1
|
Zhang J, Sheng X, Ding Q, Wang Y, Zhao J, Zhang J. Subretinal fibrosis secondary to neovascular age-related macular degeneration: mechanisms and potential therapeutic targets. Neural Regen Res 2025; 20:378-393. [PMID: 38819041 PMCID: PMC11317958 DOI: 10.4103/nrr.nrr-d-23-01642] [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: 09/30/2023] [Revised: 12/19/2023] [Accepted: 01/15/2024] [Indexed: 06/01/2024] Open
Abstract
Subretinal fibrosis is the end-stage sequelae of neovascular age-related macular degeneration. It causes local damage to photoreceptors, retinal pigment epithelium, and choroidal vessels, which leads to permanent central vision loss of patients with neovascular age-related macular degeneration. The pathogenesis of subretinal fibrosis is complex, and the underlying mechanisms are largely unknown. Therefore, there are no effective treatment options. A thorough understanding of the pathogenesis of subretinal fibrosis and its related mechanisms is important to elucidate its complications and explore potential treatments. The current article reviews several aspects of subretinal fibrosis, including the current understanding on the relationship between neovascular age-related macular degeneration and subretinal fibrosis; multimodal imaging techniques for subretinal fibrosis; animal models for studying subretinal fibrosis; cellular and non-cellular constituents of subretinal fibrosis; pathophysiological mechanisms involved in subretinal fibrosis, such as aging, infiltration of macrophages, different sources of mesenchymal transition to myofibroblast, and activation of complement system and immune cells; and several key molecules and signaling pathways participating in the pathogenesis of subretinal fibrosis, such as vascular endothelial growth factor, connective tissue growth factor, fibroblast growth factor 2, platelet-derived growth factor and platelet-derived growth factor receptor-β, transforming growth factor-β signaling pathway, Wnt signaling pathway, and the axis of heat shock protein 70-Toll-like receptors 2/4-interleukin-10. This review will improve the understanding of the pathogenesis of subretinal fibrosis, allow the discovery of molecular targets, and explore potential treatments for the management of subretinal fibrosis.
Collapse
Affiliation(s)
- Jingxiang Zhang
- Department of Ophthalmology, People’s Hospital of Huangdao District, Qingdao, Shandong Province, China
| | - Xia Sheng
- Department of Ophthalmology, People’s Hospital of Huangdao District, Qingdao, Shandong Province, China
| | - Quanju Ding
- Department of Ophthalmology, People’s Hospital of Huangdao District, Qingdao, Shandong Province, China
| | - Yujun Wang
- Department of Urology, People’s Hospital of Huangdao District, Qingdao, Shandong Province, China
| | - Jiwei Zhao
- Department of Ophthalmology, People’s Hospital of Huangdao District, Qingdao, Shandong Province, China
| | - Jingfa Zhang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People’s Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| |
Collapse
|
2
|
Qiao X, Shah W, Gao X, Gong Y, Li Y, Gao Y, Li J. Understanding how the immune system environment is controlled in high myopia cases. Int Immunopharmacol 2024; 143:113138. [PMID: 39362012 DOI: 10.1016/j.intimp.2024.113138] [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: 05/15/2024] [Revised: 09/03/2024] [Accepted: 09/08/2024] [Indexed: 10/05/2024]
Abstract
High myopia (HM) is characterized by a significant extension of the eye axis; it has emerged as a serious global public health issue recently. In addition to causing severe visual impairment, HM is associated with several problems that may compromise an individual's vision. Although genetic and environmental factors in HM have been extensively investigated, increasing evidence implicates the immune system and its microenvironment in its pathogenesis. In this review, we explore the complex interactions between cytokines, immune cells, and the eye environment to elucidate the complex processes controlling the immune response in HM. Furthermore, we investigated treatments modulating the immune response and alleviating the progression of HM and its complications. Through a review of the current relevant studies, we highlight the critical functions of the immune system in the multifactorial development of HM. With the evolving understanding of the immune system's involvement in HM, this review provides a valuable resource to clinicians and researchers to develop targeted interventions and personalized treatments for individuals with this vision-threatening condition.
Collapse
Affiliation(s)
- Xin Qiao
- Shanxi Eye Hospital Affiliated to Shanxi Medical University, Taiyuan 030002, China
| | - Wahid Shah
- Shanxi Eye Hospital Affiliated to Shanxi Medical University, Taiyuan 030002, China; Translational Medicine Research Center, Shanxi Medical University, Taiyuan 030001, China
| | - Xiaoqin Gao
- Shanxi Eye Hospital Affiliated to Shanxi Medical University, Taiyuan 030002, China
| | - Yuxing Gong
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan 030001, China
| | - Yanan Li
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan 030001, China
| | - Yuan Gao
- Shanxi Eye Hospital Affiliated to Shanxi Medical University, Taiyuan 030002, China; Translational Medicine Research Center, Shanxi Medical University, Taiyuan 030001, China.
| | - Junhong Li
- Shanxi Eye Hospital Affiliated to Shanxi Medical University, Taiyuan 030002, China.
| |
Collapse
|
3
|
Sun J, Song Y, Gong Y, Tao L, Wang H, Shu X, Wen Y, Cui L, Ye J, Lu S, Deng J, Li H, Xu Y, Qian L, Wu Z, Bi Y, Liu Q, Xu X, Wu M, Zhang J, Hao J, Tong J, Dai H, Wang F, Zhao M, Mao J, Li C, He T, Pei C, Liu X, Wang X, Li M, Wang W, Zheng Q, Guan H, Peng H, Fan K, Zhang W, Zhu D, Yu S, Wei W, Ding L, Li J, Lu P, Yan M, Liu W, Jia H, Sun X. Efficacy and Safety of Efdamrofusp Alfa versus Aflibercept in Participants with Neovascular Age-Related Macular Degeneration: A Randomized, Double-Masked, Active-Controlled, Noninferiority, Phase II Trial. Ophthalmol Retina 2024:S2468-6530(24)00401-9. [PMID: 39214250 DOI: 10.1016/j.oret.2024.08.014] [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: 12/22/2023] [Revised: 08/20/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Abstract
PURPOSE To evaluate efficacy and safety of efdamrofusp alfa compared with aflibercept in neovascular age-related macular degeneration (nAMD). DESIGN Randomized, double-masked, multicenter, active-controlled, noninferiority phase II study. PARTICIPANTS A total of 231 treatment-naive and previously treated participants with active choroidal neovascularization secondary to nAMD were enrolled. METHODS Eligible participants were randomized (1:1:1) to 2 mg efdamrofusp alfa, 4 mg efdamrofusp alfa, or 2 mg aflibercept groups. Participants in all groups received 3 initial monthly loading doses, followed by treatment every 8 weeks, with assessment every 4 weeks up to week 52. MAIN OUTCOME MEASURES The primary end point was the mean best-corrected visual acuity (BCVA) change from baseline to week 36. The prespecified noninferiority margin was set as -5 letters (80% confidence interval [CI]). RESULTS Each treatment group included 77 participants. The mean BCVA changes from baseline to week 36 for 2 mg efdamrofusp alfa, 4 mg efdamrofusp alfa and aflibercept groups were +10.6, +11.4, and +12.0 letters, respectively; least squares mean difference were -1.4 (80% CI: -3.5 to 0.7) between 2 mg efdamrofusp alfa and aflibercept, and -0.6 (80% CI: -2.7 to 1.6) between 4 mg efdamrofusp alfa and aflibercept. Mean central retinal thickness changes were consistent across groups. Adverse event rate was comparable among the groups. CONCLUSIONS Efdamrofusp alfa demonstrated noninferiority to aflibercept in BCVA improvement, accompanied by a similar safety profile. FINANCIAL DISCLOSURE(S) The author(s) have no proprietary or commercial interest in any materials discussed in this article.
Collapse
Affiliation(s)
- Junran Sun
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Clinical Research Center for Ophthalmic Diseases, Shanghai, China; Shanghai Key Laboratory of Fundus Diseases, Shanghai, China; Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China
| | - Yanping Song
- Department of Ophthalmology, General Hospital of Central Theater Command, Wuhan, China
| | - Yuanyuan Gong
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Clinical Research Center for Ophthalmic Diseases, Shanghai, China
| | - Liming Tao
- Department of Ophthalmology, The Second Hospital of Anhui Medical University, Hefei, China
| | - Hong Wang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Clinical Research Center for Ophthalmic Diseases, Shanghai, China
| | - Xiangwen Shu
- Department of Ophthalmology, Jinan Second People's Hospital, Jinan, China
| | - Ying Wen
- The Eye Hospital Affiliated to Shandong Traditional Chinese Medicine University, Jinan, China
| | - Ling Cui
- Department of Ophthalmology, The People's Hospital of Guangxi Zhuang Autonomous Region, Guilin, China
| | - Jian Ye
- Department of Ophthalmology, Army Medical Center of PLA, Xi'an, China
| | - Shujie Lu
- Innovent Biologics, Inc., Suzhou, China
| | | | - Haoyu Li
- Innovent Biologics, Inc., Suzhou, China
| | - Yihua Xu
- Innovent Biologics, Inc., Suzhou, China
| | - Lei Qian
- Innovent Biologics, Inc., Suzhou, China
| | - Zhifeng Wu
- Department of Ophthalmology, Jiangnan University Medical Center, Wuxi, China
| | - Yanlong Bi
- Department of Ophthalmology, Tongji Hospital of Tongji University, Shanghai, China
| | - Qinghuai Liu
- Department of Ophthalmology, Jiangsu Province Hospital, Nanjing, China
| | - Xiangzhong Xu
- The Affiliated Eye Hospital of Nanjing Medical University, Nanjing, China
| | - Miaoqin Wu
- Department of Ophthalmology, Zhejiang Provincial People's Hospital, Hangzhou, China
| | | | - Jilong Hao
- Department of Ophthalmology, The First Bethune Hospital of Jinlin University, Changchun, China
| | - Jianping Tong
- Department of Ophthalmology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Hong Dai
- Department of Ophthalmology, Beijing Hospital, Beijing, China
| | - Feng Wang
- Department of Ophthalmology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - MingWei Zhao
- Department of Ophthalmology, Peking University People's Hospital, Beijing, China
| | - Junfeng Mao
- Department of Ophthalmology, Xiangya Hospital Central South University, Changsha, China
| | - Chaopeng Li
- Department of Ophthalmology, Huai'an First People's Hospital, Huai'an, China
| | - Tao He
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Cheng Pei
- Department of Ophthalmology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiaoling Liu
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Xian Wang
- Department of Ophthalmology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Mingxin Li
- Department of Ophthalmology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Wei Wang
- Hebei Eye Hospital, Hebei, China
| | | | - Huaijin Guan
- Department of Ophthalmology, Affiliated Hospital of Nantong University, Nantong, China
| | - Hui Peng
- Department of Ophthalmology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ke Fan
- Henan Provincial Eye Hospital, Zhengzhou, China
| | - Wenfang Zhang
- Department of Ophthalmology, Lanzhou University Second Hospital, Lanzhou, China
| | - Dan Zhu
- Department of Ophthalmology, The Affiliated Hospital of Inner Mongolia Medical University, Huhehaote, China
| | - Songping Yu
- Department of Ophthalmology, Lishui Municipal Central Hospital, Zhejiang, China
| | - Wenbin Wei
- Department of Ophthalmology, Beijing Tongren Hospital, Beijing, China
| | - Lin Ding
- Department of Ophthalmology, Xinjiang UIGER Municipal People's Hospital, Xinjiang, China
| | - Jinying Li
- Department of Ophthalmology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Peirong Lu
- Department of Ophthalmology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Ming Yan
- Department of Ophthalmology, General Hospital of Central Theater Command, Wuhan, China
| | - Wei Liu
- Department of Ophthalmology, Army Medical Center of PLA, Xi'an, China
| | - Huixun Jia
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Clinical Research Center for Ophthalmic Diseases, Shanghai, China; Shanghai Key Laboratory of Fundus Diseases, Shanghai, China; Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China.
| | - Xiaodong Sun
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Clinical Research Center for Ophthalmic Diseases, Shanghai, China; Shanghai Key Laboratory of Fundus Diseases, Shanghai, China; Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China.
| |
Collapse
|
4
|
Gill K, Yoo HS, Chakravarthy H, Granville DJ, Matsubara JA. Exploring the role of granzyme B in subretinal fibrosis of age-related macular degeneration. Front Immunol 2024; 15:1421175. [PMID: 39091492 PMCID: PMC11291352 DOI: 10.3389/fimmu.2024.1421175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 07/02/2024] [Indexed: 08/04/2024] Open
Abstract
Age-related macular degeneration (AMD), a prevalent and progressive degenerative disease of the macula, is the leading cause of blindness in elderly individuals in developed countries. The advanced stages include neovascular AMD (nAMD), characterized by choroidal neovascularization (CNV), leading to subretinal fibrosis and permanent vision loss. Despite the efficacy of anti-vascular endothelial growth factor (VEGF) therapy in stabilizing or improving vision in nAMD, the development of subretinal fibrosis following CNV remains a significant concern. In this review, we explore multifaceted aspects of subretinal fibrosis in nAMD, focusing on its clinical manifestations, risk factors, and underlying pathophysiology. We also outline the potential sources of myofibroblast precursors and inflammatory mechanisms underlying their recruitment and transdifferentiation. Special attention is given to the potential role of mast cells in CNV and subretinal fibrosis, with a focus on putative mast cell mediators, tryptase and granzyme B. We summarize our findings on the role of GzmB in CNV and speculate how GzmB may be involved in the pathological transition from CNV to subretinal fibrosis in nAMD. Finally, we discuss the advantages and drawbacks of animal models of subretinal fibrosis and pinpoint potential therapeutic targets for subretinal fibrosis.
