitor, on a murine RVO model. In vivo, RVO was induced by retinal vein laser irradiation in mice, and evaluated with ripasudil. In vitro, the effects of ripasudil were examined on tight junction protein integrity in human retinal microvascular endothelial cells (HRMECs). Moreover, we investigated the expression level of the phosphorylated myosin phosphatase target protein (MYPT)-1 after administration of ripasudil. Ripasudil significantly prevented deterioration, such as retinal edema, reduced the size of the nonperfusion area, and improved retinal blood flow. Ripasudil treatment inhibited disintegration of ZO-1 in HRMECs. Administration of ripasudil suppressed retinal phosphorylation of MYPT-1 in a murine RVO model. These findings indicate that ripasudil might be as a possible therapeutic agent for RVO.
Collapse
|
38
|
Gene expression profiling in a mouse model of retinal vein occlusion induced by laser treatment reveals a predominant inflammatory and tissue damage response. PLoS One 2018. [PMID: 29529099 PMCID: PMC5846732 DOI: 10.1371/journal.pone.0191338] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Purpose Retinal vein occlusion (RVO) has been investigated in several laser-induced animal models using pigs, rabbits and rats. However, laser-induced RVO has been rarely reported in mice, despite the impressive number of available mutants, ease of handling and cost effectiveness. The aim of this study was to further assess the feasibility of a RVO mouse model for gene expression analysis and its possible use to investigate effects of hypoxia. Methods C57Bl/6J mice were injected with eosin Y for photo-sensitization. Subsequently, large retinal veins were laser-treated in one eye to induce vascular occlusion. Contralateral control eyes received non-occlusive retinal laser treatment sparing large vessels. The animals were followed for up to eight days and assessed by funduscopy, angiography, hypoxyprobe staining, histopathology and gene expression analysis by qPCR and RNA sequencing (RNAseq). Another group of mice was left untreated and studied at a single time point to determine baseline characteristics. Results Laser-induced RVO persisted in half of the treated veins for three days, and in a third of the veins for the whole observation period of 8 days. Funduscopy revealed large areas of retinal swelling in all laser-treated eyes, irrespective of vascular targeting or occlusion status. Damage of the outer retina, retinal pigment epithelium (RPE), and even choroid and sclera at the laser site was observed in histological sections. Genes associated with inflammation or cell damage were highly up-regulated in all laser-treated eyes as detected by RNAseq and qPCR. Retinal hypoxia was observed by hypoxyprobe staining in all RVO eyes for up to 5 days with a maximal extension at days 2 and 3, but no significant RVO-dependent changes in gene expression were detected for angiogenesis- or hypoxia-related genes. Conclusion The laser-induced RVO mouse model is characterized by a predominant general inflammatory and tissue damage response, which may obscure distinct hypoxia- and angiogenesis-related effects. A non-occlusive laser treatment control is essential to allow for proper data interpretation and should be mandatory in animal studies of laser-induced RVO to dissect laser-induced tissue damage from vascular occlusion effects.
Collapse
|
39
|
Nishinaka A, Fuma S, Inoue Y, Shimazawa M, Hara H. Effects of kallidinogenase on retinal edema and size of non-perfused areas in mice with retinal vein occlusion. J Pharmacol Sci 2017; 134:86-92. [PMID: 28619445 DOI: 10.1016/j.jphs.2017.05.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 05/16/2017] [Accepted: 05/18/2017] [Indexed: 11/30/2022] Open
Abstract
Kallidinogenase has been used to treat retinal vein occlusion (RVO) in patients, although there are no evidences on the effects of kallidinogenase on the retinal edema and the non-perfused areas in eyes with a RVO. We have established a murine RVO model with retinal edema and non-perfused areas. The purpose of this study was to evaluate the effects of kallidinogenase on the retinal edema and size of the non-perfused areas in the mouse RVO model. We evaluated the thickness of the retinal layers and size of the non-perfused areas, and the blood flow by laser speckle flowgraphy in RVO model. The effects of an intravenous injection of kallidinogenase on the retinal edema and size of the non-perfused areas were determined. In addition, the expressions of phosphorylated protein kinase B (Akt) and endothelial nitric oxide synthase (eNOS) were measured by Western blotting. Our results showed that kallidinogenase reduced the degree of retinal edema and size of the non-perfused areas by an increase in the blood flow in RVO model. Kallidinogenase also increased the levels of phosphorylated Akt and eNOS. These findings indicate that kallidinogenase acted through Akt/eNOS-dependent phosphorylation. Thus, kallidinogenase should be considered as a possible therapeutic agent for RVO patients.
Collapse
Affiliation(s)
- Anri Nishinaka
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Shinichiro Fuma
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Yuki Inoue
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Masamitsu Shimazawa
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan.
| | - Hideaki Hara
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| |
Collapse
|
40
|
Shimazawa M, Nishinaka A, Hara H. [Experimental techniques for animal models of retinal vein occlusion in mice]. Nihon Yakurigaku Zasshi 2017; 150:293-297. [PMID: 29225292 DOI: 10.1254/fpj.150.293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
|