Collapse
Affiliation(s)
- Karanvir Gill
- Department of Ophthalmology and Visual Sciences, University of British Columbia (UBC), Vancouver, BC, Canada
| | - Hyung-Suk Yoo
- Department of Ophthalmology and Visual Sciences, University of British Columbia (UBC), Vancouver, BC, Canada
| | - Harshini Chakravarthy
- Department of Ophthalmology and Visual Sciences, University of British Columbia (UBC), Vancouver, BC, Canada
| | - David J. Granville
- International Collaboration on Repair Discoveries (ICORD), Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Joanne A. Matsubara
- Department of Ophthalmology and Visual Sciences, University of British Columbia (UBC), Vancouver, BC, Canada
| |
Collapse
|
5
|
Li X, Li J, Zeng W, Wang B, Du M, Liang L, Gao Y. Mingjing granule inhibits the subretinal fibrovascular membrane of two-stage laser-induced neovascular age-related macular degeneration in rats. Front Pharmacol 2024; 15:1384418. [PMID: 38983912 PMCID: PMC11231192 DOI: 10.3389/fphar.2024.1384418] [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: 02/09/2024] [Accepted: 05/31/2024] [Indexed: 07/11/2024] Open
Abstract
Objective The study aims to investigate the protective effect of Mingjing granule (MG) in a fibrovascular membrane rat model of neovascular age-related macular degeneration (nAMD) and explore the underlying mechanism. Methods The nAMD fibrovascular membrane model was established by two-stage laser photocoagulation. BN rats were randomly divided into four groups: the model group was gavaged with distilled water, the anti-VEGF group was given an intravitreous injection of ranibizumab, the MG + anti-VEGF group was gavaged with MG combined with an intravitreous injection of ranibizumab, and the normal group not modeled only fed conventionally. Lesions were evaluated by color fundus photograph, optical coherence tomography, fundus fluorescein angiography, and retinal pigment epithelial-choroid-sclera flat mount. The changes in the retinal structure were observed by histopathology. The expression of inflammatory cell markers F4/80, Iba-1, and glial fibrillary acidic protein (GFAP); the fibrosis-related factors collagen-1, fibronectin, α-smooth muscle actin (α-SMA), and transforming growth factor-beta (TGF-β); and the complement system-related factors C3a and C3aR in the retina were detected by immunofluorescence or qRT-PCR. Results The current study revealed that MG + anti-VEGF administration more significantly reduced the thickness of fibrovascular lesions, suppressed vascular leakage (exudation area and mean density value), inhibited the area of fibrovascular lesions, and restrained the formation of the fibrovascular membrane than the anti-VEGF agent alone in the two-stage laser-induced rat model. The fluorescence intensities of F4/80, Iba-1, collagen-1, fibronectin, TGF-β, and C3aR showed more significant inhibition in MG + anti-VEGF-treated rats than the anti-VEGF agent alone. The mRNA expression levels of F4/80, Iba-1, GFAP, collagen-1, fibronectin, α-SMA, TGF-β, and C3a showed lower levels in rats treated with MG + anti-VEGF than the anti-VEGF agent alone. Conclusion Combining MG with anti-VEGF treatment inhibits the growth of the fibrovascular membrane more effectively than using anti-VEGF treatment alone. The mechanism underlying this effect may involve limiting inflammatory cell aggregation, controlling complement system activation, and decreasing the expression of the fibrotic protein.
Collapse
Affiliation(s)
- Xiaoyu Li
- Eye Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Ophthalmic Disease Project Group, China Evidence-based Medicine Center of Traditional Chinese Medicine, Beijing, China
| | - Jiaxian Li
- Eye Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Weixin Zeng
- Eye Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Baoli Wang
- Eye Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Maobo Du
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Lina Liang
- Eye Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Ophthalmic Disease Project Group, China Evidence-based Medicine Center of Traditional Chinese Medicine, Beijing, China
| | - Yun Gao
- Eye Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Ophthalmic Disease Project Group, China Evidence-based Medicine Center of Traditional Chinese Medicine, Beijing, China
| |
Collapse
|
6
|
Xu H, Yi C, Chen M. The complement pathway as a therapeutic target for neovascular age-related macular degeneration-mediated subretinal fibrosis. Curr Opin Pharmacol 2024; 76:102448. [PMID: 38555699 DOI: 10.1016/j.coph.2024.102448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 03/07/2024] [Accepted: 03/07/2024] [Indexed: 04/02/2024]
Abstract
Neovascular age-related macular degeneration (nAMD) is the leading cause of blindness in the elderly in developed countries. Intravitreal injection of VEGF inhibitors is the mainstream therapy for nAMD, although nearly 50% of the patients do not respond or respond poorly to the therapy. One of the main reasons for the poor outcome of the therapy is the development of subretinal macular fibrosis, a process of excessive deposition of extracellular matrix proteins around the diseased blood vessels. Currently, there is no medication to prevent or treat the condition. Here, we discussed recent advances in the pathogenesis of nAMD-mediated macular fibrosis, with a focus on the role of the complement system. We further proposed approaches to target the complement system for the management of macular fibrosis and highlighted the area of further research for future clinical applications of complement-based therapy.
Collapse
Affiliation(s)
- Heping Xu
- Changsha Aier Eye Hospital, Changsha, Hunan, China; Aier Eye Institute, Aier Academy of Ophthalmology, Central South University, Hunan, China; The Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, United Kingdom.
| | - Caijiao Yi
- Changsha Aier Eye Hospital, Changsha, Hunan, China; Aier Eye Institute, Aier Academy of Ophthalmology, Central South University, Hunan, China; Changsha Xiangjiang Aier Eye Hospital, Changsha, Hunan, China
| | - Mei Chen
- The Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, United Kingdom.
| |
Collapse
|
7
|
Chu L, Bi C, Wang C, Zhou H. The Relationship between Complements and Age-Related Macular Degeneration and Its Pathogenesis. J Ophthalmol 2024; 2024:6416773. [PMID: 38205100 PMCID: PMC10776198 DOI: 10.1155/2024/6416773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 06/08/2023] [Accepted: 12/23/2023] [Indexed: 01/12/2024] Open
Abstract
Age-related macular degeneration is a retinal disease that causes permanent loss of central vision in people over the age of 65. Its pathogenesis may be related to mitochondrial dysfunction, inflammation, apoptosis, autophagy, complement, intestinal flora, and lipid disorders. In addition, the patient's genes, age, gender, cardiovascular disease, unhealthy diet, and living habits may also be risk factors for this disease. Complement proteins are widely distributed in serum and tissue fluid. In the early 21st century, a connection was found between the complement cascade and age-related macular degeneration. However, little is known about the effect of complement factors on the pathogenesis of age-related macular degeneration. This article reviews the factors associated with age-related macular degeneration, the relationship between each factor and complement, the related functions, and variants and provides new ideas for the treatment of this disease.
Collapse
Affiliation(s)
- Liyuan Chu
- Department of Ophthalmology, China–Japan Union Hospital of Jilin University, Changchun, China
| | - Chaoran Bi
- College of Traditional Chinese Medicine, Hainan Medical University, Haikou, Hainan, China
| | - Caiming Wang
- Department of Ophthalmology, China–Japan Union Hospital of Jilin University, Changchun, China
| | - Hongyan Zhou
- Department of Ophthalmology, China–Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
8
|
Zhou S, Wang Z, Gao L, Chen M, Duan Y, Zhou P, Liu Z, Wu C, Zhang J, Zhu Q. C5a/C5aR1 axis as a key driver promotes epithelial-to-mesenchymal transition in airway epithelial cells in silica nanoparticles-induced pulmonary fibrosis. Int Immunopharmacol 2023; 125:111112. [PMID: 37948857 DOI: 10.1016/j.intimp.2023.111112] [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: 07/31/2023] [Revised: 10/12/2023] [Accepted: 10/19/2023] [Indexed: 11/12/2023]
Abstract
Previous studies have shown that silica nanoparticles (SiNPs) exposure can affect the respiratory, cardiovascular, reproductive and other systems, with the lung being the primary target organ for the direct effect, causing damage with a central feature of pulmonary inflammation and fibrosis. However, the underlying mechanisms of pulmonary fibrosis due to SiNPs are not fully understood. The aim of the study was to investigate the role of complement anaphylatoxin C5a in SiNPs-induced pulmonary fibrosis. A mouse model of SiNPs-induced pulmonary fibrosis was established, and pulmonary fibrosis-related indicators, epithelial-to-mesenchymal transition (EMT), C5a/C5aR1 and high mobility group protein B1 (HMGB1) proteins were measured. An in vitro study using the human lung epithelial cell line BEAS-2B investigated whether C5a leads to epithelial-to-mesenchymal trans-differentiation. In vivo studies revealed that SiNPs-induced pulmonary fibrosis mainly manifested as EMT trans-differentiation in airway epithelial cells, which subsequently led to excessive deposition of extracellular matrix (ECM). Furthermore, we found that C5a and C5aR1 proteins were also increased in SiNPs-induced pulmonary fibrosis tissue. In vitro studies also showed that C5a directly activated HMGB1/RAGE signaling and induced EMT in BEAS-2B cells. Finally, treatment of SiNPs-exposed mice with the C5aR1 inhibitor PMX205 effectively reduced C5aR1 levels and inhibited the activation of HMGB1/RAGE signaling and the expression of EMT-related proteins, culminating in a significant alleviation of pulmonary fibrosis. Taken together, our results suggest that C5a/C5aR1 is the main signaling pathway for SiNPs-induced pulmonary fibrosis, which induces EMT in airway epithelial cells via the HMGB1/RAGE axis.
Collapse
Affiliation(s)
- Sifan Zhou
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Zhoujian Wang
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Lei Gao
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Muyue Chen
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Yuansheng Duan
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Pengcheng Zhou
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Zhibing Liu
- Department of Dermatology, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, Anhui, China
| | - Changhao Wu
- Department of Biochemistry and Physiology, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - Jiaxiang Zhang
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China.
| | - Qixing Zhu
- Department of Dermatology, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, Anhui, China.
| |
Collapse
|
9
|
Patel SS, Lally DR, Hsu J, Wykoff CC, Eichenbaum D, Heier JS, Jaffe GJ, Westby K, Desai D, Zhu L, Khanani AM. Avacincaptad pegol for geographic atrophy secondary to age-related macular degeneration: 18-month findings from the GATHER1 trial. Eye (Lond) 2023; 37:3551-3557. [PMID: 36964259 PMCID: PMC10686386 DOI: 10.1038/s41433-023-02497-w] [Citation(s) in RCA: 33] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/24/2023] [Accepted: 03/07/2023] [Indexed: 03/26/2023] Open
Abstract
BACKGROUND/OBJECTIVES To assess the safety and efficacy of avacincaptad pegol (ACP), a C5 inhibitor, for geographic atrophy (GA) secondary to age-related macular degeneration (AMD) over an 18-month treatment course. SUBJECTS/METHODS This study was an international, prospective, randomized, double-masked, sham-controlled, phase 2/3 clinical trial that consisted of 2 parts. In part 1, 77 participants were randomized 1:1:1 to receive monthly intravitreal injections of ACP 1 mg, ACP 2 mg, or sham. In part 2, 209 participants were randomized 1:2:2 to receive monthly ACP 2 mg, ACP 4 mg, or sham. The mean rate of change of GA over 18 months was measured by fundus autofluorescence. RESULTS Compared with their respective sham cohorts, monthly ACP treatment reduced the mean GA growth (square root transformation) over 18 months by 28.1% (0.168 mm, 95% CI [0.066, 0.271]) for the 2 mg cohort and 30.0% (0.167 mm, 95% CI [0.062, 0.273]) for the 4 mg cohort. ACP treatment was generally well tolerated over 18 months, with most ocular adverse events (AEs) related to the injection procedure. Macular neovascularization (MNV) was more frequent in both 2 mg (11.9%) and 4 mg (15.7%) cohorts than their respective sham control groups (2.7% and 2.4%). CONCLUSIONS Over this 18-month study, ACP 2 mg and 4 mg showed continued reductions in the progression of GA growth compared to sham and continued to be generally well tolerated. A pivotal phase 3 GATHER2 trial is currently underway to support the efficacy and safety of ACP as a potential treatment for GA.
Collapse
Affiliation(s)
| | - David R Lally
- New England Retina Consultants, Springfield, MA, USA
| | - Jason Hsu
- The Retina Service of Wills Eye Hospital, Wills Eye Physicians-Mid Atlantic Retina, Philadelphia, PA, USA
| | | | - David Eichenbaum
- Retina Vitreous Associates of Florida, St. Petersburg, FL, USA
- Morsani College of Medicine at The University of South Florida, Tampa, FL, USA
| | | | - Glenn J Jaffe
- Department of Ophthalmology, Duke University, Durham, NC, USA
| | | | | | | | - Arshad M Khanani
- Sierra Eye Associates and The University of Nevada, Reno School of Medicine, Reno, NV, USA.
| |
Collapse
|
10
|
Deng W, Yi C, Pan W, Liu J, Qi J, Chen J, Zhou Z, Duan Y, Ning X, Li J, Ye C, Chen Z, Xu H. Vascular Cell Adhesion Molecule-1 (VCAM-1) contributes to macular fibrosis in neovascular age-related macular degeneration through modulating macrophage functions. Immun Ageing 2023; 20:65. [PMID: 37985993 PMCID: PMC10659061 DOI: 10.1186/s12979-023-00389-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Accepted: 11/06/2023] [Indexed: 11/22/2023]
Abstract
BACKGROUND Neovascular age-related macular degeneration (nAMD) is a major cause of blindness in the elderly. The disease is due to the growth of abnormal blood vessels into the macula, leading to the loss of central vision. Intravitreal injection of vascular endothelial growth factor (VEGF) inhibitors (e.g., anti-VEGF) is the standard of care for nAMD. However, nearly 50% of patients do not respond or respond poorly to the therapy. More importantly, up to 70% of nAMD patients develop macular fibrosis after 10 years of anti-VEGF therapy. The underlying mechanism of nAMD-mediated macular fibrosis is unknown although inflammation is known to play an important role in the development of abnormal macular blood vessels and its progression to fibro-vascular membrane. In this study, we measured the intraocular levels of adhesion molecule VCAM-1, ICAM-1, CD44, CD62L, and CD62P in nAMD patients with and without macular fibrosis and investigated the link between the levels of adhesion molecule and clinical features (e.g., visual improvement, retinal thickness, etc.). We further investigated the effect of VCAM-1 in macrophage function in vitro and the development of subretinal fibrosis in vivo using a two-stage laser-induced protocol. RESULTS The aqueous levels of ICAM-1, VCAM-1, CD44, and CD62L were significantly higher in nAMD patients compared to cataract controls. The aqueous level of VCAM-1 (but not other adhesion molecules) was significantly higher in patients with macular fibrosis than those without and the level correlated positively with the retinal thickness. VCAM-1 was highly expressed at the lesion site in the mouse model of subretinal fibrosis. Blocking VCAM-1 or its receptor VLA-4 significantly prevented macrophage infiltration and reduced subretinal fibrosis in vivo. VCAM-1 induced macrophage migration and upregulated the expression of Arg-1, Mmp12 and Il6 but down-regulated the expression of iNOS and Il1b in macrophages. CONCLUSIONS VCAM-1 may contribute to the development of macular fibrosis in nAMD patients by modulating macrophage functions, including migration and profibrotic polarization.
Collapse
Affiliation(s)
- Wen Deng
- Aier School of Ophthalmology, Central South University, Changsha, China
- Aier Institute of Optometry and Vision Science, Changsha, China
| | - Caijiao Yi
- Aier Institute of Optometry and Vision Science, Changsha, China
| | - Wei Pan
- Aier Institute of Optometry and Vision Science, Changsha, China
| | - Jian Liu
- Aier Institute of Optometry and Vision Science, Changsha, China
| | - Jinyan Qi
- Aier School of Ophthalmology, Central South University, Changsha, China
- Aier Institute of Optometry and Vision Science, Changsha, China
| | - Juan Chen
- Changsha Aier Eye Hospital, Changsha, China
| | | | - Yiqin Duan
- Changsha Aier Eye Hospital, Changsha, China
| | | | - Jun Li
- Changsha Aier Eye Hospital, Changsha, China
| | - Changhua Ye
- Aier School of Ophthalmology, Central South University, Changsha, China
- Changsha Aier Eye Hospital, Changsha, China
| | - Zhongping Chen
- Aier School of Ophthalmology, Central South University, Changsha, China
- Changsha Aier Eye Hospital, Changsha, China
| | - Heping Xu
- Aier School of Ophthalmology, Central South University, Changsha, China.
- Aier Institute of Optometry and Vision Science, Changsha, China.
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, BT9 7BL, UK.
| |
Collapse
|
11
|
Liu D, Zhang C, Zhang J, Xu GT, Zhang J. Molecular pathogenesis of subretinal fibrosis in neovascular AMD focusing on epithelial-mesenchymal transformation of retinal pigment epithelium. Neurobiol Dis 2023; 185:106250. [PMID: 37536385 DOI: 10.1016/j.nbd.2023.106250] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 07/11/2023] [Accepted: 07/31/2023] [Indexed: 08/05/2023] Open
Abstract
Age-related macular degeneration (AMD) is a leading cause of vision loss among elderly people in developed countries. Neovascular AMD (nAMD) accounts for more than 90% of AMD-related vision loss. At present, intravitreal injection of anti-vascular endothelial growth factor (anti-VEGF) is widely used as the first-line therapy to decrease the choroidal and retinal neovascularizations, and thus to improve or maintain the visual acuity of the patients with nAMD. However, about 1/3 patients still progress to irreversible visual impairment due to subretinal fibrosis even with adequate anti-VEGF treatment. Extensive literatures support the critical role of epithelial-mesenchymal transformation (EMT) of retinal pigment epithelium (RPE) in the pathogenesis of subretinal fibrosis in nAMD, but the underlying mechanisms still remain largely unknown. This review summarized the molecular pathogenesis of subretinal fibrosis in nAMD, especially focusing on the transforming growth factor-β (TGF-β)-induced EMT pathways. It was also discussed how these pathways crosstalk and respond to signals from the microenvironment to mediate EMT and contribute to the progression of nAMD-related subretinal fibrosis. Targeting EMT signaling pathways might provide a promising and effective therapeutic strategy to treat subretinal fibrosis secondary to nAMD.
Collapse
Affiliation(s)
- Dandan Liu
- Department of Ophthalmology of Tongji Hospital and Laboratory of Clinical and Visual Sciences of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, China
| | - Chaoyang Zhang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University, Shanghai, China; National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Jingting Zhang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University, Shanghai, China; National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Guo-Tong Xu
- Department of Ophthalmology of Tongji Hospital and Laboratory of Clinical and Visual Sciences of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, China.
| | - Jingfa Zhang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University, Shanghai, China; National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China.
| |
Collapse
|
12
|
Jia H, Li T, Sun J, Gong Y, Liu H, Wang H, Chen J, Liu W, Lu S, Feng L, Wan Q, Qian L, Wang F, Liu X, Sun X. A Novel Bispecific Fusion Protein Targeting C3b/C4b and VEGF in Patients With nAMD: A Randomized, Open-Label, Phase 1b Study. Am J Ophthalmol 2023; 248:8-15. [PMID: 36410472 DOI: 10.1016/j.ajo.2022.11.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 11/09/2022] [Accepted: 11/11/2022] [Indexed: 11/23/2022]
Abstract
PURPOSE To evaluate the safety, tolerability, and efficacy of efdamrofusp alfa in patients with neovascular age-related macular degeneration (nAMD). DESIGN Prospective randomized, open-label, multiple ascending-dose, phase 1b study. METHODS Patients aged 50 years or older with active choroid neovascularization (CNV) secondary to nAMD were screened from 2 hospitals in 2 provinces in China. The first 9 patients were randomized 2:1 to intravitreally receive efdamrofusp alfa 2 mg at weeks 0, 4, and 8 or aflibercept 2 mg at weeks 0, 4, 8, and 16. After the dose-limiting toxicity assessment, 9 additional patients were randomized 2:1 to intravitreally receive efdamrofusp alfa 4 mg at weeks 0, 4, and 8 or aflibercept 2 mg at weeks 0, 4, 8, and 16. All patients were followed until week 20. Primary outcomes were safety and tolerability of efdamrofusp alfa. Secondary outcomes included changes from baseline in best-corrected visual acuity (BCVA), central subfield thickness (CST) as measured by spectral domain optical coherence tomography (SD-OCT), and CNV area as measured by fluorescein angiography (FA). RESULTS A total of 18 patients were enrolled. Six each of them received efdamrofusp alfa 2 mg, efdamrofusp alfa 4 mg, or aflibercept 2 mg, respectively. No dose-limiting toxicity was reported, and all patients completed the study. No ocular serious adverse events were reported. All ocular treatment-emergent adverse events were intravitreal injection related and were mild or moderate in severity. At week 20, mean changes from baseline in BCVA were 5.64 ± 3.56, 8.93 ± 3.59, and 7.92 ± 3.55 letters for patients receiving efdamrofusp alfa 2 mg, efdamrofusp alfa 4 mg and aflibercept 2 mg, respectively. Meanwhile, CST and CNV area reductions indicative of anatomic improvement were observed in the majority of the patients receiving both doses of efdamrofusp alfa and aflibercept. CONCLUSIONS Intravitreal efdamrofusp alfa dosed up to 4 mg every 4 weeks was well tolerated in nAMD patients with similar vision acuity and anatomic improvements.
Collapse
Affiliation(s)
- Huixun Jia
- From the Department of Ophthalmology (H.J., T.L., J.S., Y.G., H.L., H.W., J.C., W.L., F.W., X.S.) Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Clinical Research Center for Ophthalmic Diseases (H.J., T.L., J.S., J.C., W.L., F.W. X.S.), Shanghai, China; Shanghai Key Laboratory of Fundus Diseases (H.J., F.W., X.S.) Shanghai, China
| | - Tong Li
- From the Department of Ophthalmology (H.J., T.L., J.S., Y.G., H.L., H.W., J.C., W.L., F.W., X.S.) Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Clinical Research Center for Ophthalmic Diseases (H.J., T.L., J.S., J.C., W.L., F.W. X.S.), Shanghai, China
| | - Junran Sun
- From the Department of Ophthalmology (H.J., T.L., J.S., Y.G., H.L., H.W., J.C., W.L., F.W., X.S.) Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Clinical Research Center for Ophthalmic Diseases (H.J., T.L., J.S., J.C., W.L., F.W. X.S.), Shanghai, China
| | - Yuanyuan Gong
- From the Department of Ophthalmology (H.J., T.L., J.S., Y.G., H.L., H.W., J.C., W.L., F.W., X.S.) Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haiyun Liu
- From the Department of Ophthalmology (H.J., T.L., J.S., Y.G., H.L., H.W., J.C., W.L., F.W., X.S.) Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hong Wang
- From the Department of Ophthalmology (H.J., T.L., J.S., Y.G., H.L., H.W., J.C., W.L., F.W., X.S.) Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jieqiong Chen
- From the Department of Ophthalmology (H.J., T.L., J.S., Y.G., H.L., H.W., J.C., W.L., F.W., X.S.) Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Clinical Research Center for Ophthalmic Diseases (H.J., T.L., J.S., J.C., W.L., F.W. X.S.), Shanghai, China
| | - Wenjia Liu
- From the Department of Ophthalmology (H.J., T.L., J.S., Y.G., H.L., H.W., J.C., W.L., F.W., X.S.) Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Clinical Research Center for Ophthalmic Diseases (H.J., T.L., J.S., J.C., W.L., F.W. X.S.), Shanghai, China
| | - Shujie Lu
- Innovent Biologics, Inc. (S.L., L.F., Q.W., L.Q.), Suzhou, China
| | - Liqi Feng
- Innovent Biologics, Inc. (S.L., L.F., Q.W., L.Q.), Suzhou, China
| | - Qiuchen Wan
- Innovent Biologics, Inc. (S.L., L.F., Q.W., L.Q.), Suzhou, China
| | - Lei Qian
- Innovent Biologics, Inc. (S.L., L.F., Q.W., L.Q.), Suzhou, China
| | - Fenghua Wang
- From the Department of Ophthalmology (H.J., T.L., J.S., Y.G., H.L., H.W., J.C., W.L., F.W., X.S.) Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Clinical Research Center for Ophthalmic Diseases (H.J., T.L., J.S., J.C., W.L., F.W. X.S.), Shanghai, China; Shanghai Key Laboratory of Fundus Diseases (H.J., F.W., X.S.) Shanghai, China
| | - Xiaoling Liu
- Eye Hospital of Wenzhou Medical University (X.L.), Wenzhou, China
| | - Xiaodong Sun
- From the Department of Ophthalmology (H.J., T.L., J.S., Y.G., H.L., H.W., J.C., W.L., F.W., X.S.) Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Clinical Research Center for Ophthalmic Diseases (H.J., T.L., J.S., J.C., W.L., F.W. X.S.), Shanghai, China; Shanghai Key Laboratory of Fundus Diseases (H.J., F.W., X.S.) Shanghai, China; Shanghai Engineering Center for Visual Science and Photomedicine (X.S.), Shanghai, China.
| |
Collapse
|
13
|
Higashijima F, Hasegawa M, Yoshimoto T, Kobayashi Y, Wakuta M, Kimura K. Molecular mechanisms of TGFβ-mediated EMT of retinal pigment epithelium in subretinal fibrosis of age-related macular degeneration. FRONTIERS IN OPHTHALMOLOGY 2023; 2:1060087. [PMID: 38983569 PMCID: PMC11182173 DOI: 10.3389/fopht.2022.1060087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 12/30/2022] [Indexed: 07/11/2024]
Abstract
Age-related macular degeneration (AMD) is one of the leading causes of blindness in the elderly, affecting the macula of the retina and resulting in vision loss. There are two types of AMD, wet and dry, both of which cause visual impairment. Wet AMD is called neovascular AMD (nAMD) and is characterized by the formation of choroidal neovascular vessels (CNVs) in the macula. nAMD can be treated with intravitreal injections of vascular endothelial growth factor (VEGF) inhibitors, which help improve vision. However, approximately half the patients do not achieve satisfactory results. Subretinal fibrosis often develops late in nAMD, leading to irreversible photoreceptor degeneration and contributing to visual loss. Currently, no treatment exists for subretinal fibrosis, and the molecular mechanisms of fibrous tissue formation following neovascular lesions remain unclear. In this review, we describe the clinical features and molecular mechanisms of macular fibrosis secondary to nAMD. Myofibroblasts play an essential role in the development of fibrosis. This review summarizes the latest findings on the clinical features and cellular and molecular mechanisms of the pathogenesis of subretinal fibrosis in nAMD and discusses the potential therapeutic strategies to control subretinal fibrosis in the future.
Collapse
Affiliation(s)
| | | | | | | | | | - Kazuhiro Kimura
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
| |
Collapse
|
14
|
de Jong S, Tang J, Clark SJ. Age-related macular degeneration: A disease of extracellular complement amplification. Immunol Rev 2023; 313:279-297. [PMID: 36223117 DOI: 10.1111/imr.13145] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Age-related macular degeneration (AMD) is a major cause of vision impairment in the Western World, and with the aging world population, its incidence is increasing. As of today, for the majority of patients, no treatment exists. Multiple genetic and biochemical studies have shown a strong association with components in the complement system and AMD, and evidence suggests a major role of remodeling of the extracellular matrix underlying the outer blood/retinal barrier. As part of the innate immune system, the complement cascade acts as a first-line defense against pathogens, and upon activation, its amplification loop ensures a strong, rapid, and sustained response. Excessive activation, however, can lead to host tissue damage and cause complement-associated diseases like AMD. AMD patients present with aberrant activation of the alternative pathway, especially in ocular tissues but also on a systemic level. Here, we review the latest findings of complement activation in AMD, and we will discuss in vivo observations made in human tissue, cellular models, the potential synergy of different AMD-associated pathways, and conclude on current clinical trials and the future outlook.
Collapse
Affiliation(s)
- Sarah de Jong
- Department for Ophthalmology, University Eye Clinic, Eberhard Karls University of Tübingen, Tübingen, Germany.,Department for Ophthalmology, Institute for Ophthalmic Research, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Jiaqi Tang
- Department for Ophthalmology, University Eye Clinic, Eberhard Karls University of Tübingen, Tübingen, Germany.,Department for Ophthalmology, Institute for Ophthalmic Research, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Simon J Clark
- Department for Ophthalmology, University Eye Clinic, Eberhard Karls University of Tübingen, Tübingen, Germany.,Department for Ophthalmology, Institute for Ophthalmic Research, Eberhard Karls University of Tübingen, Tübingen, Germany.,Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| |
Collapse
|
15
|
Suemanotham N, Photcharatinnakorn P, Chantong B, Buranasinsup S, Phochantachinda S, Sakcamduang W, Reamtong O, Thiangtrongjit T, Chatchaisak D. Curcuminoid supplementation in canine diabetic mellitus and its complications using proteomic analysis. Front Vet Sci 2022; 9:1057972. [PMID: 36619946 PMCID: PMC9816143 DOI: 10.3389/fvets.2022.1057972] [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: 09/30/2022] [Accepted: 12/09/2022] [Indexed: 12/24/2022] Open
Abstract
Introduction Inflammation and oxidative stress contribute to diabetes pathogenesis and consequences. Therapeutic approaches for canine diabetes remain a challenge. Curcumin has anti-inflammatory and anti-oxidative effects and is beneficial for humans with diabetes mellitus (DM); however, data on its impact on canine diabetes is limited. This study aimed to evaluate the potential for causing adverse effects, anti-inflammatory effects, anti-oxidative effects and proteomic patterns of curcuminoid supplementation on canine DM. Methods Altogether, 18 dogs were divided into two groups: DM (n = 6) and healthy (n = 12). Curcuminoid 250 mg was given to the DM group orally daily for 180 days. Blood and urine sample collection for hematological parameters, blood biochemistry, urinalysis, oxidative stress parameters, inflammatory markers and proteomics were performed every 6 weeks. Results and discussion Curcuminoid supplementation with standard therapy significantly decreased oxidative stress with the increased glutathione/oxidized glutathione ratio, but cytokine levels were unaffected. According to the proteomic analysis, curcuminoid altered the expression of alpha-2-HS-glycoprotein, transthyretin, apolipoprotein A-I and apolipoprotein A-IV, suggesting that curcuminoid improves insulin sensitivity and reduces cardiovascular complications. No negative impact on clinical symptoms, kidneys or liver markers was identified. This study proposed that curcuminoids might be used as a targeted antioxidant strategy as an adjunctive treatment to minimize diabetes complications in dogs.
Collapse
Affiliation(s)
- Namphung Suemanotham
- Department of Clinical Sciences and Public Health, Faculty of Veterinary Science, Mahidol University, Nakhon Pathom, Thailand,Department of Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | | | - Boonrat Chantong
- Department of Pre-clinic and Applied Animal Science, Faculty of Veterinary Science, Mahidol University, Nakhon Pathom, Thailand
| | - Shutipen Buranasinsup
- Department of Pre-clinic and Applied Animal Science, Faculty of Veterinary Science, Mahidol University, Nakhon Pathom, Thailand
| | - Sataporn Phochantachinda
- Department of Clinical Sciences and Public Health, Faculty of Veterinary Science, Mahidol University, Nakhon Pathom, Thailand
| | - Walasinee Sakcamduang
- Department of Clinical Sciences and Public Health, Faculty of Veterinary Science, Mahidol University, Nakhon Pathom, Thailand
| | - Onrapak Reamtong
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Tipparat Thiangtrongjit
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Duangthip Chatchaisak
- Department of Clinical Sciences and Public Health, Faculty of Veterinary Science, Mahidol University, Nakhon Pathom, Thailand,*Correspondence: Duangthip Chatchaisak ✉
| |
Collapse
|
16
|
Liisborg C. Age-related macular degeneration and myeloproliferative neoplasms - A common pathway. Acta Ophthalmol 2022; 100 Suppl 271:3-35. [PMID: 36200281 PMCID: PMC9828081 DOI: 10.1111/aos.15247] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 08/22/2021] [Indexed: 01/12/2023]
Abstract
DANSK RESUMÉ (DANISH SUMMARY): Aldersrelateret makuladegeneration (AMD) er den hyppigste årsag til uopretteligt synstab og blindhed i højindkomstlande. Det er en progredierende nethindesygdom som gradvist fører til ødelaeggelse af de celler som er ansvarlige for vores centralsyn. De tidlige stadier er ofte asymptomatiske, imens senstadie AMD, som opdeles i to former, neovaskulaer AMD (nAMD) og geografisk atrofi (GA), begge udviser gradvist synstab, dog generelt med forskellig hastighed. Tidlig AMD er karakteriseret ved tilstedevaerelsen af druser og pigmentforandringer i nethinden mens nAMD og GA udviser henholdsvis karnydannelse i og atrofi af nethinden. AEtiologien er multifaktoriel og udover alder omfatter patogenesen miljø- og genetiske risikofaktorer. Forskning har specielt fokuseret på lokale forandringer i øjet hvor man har fundet at inflammation spiller en betydelig rolle for udviklingen af sygdommen, men flere studier tyder også på at systemiske forandringer og specielt systemisk inflammation spiller en vaesentlig rolle i patogenesen. De Philadelphia-negative myeloproliferative neoplasier (MPNs) er en gruppe af haematologiske kraeftsygdomme med en erhvervet genetisk defekt i den tidlige pluripotente stamcelle som medfører en overproduktion af en eller flere af blodets modne celler. Sygdommene er fundet at udvikle sig i et biologisk kontinuum fra tidligt cancerstadie, essentiel trombocytose (ET) over polycytaemi vera (PV) og endelig til det sene myelofibrose stadie (PMF). Symptomer hos disse patienter skyldes isaer den aendrede sammensaetning af blodet, hyperviskositet, kompromitteret mikrocirkulation og nedsat vaevsgennemblødning. Den øgede morbiditet og mortalitet beror i høj grad på tromboembolier, blødninger og leukemisk transformation. En raekke mutationer som driver MPN sygdommene er identificeret, bl.a. JAK2V617F-mutationen som medfører en deregulering JAK/STAT signalvejen, der bl.a. har betydning for cellers vaekst og overlevelse. Et tidligere stort registerstudie har vist at patienter med MPNs har en øget risiko for neovaskulaer AMD og et pilotstudie har vist øget forekomst af intermediaer AMD. Dette ønsker vi at undersøge naermere i et større studie i dette Ph.d.- projekt. Flere studier har også vist at kronisk inflammation spiller en vigtig rolle for både initiering og udvikling af den maligne celleklon hos MPNs og herfra er en "Human Inflammationsmodel" blevet udviklet. Siden er MPN sygdommene blevet anvendt som "model sygdomme" for en tilsvarende inflammationsmodel for udvikling af Alzheimers sygdom. I dette Ph.d.-projekt vil vi tilsvarende forsøge at undersøge systemisk inflammation i forhold til forekomst af druser. Det vil vi gøre ved at sammenligne systemiske immunologiske markører som tidligere er undersøgt hos patienter med AMD og sammenligne med MPN. Specielt er vi interesseret i systemiske immunologiske forskelle på patienter med MPN og druser (MPNd) og MPN med normale nethinder (MPNn). Denne afhandling består af to overordnede studier. I Studie I, undersøgte vi forekomsten af retinale forandringer associeret med AMD hos 200 patienter med MPN (artikel I). Studie II, omhandlede immunologiske ligheder ved AMD og MPN, og var opdelt i yderligere tre delstudier hvor vi undersøgte hhv. systemiske markører for inflammation, aldring og angiogenese (artikel II, III og IV). Vi undersøgte markørerne i fire typer af patienter: nAMD, intermediaer AMD (iAMD), MPNd og MPNn. Undersøgelsen af forskelle mellem MPNd og MPNn, vil gøre det muligt at identificere forandringer i immunsystemet som kunne vaere relevante for AMD-patogenesen. Vi vil endvidere sammenholde resultaterne for patienter med MPN med patienter som har iAMD og nAMD. I studie I (Artikel I) fandt vi at patienter med MPN har en signifikant højere praevalens af store druser og AMD tidligere i livet sammenlignet med estimater fra tre store befolkningsundersøgelser. Vi fandt også at forekomst af druser var associeret med højere neutrofil-lymfocyt ratio, hvilket indikerer et højere niveau af kronisk inflammation i patienterne med druser sammenlignet med dem uden druser. I studie II (Artikel II, III og IV) fandt vi flere immunologiske forskelle mellem patienter med MPNd og MPNn. Da vi undersøgte markører for inflammation, fandt vi en højere grad af systemisk inflammation i MPNd end MPNn. Dette blev vist ved en højere inflammationsscore (udregnet på baggrund af niveauer af pro-inflammatoriske markører), en højere neutrofil-lymfocyt ratio, samt indikationer på et dereguleret komplementsystem. Ved undersøgelse af aldringsmarkører fandt vi tegn på accelereret immunaldring hos MPNd i forhold til MPNn, hvilket kommer til udtryk ved en større procentdel af "effector memory T celler". Endelig fandt vi en vaesentlig lavere ekspression af CXCR3 på T celler og monocytter hos patienter med nAMD sammenlignet med iAMD, MPNd og MPNn. Dette er i overensstemmelse med tidligere studier hvor CXCR3 ekspression er fundet lavere end hos raske kontroller. Derudover fandt vi en faldende CXCR3 ekspression på monocytter over det biologiske MPN-kontinuum. Disse studier indikerer en involvering af CXCR3 i både nAMD og PMF, begge sygdomsstadier som er karakteriseret ved angiogenese og fibrose. Ud fra resultaterne af denne afhandling kan vi konkludere at forekomsten af druser og AMD hos MPN er øget i forhold til baggrundsbefolkningen. Endvidere viser vores resultater at systemisk inflammation muligvis spiller en vaesentlig større rolle i udviklingen af AMD end tidligere antaget. Vi foreslår derfor en AMD-model (Figur 18) hvor inflammation kan initiere og accelerere den normale aldersafhaengige akkumulation af affaldsstoffer i nethinden, som senere udvikler sig til druser, medførende øget lokal inflammation og med tiden tidlig og intermediaer AMD. Dette resulterer i den øgede risiko for udvikling til de invaliderende senstadier af AMD. ENGLISH SUMMARY: Age-related macular degeneration (AMD) is the most common cause of irreversible vision loss and blindness in high-income countries. It is a progressive retinal disease leading to damage of the cells responsible for central vision. The early stages of the disease are often asymptomatic, while late-stage AMD, which is divided into two entities, neovascular AMD and geographic atrophy (GA), both show vision loss, though generally with different progression rates. Drusen and pigmentary abnormalities in the retina characterise early AMD, while nAMD and GA show angiogenesis in and atrophy of the retina, respectively. The aetiology is multifactorial and, in addition to ageing, which is the most significant risk factor for developing AMD, environmental- and genetic risk factors are implicated in the pathogenesis. Research has focused on local changes in the eye where inflammation has been found to play an essential role, but studies also point to systemic alterations and especially systemic inflammation to be involved in the pathogenesis. The Philadelphia-negative myeloproliferative neoplasms (MPN) are a group of haematological cancers with an acquired genetic defect of the pluripotent haematopoietic stem cell, characterised by excess haematopoiesis of the myeloid cell lineage. The diseases have been found to evolve in a biological continuum from early cancer state, essential thrombocythemia, over polycythaemia vera (PV), to the advanced myelofibrosis stage (PMF). The symptoms in these patients are often a result of the changes in the blood composition, hyperviscosity, microvascular disturbances, and reduced tissue perfusion. The major causes of morbidity and mortality are thromboembolic- and haemorrhagic events, and leukemic transformation. A group of mutations that drive the MPNs has been identified, e.g., the JAK2V617F mutation, which results in deregulation of the JAK/STAT signal transduction pathway important, for instance, in cell differentiation and survival. A previous large register study has shown that patients with MPNs have an increased risk of neovascular AMD, and a pilot study has shown an increased prevalence of intermediate AMD. We wish to study this further in a larger scale study. Several studies have also shown that systemic inflammation plays an essential role in both the initiation and progression of the malignant cell clone in MPNs. From this knowledge, a "Human inflammation model" has been developed. Since then, the MPNs has been used as model diseases for a similar inflammation model for the development of Alzheimer's disease. In this PhD project, we would like to investigate systemic inflammation in relation to drusen presence. We will do this by comparing systemic immunological markers previously investigated in patients with AMD and compare with MPN. We are primarily interested in systemic immunological differences between patients with MPN and drusen (MPNd) and MPN with normal retinas (MPNn). This thesis consists of two main studies. Study I investigated the prevalence of retinal changes associated with AMD and the prevalence of different AMD stages in 200 patients with MPN (paper I). Study II examined immunological similarities between AMD and MPNs. This study was divided into three substudies exploring systemic markers of inflammation, ageing and angiogenesis, respectively. This was done in four types of patients: nAMD, intermediate AMD (iAMD), MPNd and MPNn. Investigating, differences between MPNd and MPNn, will make it possible to identify changes in the immune system, relevant for AMD pathogenesis. Additionally, we will compare patients with MPNs with patients with iAMD and nAMD. In study I (Paper I), we found that patients with MPNs have a significantly higher prevalence of large drusen and consequently AMD from an earlier age compared to the estimates from three large population-based studies. We also found that drusen prevalence was associated with a higher neutrophil-to-lymphocyte ratio indicating a higher level of chronic low-grade inflammation in patients with drusen compared to those without drusen. In study II (papers II, III and IV), we found immunological differences between patients with MPNd and MPNn. When we investigated markers of inflammation, we found a higher level of systemic inflammation in MPNd than MPNn. This was indicated by a higher inflammation score (based on levels of pro-inflammatory markers), a higher neutrophil-to-lymphocyte ratio, and indications of a deregulated complement system. When examining markers of ageing, we found signs of accelerated immune ageing in MPNd compared to MPNn, shown by more senescent effector memory T cells. Finally, when exploring a marker of angiogenesis, we found a lower CXCR3 expression on monocytes and T cells in nAMD compared to iAMD, MPNd and MPNn, in line with previous studies of nAMD compared to healthy controls. Further, we found decreasing CXCR3 expression over the MPN biological continuum. These studies indicate CXCR3 involvement in both nAMD and PMF, two disease stages characterised by angiogenesis and fibrosis. From the results of this PhD project, we can conclude that the prevalence of drusen and AMD is increased in patients with MPN compared to the general population. Further, our results show that systemic inflammation may play a far more essential role in AMD pathogenesis than previously anticipated. We, therefore, propose an AMD model (Figure 18) where inflammation can initiate and accelerate the normal age-dependent accumulation of debris in the retina, which later evolve into drusen, resulting in increased local inflammation, and over time early- and intermediate AMD. This results in the increased risk of developing the late debilitating stages of AMD.
Collapse
|
17
|
Patel PN, Patel PA, Land MR, Bakerkhatib-Taha I, Ahmed H, Sheth V. Targeting the Complement Cascade for Treatment of Dry Age-Related Macular Degeneration. Biomedicines 2022; 10:1884. [PMID: 36009430 PMCID: PMC9405256 DOI: 10.3390/biomedicines10081884] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/28/2022] [Accepted: 08/01/2022] [Indexed: 11/16/2022] Open
Abstract
Age-related macular degeneration (AMD) is the leading cause of irreversible vision loss in the elderly population. AMD is characterized in its late form by neovascularization (wet type) or geographic atrophy of the retinal pigment epithelium cell layer (dry type). Regarding the latter type, there is growing evidence supporting an association between the pathophysiology of dry AMD and key proteins in the complement cascade. The complement cascade works as a central part of the innate immune system by defending against foreign pathogens and modified self-tissues. Through three distinct pathways, a series of plasma and membrane-associated serum proteins are activated upon identification of a foreign entity. Several of these proteins have been implicated in the development and progression of dry AMD. Potential therapeutic targets include C1q, C3, C5, complement factors (B, D, H, I), membrane attack complex, and properdin. In this review, we provide an understanding of the role of the complement system in dry AMD and discuss the emerging therapies in early phase clinical trials. The tentative hope is that these drugs may offer the potential to intervene at earlier stages in dry AMD pathogenesis, thereby preventing progression to late disease.
Collapse
Affiliation(s)
- Prem N. Patel
- Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Parth A. Patel
- Department of Ophthalmology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Matthew R. Land
- Department of Ophthalmology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | | | - Harris Ahmed
- Department of Ophthalmology, Loma Linda University Medical Center, Loma Linda, CA 92350, USA
| | - Veeral Sheth
- University Retina and Macula Associates, Oak Forest, IL 60452, USA
| |
Collapse
|
18
|
Classical complement pathway factor alterations in narcolepsy. Acta Neuropsychiatr 2022; 34:212-219. [PMID: 35034679 DOI: 10.1017/neu.2021.42] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Narcolepsy is a chronic sleep disorder long hypothesised to be an autoimmune disease. Complement-mediated immune mechanisms have not been investigated in detail in narcolepsy. Our aim was to establish the significance of classical pathway activation in narcolepsy. METHODS Sera of 42 narcolepsy patients and 26 healthy controls were screened with ELISA to determine the levels of C1q, C3a, C4d and complement component 4 binding protein (C4BP). A home-made ELISA method was developed to detect antibodies to C4BP-alpha (anti-C4BPA). The correlation between complement levels and clinical findings was examined. RESULTS C1q levels were significantly higher in narcolepsy patients while C4d and C4BP levels were significantly lower compared to healthy controls. C3a levels were comparable among patients and controls. Eleven narcolepsy patients showed serum anti-C4BPA levels. Total rapid eye movements (REM) time, sleep onset latency, REM sleep latency, sleep activity, percentage of wakefulness after sleep onset and Epworth sleepiness scale scores were correlated with levels of different complement factors. CONCLUSION Complement-mediated immune mechanisms might partake in narcolepsy pathogenesis. The precise role of autoantibodies on complement level alterations needs to be investigated. Levels of complement factors and degradation products may potentially be utilised as biomarkers to predict the clinical severity of narcolepsy.
Collapse
|
19
|
Llorián-Salvador M, Byrne EM, Szczepan M, Little K, Chen M, Xu H. Complement activation contributes to subretinal fibrosis through the induction of epithelial-to-mesenchymal transition (EMT) in retinal pigment epithelial cells. J Neuroinflammation 2022; 19:182. [PMID: 35831910 PMCID: PMC9447479 DOI: 10.1186/s12974-022-02546-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 06/22/2022] [Indexed: 11/16/2022] Open
Abstract
Background We previously reported higher plasma levels of complement fragments C3a and C5a in neovascular Age-related Macular Degeneration (nAMD) patients with macular fibrosis. This study aimed to understand whether complement activation contributes to the development of macular fibrosis and the underlying mechanisms involved. Methods Complement activation was blocked using a C5 neutralizing antibody (BB5.1) in C57BL/6J mice after induction of subretinal fibrosis using the two-stage laser protocol. Fibrotic lesions were examined 10 days after the 2nd laser through fundus examination and immunohistochemistry. The expression of C5aR in fibrotic lesions and retinal pigment epithelial (RPE) cultures were examined by confocal microscopy. Primary murine RPE cells were treated with C3a or C5a (10–100 ng/mL) or TGF-β2 (10 ng/mL). Epithelial-to-mesenchymal transition (EMT) was assessed through various readouts. The expression of E-cadherin, vimentin, fibronectin, α-SMA, Slug, ERK/AKT and pSMAD2/3 were determined by Western blot and immunocytochemistry. Collagen contraction and wound-healing assays were used as functional readouts of EMT. The production of IL-6, TGF-β1, TGF-β2 and VEGF by RPE cells were determined by ELISA. PMX53 was used to block C5aR in RPE cultures and in vivo in mice with subretinal fibrosis. Results Extensive C5b-9 deposition was detected at the site of subretinal fibrosis. BB5.1 treatment completely abrogated complement activation and significantly reduced subretinal fibrosis. C5aR was detected in RPE and infiltrating MHC-II+ cells in subretinal fibrosis. In vitro, RPE cells constitutively express C5/C5a and C5aR, and their expression was increased by TGF-β2 treatment. C5a but not C3a increased fibronectin, α-SMA, vimentin and Slug expression, and decreased E-cadherin expression in RPE cells. C5a treatment also increased the contractility and migration of RPE cells and enhanced the production of VEGF and TGF-β1/2. C5a treatment induced pSmad2/3 and pERK1/2 expression in RPE cells and this was blocked by PMX53. PMX53 treatment significantly reduced sodium fluorescein leakage in the subretinal fibrosis model, while collagen-I+ lesions only mildly reduced. Conclusions Complement activation is critically involved in the development of subretinal fibrosis, partially through C5a–C5aR-mediated EMT in RPE cells. Targeting complement activation rather than C5a may be a novel approach for the management of macular fibrosis. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02546-3.
Collapse
Affiliation(s)
- María Llorián-Salvador
- The Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, Northern Ireland, UK.,Vall d´Hebron Research Institute (VHIR), Universitat Autonòma de Barcelona, 08035, Barcelona, Spain
| | - Eimear M Byrne
- The Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, Northern Ireland, UK.,Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, 08003, Barcelona, Spain
| | - Manon Szczepan
- The Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, Northern Ireland, UK
| | - Karis Little
- The Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, Northern Ireland, UK
| | - Mei Chen
- The Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, Northern Ireland, UK
| | - Heping Xu
- The Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, Northern Ireland, UK.
| |
Collapse
|
20
|
Szczepan M, Llorián-Salvador M, Chen M, Xu H. Immune Cells in Subretinal Wound Healing and Fibrosis. Front Cell Neurosci 2022; 16:916719. [PMID: 35755781 PMCID: PMC9226489 DOI: 10.3389/fncel.2022.916719] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 05/16/2022] [Indexed: 11/22/2022] Open
Abstract
The subretinal space is devoid of any immune cells under normal conditions and is an immune privileged site. When photoreceptors and/or retinal pigment epithelial cells suffer from an injury, a wound healing process will be initiated. Retinal microglia and the complement system, as the first line of retinal defense, are activated to participate in the wound healing process. If the injury is severe or persists for a prolonged period, they may fail to heal the damage and circulating immune cells will be summoned leading to chronic inflammation and abnormal wound healing, i.e., subretinal or intraretinal fibrosis, a sight-threatening condition frequently observed in rhematogenous retinal detachment, age-related macular degeneration and recurrent uveoretinitis. Here, we discussed the principles of subretinal wound healing with a strong focus on the conditions whereby the damage is beyond the healing capacity of the retinal defense system and highlighted the roles of circulating immune cells in subretinal wound healing and fibrosis.
Collapse
Affiliation(s)
- Manon Szczepan
- The Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Sciences, Queen’s University Belfast, Belfast, United Kingdom
| | - María Llorián-Salvador
- The Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Sciences, Queen’s University Belfast, Belfast, United Kingdom
| | - Mei Chen
- The Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Sciences, Queen’s University Belfast, Belfast, United Kingdom
| | - Heping Xu
- The Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Sciences, Queen’s University Belfast, Belfast, United Kingdom,Aier Institute of Optometry and Vision Science, Changsha, China,*Correspondence: Heping Xu,
| |
Collapse
|
21
|
Marin AI, Poppelaars F, Wagner BD, Palestine AG, Patnaik JL, Holers VM, Frazer-Abel AA, Mathias MT, Manoharan N, Fonteh CN, Mandava N, Lynch AM. Sex and Age-Related Differences in Complement Factors Among Patients With Intermediate Age-Related Macular Degeneration. Transl Vis Sci Technol 2022; 11:22. [PMID: 35594041 PMCID: PMC9145081 DOI: 10.1167/tvst.11.5.22] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Purpose Age-related macular degeneration (AMD) is an acquired degenerative disease of the retina classified into early, intermediate, and advanced AMD. A key factor in the pathogenesis of AMD is the complement system. The interaction of age and sex with the complement system may affect the risk of developing AMD. The purpose of this study was to determine if there were sex-specific differences in levels of complement factors among patients with the intermediate phenotype of AMD (iAMD) and explore the correlation between age and complement proteins. Methods We studied complement factors in patients with iAMD and controls without AMD. Nonparametric, rank-based linear regressions including a sex by AMD interaction were used to compare levels for each analyte. Correlations between age and complement proteins were evaluated using the Spearman rank correlation coefficient. Results We found significantly higher levels of factor B and factor I in females compared with males with iAMD, whereas no differences were seen in complement levels in male and female controls. The ratios of Ba/factor B, C3a/C3, C4b/C4, and C5a/C5 were not different in males and females with iAMD. Conclusions We demonstrate disparities in a subset of systemic complement factors between females and males with iAMD, but apparent complement turnover as measured by ratios of activation fragments to intact molecules was not different between these groups. The results suggest that complement system levels, including complement regulator factor I, exhibits sex-related differences in patients with iAMD and highlights that stratification by sex might be helpful in the interpretation of clinical trials of anticomplement therapy.
Collapse
Affiliation(s)
- A Itzam Marin
- Department of Ophthalmology, University of Colorado Denver School of Medicine, Aurora, Colorado, USA
| | - Felix Poppelaars
- Division of Nephrology and Hypertension, University of Colorado Denver School of Medicine, Aurora, Colorado, USA
| | - Brandie D Wagner
- Department of Biostatistics and Informatics, University of Colorado School of Public Health, Aurora, Colorado, USA
| | - Alan G Palestine
- Department of Ophthalmology, University of Colorado Denver School of Medicine, Aurora, Colorado, USA
| | - Jennifer L Patnaik
- Department of Ophthalmology, University of Colorado Denver School of Medicine, Aurora, Colorado, USA
| | - V Michael Holers
- Departments of Medicine and Immunology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Ashley A Frazer-Abel
- Exsera BioLabs, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Marc T Mathias
- Department of Ophthalmology, University of Colorado Denver School of Medicine, Aurora, Colorado, USA
| | - Niranjan Manoharan
- Department of Ophthalmology, University of Colorado Denver School of Medicine, Aurora, Colorado, USA
| | - Cheryl N Fonteh
- Department of Ophthalmology, University of Colorado Denver School of Medicine, Aurora, Colorado, USA
| | - Naresh Mandava
- Department of Ophthalmology, University of Colorado Denver School of Medicine, Aurora, Colorado, USA
| | - Anne M Lynch
- Department of Ophthalmology, University of Colorado Denver School of Medicine, Aurora, Colorado, USA
| | | |
Collapse
|
22
|
Yi C, Liu J, Deng W, Luo C, Qi J, Chen M, Xu H. Macrophage elastase (MMP12) critically contributes to the development of subretinal fibrosis. J Neuroinflammation 2022; 19:78. [PMID: 35382832 PMCID: PMC8985356 DOI: 10.1186/s12974-022-02433-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 03/22/2022] [Indexed: 11/16/2022] Open
Abstract
Background Macular subretinal fibrosis is the end-stage complication of neovascular age-related macular degeneration (nAMD). We previously developed a mouse model of two-stage laser-induced subretinal fibrosis that mimics closely the dynamic course of macular fibrosis in nAMD patients. This study was aimed to understand the molecular mechanism of subretinal fibrosis. Methods Subretinal fibrosis was induced in C57BL/6J mice using the two-stage laser-induced protocol. Twenty days later, eyes were collected and processed for RNA sequencing (RNA-seq) analysis. DESeq2 was used to determine the differentially expressed genes (DEGs). Gene Ontology (GO) and KEGG were used to analyze the enriched pathways. The expression of the selected DEGs including Mmp12 was verified by qPCR. The expression of MMP12 in subretinal fibrosis of mouse and nAMD donor eyes was examined by immunofluorescence and confocal microscopy. The expression of collagen 1, αSMA and fibronectin and cytokines in bone marrow-derived macrophages from control and subretinal fibrosis mice were examined by qPCR, immunocytochemistry and Luminex multiplex cytokine assay. The MMP12 specific inhibitor MMP408 was used to evaluate the effect of MMP12 on TGFβ-induced macrophage-to-myofibroblast transition (MMT) in vitro and its role in subretinal fibrosis in vivo. Results RNA-seq analysis of RPE-choroid from subretinal fibrosis eyes uncovered 139 DEGs (fold change log2(fc) ≥ 0.5, FDR < 0.05), including 104 up-regulated and 35 were down-regulated genes. The top 25 enrichment GO terms were related to inflammation, blood vessels/cardiovascular development and angiogenesis. One of the most significantly upregulated genes, Mmp12, contributed to 12 of the top 25 GO terms. Higher levels of MMP12 were detected in subretinal fibrotic lesions in nAMD patients and the mouse model, including in F4/80+ or Iba1+ macrophages. BMDMs from subretinal fibrosis mice expressed higher levels of MMP12, collagen-1, αSMA and fibronectin. MMP408 dose-dependently suppressed TGFβ-induced MMT in BMDMs. In vivo treatment with MMP408 (5 mg/kg) significantly reduced subretinal fibrosis accompanied by reduced F4/80+ macrophage infiltration. Conclusions MMP12 critically contributes to the development of subretinal fibrosis, partially through promoting MMT. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02433-x.
Collapse
Affiliation(s)
- Caijiao Yi
- Aier School of Ophthalmology, Central South University, Changsha, 410000, China
| | - Jian Liu
- Aier Institute of Optometry and Vision Science, Changsha, 410000, China
| | - Wen Deng
- Aier School of Ophthalmology, Central South University, Changsha, 410000, China
| | - Chang Luo
- Aier School of Ophthalmology, Central South University, Changsha, 410000, China
| | - Jinyan Qi
- Aier School of Ophthalmology, Central South University, Changsha, 410000, China
| | - Mei Chen
- The Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK
| | - Heping Xu
- Aier School of Ophthalmology, Central South University, Changsha, 410000, China. .,Aier Institute of Optometry and Vision Science, Changsha, 410000, China. .,The Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK.
| |
Collapse
|
23
|
Wagner BD, Patnaik JL, Palestine AG, Frazer-Abel AA, Baldermann R, Holers VM, Mathias MT, Mandava N, Lynch AM. Association of Systemic Inflammatory Factors with Progression to Advanced Age-related Macular Degeneration. Ophthalmic Epidemiol 2022; 29:139-148. [PMID: 33827374 PMCID: PMC8497647 DOI: 10.1080/09286586.2021.1910314] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 02/01/2021] [Accepted: 03/22/2021] [Indexed: 01/10/2023]
Abstract
PURPOSE Age-related macular degeneration (AMD) is a leading cause of vision loss in the elderly. The role of systemic inflammation in AMD remains unclear specifically in patients with intermediate AMD (iAMD). We sought to determine whether systemic inflammation was associated with future iAMD progression. METHODS Combinations of 27 circulating inflammatory markers including complement factors, cytokines, chemokines, and high-sensitivity C-reactive protein (hsCRP) were evaluated in iAMD patients recruited into a Colorado AMD registry. Systemic inflammatory markers were combined using principal component analysis. Risk factors for AMD progression were evaluated using Cox regression models. RESULTS This study included 99 subjects with iAMD, 21 of which progressed to advanced AMD. Two principal components (PCs) were identified that contributed to the risk of progression to advanced AMD, after adjusting for age and bilateral reticular pseudodrusen. The strongest associated PC was explained largely by the pro-inflammatory cytokine TNFα and the anti-inflammatory IL1ra antagonist of IL1. The additional PC was largely explained by IL6, IL8, C3 and factor D in the positive direction and CRP, MCP1, factor B and factor I in the negative direction. CONCLUSION When evaluated through multivariate analyses, combinations of biomarkers distinguished patients who did and did not progress to future advanced AMD. Increased risk could result from different combinations of analyte levels indicating a complex relationship rather than a simple increase in a few markers. This suggests that studying systemic inflammation in iAMD can provide insights into early pathologic events and potentially identify patients at highest risk for the development of severe AMD.
Collapse
Affiliation(s)
- Brandie D. Wagner
- Department of Biostatistics and Informatics, University of Colorado School of Public Health, Aurora, Colorado, USA
- Department of Ophthalmology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Jennifer L. Patnaik
- Department of Ophthalmology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Alan G. Palestine
- Department of Ophthalmology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | | | - Rebecca Baldermann
- Colorado Clinical and Translational Sciences Institute, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
| | - V. Michael Holers
- Departments of Medicine and Immunology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Marc T. Mathias
- Department of Ophthalmology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Naresh Mandava
- Department of Ophthalmology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Anne M. Lynch
- Department of Ophthalmology, University of Colorado School of Medicine, Aurora, Colorado, USA
| |
Collapse
|
24
|
Complement activation by RPE cells preexposed to TNFα and IFNγ. Exp Eye Res 2022; 218:108982. [DOI: 10.1016/j.exer.2022.108982] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 02/08/2022] [Accepted: 02/14/2022] [Indexed: 01/27/2023]
|
25
|
Ma Y, Ding X, Shao M, Qiu Y, Li S, Cao W, Xu G. Association of Serum Complement C1q and C3 Level with Age-Related Macular Degeneration in Women. J Inflamm Res 2022; 15:285-294. [PMID: 35058703 PMCID: PMC8765539 DOI: 10.2147/jir.s348539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 12/25/2021] [Indexed: 11/23/2022] Open
Abstract
PURPOSE To investigate the association between serum complement components and age-related macular degeneration (AMD). PATIENTS AND METHODS A total of 118 AMD patients and age- and sex-matched 106 control subjects were included. Demographic data and the level of serum complement component (C)1q, C3 and C4 were evaluated. Based on sex, the subjects were stratified into male and female subgroups. RESULTS The level of C1q (226.31±45.33mg/dL) was significantly higher and C3 (121.14±15.76mg/dL) was significantly lower than that in control group (200.03±38.54mg/dL) (128.42±19.81mg/dL) in the female AMD patients (p = 0.005, p = 0.045). Logistic regression showed that increased C1q (OR = 1.132, p = 0.016) and decreased C3 (OR = 0.960, p = 0.048) were independent risk factors for female AMD patients. No statistical significance was observed in the male. CONCLUSION Increased C1q and decreased C3 were associated with increased risk of AMD, suggesting that the complement classical pathway probably be involved in AMD, especially in female.
Collapse
Affiliation(s)
- Yingbo Ma
- Department of Clinical Laboratory, Eye and ENT Hospital of Fudan University, Shanghai, People's Republic of China
| | - Xueqing Ding
- Department of Clinical Laboratory, Eye and ENT Hospital of Fudan University, Shanghai, People's Republic of China
| | - Mingxi Shao
- Department of Clinical Laboratory, Eye and ENT Hospital of Fudan University, Shanghai, People's Republic of China
| | - Yichao Qiu
- Department of Clinical Laboratory, Eye and ENT Hospital of Fudan University, Shanghai, People's Republic of China
| | - Shengjie Li
- Department of Clinical Laboratory, Eye and ENT Hospital of Fudan University, Shanghai, People's Republic of China
| | - Wenjun Cao
- Department of Clinical Laboratory, Eye and ENT Hospital of Fudan University, Shanghai, People's Republic of China
- Department of Ophthalmology, Eye and ENT Hospital of Fudan University, Shanghai, People's Republic of China
| | - Gezhi Xu
- Department of Ophthalmology, Eye and ENT Hospital of Fudan University, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, People's Republic of China
- NHC Key Laboratory of Myopia, Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
26
|
Kim J, Lee YJ, Won JY. Molecular Mechanisms of Retinal Pigment Epithelium Dysfunction in Age-Related Macular Degeneration. Int J Mol Sci 2021; 22:ijms222212298. [PMID: 34830181 PMCID: PMC8624542 DOI: 10.3390/ijms222212298] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/12/2021] [Accepted: 11/12/2021] [Indexed: 02/06/2023] Open
Abstract
The retinal pigment epithelium (RPE), situated upon Bruch’s membrane, plays multiple roles in the ocular system by interacting with photoreceptors and. Therefore, dysfunction of the RPE causes diseases related to vision loss, such as age-related macular degeneration (AMD). Despite AMD being a global cause of blindness, the pathogenesis remains unclear. Understanding the pathogenesis of AMD is the first step for its prevention and treatment. This review summarizes the common pathways of RPE dysfunction and their effect in AMD. Potential treatment strategies for AMD based on targeting the RPE have also been discussed.
Collapse
Affiliation(s)
- Jongmin Kim
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea;
| | - Yeo Jin Lee
- Department of Ophthalmology and Visual Science, Eunpyeong St. Mary’s Hospital, The Catholic University of Korea, Seoul 03312, Korea;
- Catholic Institute for Visual Science, College of Medicine, The Catholic University of Korea, Seoul 14662, Korea
| | - Jae Yon Won
- Department of Ophthalmology and Visual Science, Eunpyeong St. Mary’s Hospital, The Catholic University of Korea, Seoul 03312, Korea;
- Catholic Institute for Visual Science, College of Medicine, The Catholic University of Korea, Seoul 14662, Korea
- Correspondence:
| |
Collapse
|
27
|
Subretinal fibrosis in neovascular age-related macular degeneration: current concepts, therapeutic avenues, and future perspectives. Cell Tissue Res 2021; 387:361-375. [PMID: 34477966 PMCID: PMC8975778 DOI: 10.1007/s00441-021-03514-8] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 07/19/2021] [Indexed: 12/13/2022]
Abstract
Age-related macular degeneration (AMD) is a progressive, degenerative disease of the human retina which in its most aggressive form is associated with the formation of macular neovascularization (MNV) and subretinal fibrosis leading to irreversible blindness. MNVs contain blood vessels as well as infiltrating immune cells, myofibroblasts, and excessive amounts of extracellular matrix proteins such as collagens, fibronectin, and laminin which disrupts retinal function and triggers neurodegeneration. In the mammalian retina, damaged neurons cannot be replaced by tissue regeneration, and subretinal MNV and fibrosis persist and thus fuel degeneration and visual loss. This review provides an overview of subretinal fibrosis in neovascular AMD, by summarizing its clinical manifestations, exploring the current understanding of the underlying cellular and molecular mechanisms and discussing potential therapeutic approaches to inhibit subretinal fibrosis in the future.
Collapse
|
28
|
Ruggiero D, Nutile T, Nappo S, Tirozzi A, Bellenguez C, Leutenegger AL, Ciullo M. Genetics of PlGF plasma levels highlights a role of its receptors and supports the link between angiogenesis and immunity. Sci Rep 2021; 11:16821. [PMID: 34413389 PMCID: PMC8376970 DOI: 10.1038/s41598-021-96256-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 07/28/2021] [Indexed: 11/29/2022] Open
Abstract
Placental growth factor (PlGF) is a member of the vascular endothelial growth factor family and is involved in bone marrow-derived cell activation, endothelial stimulation and pathological angiogenesis. High levels of PlGF have been observed in several pathological conditions especially in cancer, cardiovascular, autoimmune and inflammatory diseases. Little is known about the genetics of circulating PlGF levels. Indeed, although the heritability of circulating PlGF levels is around 40%, no studies have assessed the relation between PlGF plasma levels and genetic variants at a genome-wide level. In the current study, PlGF plasma levels were measured in a population-based sample of 2085 adult individuals from three isolated populations of South Italy. A GWAS was performed in a discovery cohort (N = 1600), followed by a de novo replication (N = 468) from the same populations. The meta-analysis of the discovery and replication samples revealed one signal significantly associated with PlGF circulating levels. This signal was mapped to the PlGF co-receptor coding gene NRP1, indicating its important role in modulating the PlGF plasma levels. Two additional signals, at the PlGF receptor coding gene FLT1 and RAPGEF5 gene, were identified at a suggestive level. Pathway and TWAS analyses highlighted genes known to be involved in angiogenesis and immune response, supporting the link between these processes and PlGF regulation. Overall, these data improve our understanding of the genetic variation underlying circulating PlGF levels. This in turn could lead to new preventive and therapeutic strategies for a wide variety of PlGF-related pathologies.
Collapse
Affiliation(s)
- Daniela Ruggiero
- Institute of Genetics and Biophysics "A. Buzzati-Traverso", National Research Council of Italy (CNR), Via Pietro Castellino, 111, 80131, Naples, Italy.
- IRCCS Neuromed, Pozzilli, Isernia, Italy.
| | - Teresa Nutile
- Institute of Genetics and Biophysics "A. Buzzati-Traverso", National Research Council of Italy (CNR), Via Pietro Castellino, 111, 80131, Naples, Italy
| | | | | | - Celine Bellenguez
- CHU Lille, U1167 - Labex DISTALZ - RID-AGE - Risk Factors and Molecular Determinants of Aging-Related Diseases, Inserm, Institut Pasteur de Lille, Univ. Lille, 59000, Lille, France
| | - Anne-Louise Leutenegger
- UMR 946, Genetic Variation and Human Diseases, Inserm, 75010, Paris, France
- UMR946, Université Paris-Diderot, Sorbonne Paris Cité, 75010, Paris, France
| | - Marina Ciullo
- Institute of Genetics and Biophysics "A. Buzzati-Traverso", National Research Council of Italy (CNR), Via Pietro Castellino, 111, 80131, Naples, Italy.
- IRCCS Neuromed, Pozzilli, Isernia, Italy.
| |
Collapse
|
29
|
Kim BJ, Mastellos DC, Li Y, Dunaief JL, Lambris JD. Targeting complement components C3 and C5 for the retina: Key concepts and lingering questions. Prog Retin Eye Res 2021; 83:100936. [PMID: 33321207 PMCID: PMC8197769 DOI: 10.1016/j.preteyeres.2020.100936] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 12/07/2020] [Accepted: 12/09/2020] [Indexed: 12/13/2022]
Abstract
Age-related macular degeneration (AMD) remains a major cause of legal blindness, and treatment for the geographic atrophy form of AMD is a significant unmet need. Dysregulation of the complement cascade is thought to be instrumental for AMD pathophysiology. In particular, C3 and C5 are pivotal components of the complement cascade and have become leading therapeutic targets for AMD. In this article, we discuss C3 and C5 in detail, including their roles in AMD, biochemical and structural aspects, locations of expression, and the functions of C3 and C5 fragments. Further, the article critically reviews developing therapeutics aimed at C3 and C5, underscoring the potential effects of broad inhibition of complement at the level of C3 versus more specific inhibition at C5. The relationships of complement biology to the inflammasome and microglia/macrophage activity are highlighted. Concepts of C3 and C5 biology will be emphasized, while we point out questions that need to be settled and directions for future investigations.
Collapse
Affiliation(s)
- Benjamin J Kim
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | | | - Yafeng Li
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Joshua L Dunaief
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - John D Lambris
- Department of Laboratory Medicine and Pathology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
30
|
Kato Y, Oguchi Y, Omori T, Shintake H, Tomita R, Kasai A, Ogasawara M, Sugano Y, Itagaki K, Ojima A, Machida T, Sekine H, Sekiryu T. Complement Activation Products and Cytokines in Pachychoroid Neovasculopathy and Neovascular Age-Related Macular Degeneration. Invest Ophthalmol Vis Sci 2021; 61:39. [PMID: 33252634 PMCID: PMC7705396 DOI: 10.1167/iovs.61.13.39] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Purpose To investigate the characteristics of complement activation products and angiogenic cytokines in the aqueous humor in eyes with pachychoroid neovasculopathy (PNV) and neovascular age-related macular degeneration (nAMD). Methods This was a prospective, comparative, observational study. All patients with choroidal neovascularization were classified as PNV without polyps, PNV with polyps (polypoidal choroidal vasculopathy [PCV]), or drusen-associated nAMD according to the presence or absence of pachychoroid features and soft drusen. This study included a total of 105 eyes. Aqueous humor samples were collected from 25 eyes with PNV without polyps, 23 eyes with PCV, and 24 eyes with drusen-associated nAMD before intravitreal anti-vascular endothelial growth factor (VEGF) injection and cataract surgery in 33 control eyes. Clinical samples were measured for complement component 3a (C3a), C4a, C5a, VEGF, and macrophage chemoattractant protein 1 (MCP-1) using a bead-based immunoassay. Results C3a and MCP-1 levels were significantly higher in PCV (P = 0.032 and P = 0.039, respectively) and drusen-associated nAMD (P = 0.01 for both comparisons) than in controls, and no difference was seen in C3a and MCP-1 levels between PNV and controls (P = 0.747 and P = 0.294, respectively). VEGF levels were significantly higher in PNV (P = 0.016), PCV (P = 0.009), and drusen-associated nAMD (P = 0.043) than in controls. In PNV, the VEGF levels elevated without elevated C3a and MCP-1. Conclusions PNV, PCV, and drusen-associated nAMD had significantly distinct profiles of complement activation products and cytokines in the aqueous humor.
Collapse
Affiliation(s)
- Yutaka Kato
- Department of Ophthalmology, Fukushima Medical University, Fukushima, Japan
| | - Yasuharu Oguchi
- Department of Ophthalmology, Fukushima Medical University, Fukushima, Japan
| | - Tomoko Omori
- Department of Immunology, Fukushima Medical University, Fukushima, Japan
| | - Hiroaki Shintake
- Department of Ophthalmology, Fukushima Medical University, Fukushima, Japan
| | - Ryutaro Tomita
- Department of Ophthalmology, Fukushima Medical University, Fukushima, Japan
| | - Akihito Kasai
- Department of Ophthalmology, Fukushima Medical University, Fukushima, Japan
| | - Masashi Ogasawara
- Department of Ophthalmology, Fukushima Medical University, Fukushima, Japan
| | - Yukinori Sugano
- Department of Ophthalmology, Fukushima Medical University, Fukushima, Japan
| | - Kanako Itagaki
- Department of Ophthalmology, Fukushima Medical University, Fukushima, Japan
| | - Akira Ojima
- Department of Ophthalmology, Fukushima Medical University, Fukushima, Japan
| | - Takeshi Machida
- Department of Immunology, Fukushima Medical University, Fukushima, Japan
| | - Hideharu Sekine
- Department of Immunology, Fukushima Medical University, Fukushima, Japan
| | - Tetsuju Sekiryu
- Department of Ophthalmology, Fukushima Medical University, Fukushima, Japan
| |
Collapse
|
31
|
Dreismann AK, McClements ME, Barnard AR, Orhan E, Hughes JP, Lachmann PJ, MacLaren RE. Functional expression of complement factor I following AAV-mediated gene delivery in the retina of mice and human cells. Gene Ther 2021; 28:265-276. [PMID: 33750925 PMCID: PMC8149295 DOI: 10.1038/s41434-021-00239-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 01/05/2021] [Accepted: 02/03/2021] [Indexed: 01/01/2023]
Abstract
Dry age-related macular degeneration (AMD) is characterised by loss of central vision and currently has no approved medical treatment. Dysregulation of the complement system is thought to play an important role in disease pathology and supplementation of Complement Factor I (CFI), a key regulator of the complement system, has the potential to provide a treatment option for AMD. In this study, we demonstrate the generation of AAV constructs carrying the human CFI sequence and expression of CFI in cell lines and in the retina of C57BL/6 J mice. Four codon optimised constructs were compared to the most common human CFI sequence. All constructs expressed CFI protein; however, most codon optimised sequences resulted in significantly reduced CFI secretion compared to the non-optimised CFI sequence. In vivo expression analysis showed that CFI was predominantly expressed in the RPE and photoreceptors. Secreted protein in vitreous humour was demonstrated to be functionally active. The findings presented here have led to the formulation of an AAV-vectored gene therapy product currently being tested in a first-in-human clinical trial in subjects with geographic atrophy secondary to dry AMD (NCT03846193).
Collapse
Affiliation(s)
- Anna K Dreismann
- Gyroscope Therapeutics Limited, Stevenage Bio-Science Catalyst, Stevenage, UK
| | | | - Alun R Barnard
- Nuffield Laboratory of Ophthalmology, University of Oxford, Oxford, UK
| | - Elise Orhan
- Gyroscope Therapeutics Limited, Stevenage Bio-Science Catalyst, Stevenage, UK
| | - Jane P Hughes
- Gyroscope Therapeutics Limited, Stevenage Bio-Science Catalyst, Stevenage, UK
| | - Peter J Lachmann
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Robert E MacLaren
- Nuffield Laboratory of Ophthalmology, University of Oxford, Oxford, UK.
- Oxford Eye Hospital NIHR Biomedical Research Centre, Oxford, UK.
| |
Collapse
|
32
|
Silawal S, Kohl B, Shi J, Schulze-Tanzil G. Complement Regulation in Human Tenocytes under the Influence of Anaphylatoxin C5a. Int J Mol Sci 2021; 22:ijms22063105. [PMID: 33803624 PMCID: PMC8003014 DOI: 10.3390/ijms22063105] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/13/2021] [Accepted: 03/15/2021] [Indexed: 12/27/2022] Open
Abstract
A central part of the complement system, the anaphylatoxin C5a was investigated in this study to learn its effects on tenocytes in respect to understanding the potential expression of other crucial complement factors and pro-inflammatory mediators involved in tendinopathy. Human hamstring tendon-derived tenocytes were treated with recombinant C5a protein in concentrations of 25 ng/mL and 100 ng/mL for 0.5 h (early phase), 4 h (intermediate phase), and 24 h (late phase). Tenocytes survival was assessed after 24 h stimulation by live-dead assay. The gene expression of complement-related factors C5aR, the complement regulatory proteins (CRPs) CD46, CD55, CD59, and of the pro-inflammatory cytokines tumor necrosis factor (TNF)-α and interleukin (IL)-6 was monitored using qPCR. Tenocytes were immunolabeled for C5aR and CD55 proteins. TNFα production was monitored by ELISA. Tenocyte survival was not impaired through C5a stimulation. Interestingly, the gene expression of C5aR and that of the CRPs CD46 and CD59 was significantly reduced in the intermediate and late phase, and that of TNFα only in an early phase, compared to the control group. ELISA analysis indicated a concomitant not significant trend of impaired TNFα protein synthesis at 4 h. However, there was also an early significant induction of CD55 and CD59 mediated by 25 ng/mL anaphylatoxin C5a. Hence, exposure of tenocytes to C5a obviously evokes a time and concentration-dependent response in their expression of complement and pro-inflammatory factors. C5a, released in damaged tendons, might directly contribute to tenocyte activation and thereby be involved in tendon healing and tendinopathy.
Collapse
Affiliation(s)
- Sandeep Silawal
- Institute of Anatomy and Cell Biology, Paracelsus Medical University, Nuremberg and Salzburg, General Hospital Nuremberg, Prof. Ernst Nathan Str. 1, 90419 Nuremberg, Germany; (S.S.); (J.S.)
| | - Benjamin Kohl
- Department of Traumatology and Reconstructive Surgery, Campus Benjamin Franklin, Charité—Universitätsmedizin Berlin, Corporate Member of Berlin Institute of Health, Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany;
| | - Jingjian Shi
- Institute of Anatomy and Cell Biology, Paracelsus Medical University, Nuremberg and Salzburg, General Hospital Nuremberg, Prof. Ernst Nathan Str. 1, 90419 Nuremberg, Germany; (S.S.); (J.S.)
| | - Gundula Schulze-Tanzil
- Institute of Anatomy and Cell Biology, Paracelsus Medical University, Nuremberg and Salzburg, General Hospital Nuremberg, Prof. Ernst Nathan Str. 1, 90419 Nuremberg, Germany; (S.S.); (J.S.)
- Correspondence: ; Tel.: +49-911-398-11-6772
| |
Collapse
|
33
|
van Dijk EHC, Boon CJF. Serous business: Delineating the broad spectrum of diseases with subretinal fluid in the macula. Prog Retin Eye Res 2021; 84:100955. [PMID: 33716160 DOI: 10.1016/j.preteyeres.2021.100955] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 02/14/2021] [Accepted: 02/19/2021] [Indexed: 02/08/2023]
Abstract
A wide range of ocular diseases can present with serous subretinal fluid in the macula and therefore clinically mimic central serous chorioretinopathy (CSC). In this manuscript, we categorise the diseases and conditions that are part of the differential diagnosis into 12 main pathogenic subgroups: neovascular diseases, vitelliform lesions, inflammatory diseases, ocular tumours, haematological malignancies, paraneoplastic syndromes, genetic diseases, ocular developmental anomalies, medication-related conditions and toxicity-related diseases, rhegmatogenous retinal detachment and tractional retinal detachment, retinal vascular diseases, and miscellaneous diseases. In addition, we describe 2 new clinical pictures associated with macular subretinal fluid accumulation, namely serous maculopathy with absence of retinal pigment epithelium (SMARPE) and serous maculopathy due to aspecific choroidopathy (SMACH). Differentiating between these various diseases and CSC can be challenging, and obtaining the correct diagnosis can have immediate therapeutic and prognostic consequences. Here, we describe the key differential diagnostic features of each disease within this clinical spectrum, including representative case examples. Moreover, we discuss the pathogenesis of each disease in order to facilitate the differentiation from typical CSC.
Collapse
Affiliation(s)
- Elon H C van Dijk
- Department of Ophthalmology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, the Netherlands
| | - Camiel J F Boon
- Department of Ophthalmology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, the Netherlands; Department of Ophthalmology, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands.
| |
Collapse
|
34
|
Han X, de la Fuente M, Nieman MT. Complement factor C4a does not activate protease-activated receptor 1 (PAR1) or PAR4 on human platelets. Res Pract Thromb Haemost 2021; 5:104-110. [PMID: 33537534 PMCID: PMC7845074 DOI: 10.1002/rth2.12459] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 10/22/2020] [Accepted: 11/04/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Protease-activated receptor (PAR) 1 and PAR4 are key thrombin signal mediators for human platelet activation and aggregation in response to vascular injury. They are primarily activated by thrombin cleavage of the N-terminus to expose a tethered ligand. In addition to the canonical activation by thrombin, a growing panel of proteases can also elicit PAR1- or PAR4-mediated signal transduction. Recently, complement factor C4a was reported as the first endogenous agonist for both PAR1 and PAR4. Further, it is the first endogenous nontethered ligand that activates PAR1 and PAR4. These studies were conducted with human microvascular cells; the impact of C4a on platelet PARs is unknown. OBJECTIVES The goal of this study was to interrogate PAR1 and PAR4 activation by C4a on human platelets. METHODS Platelet-rich plasma (PRP) was isolated from healthy donors. PRP was stimulated with C4a, and the platelet aggregation was measured. Human embryonic kidney (HEK) 293 Flp-In T-rex cells were used to further test if C4a stimulation can initiate PAR1- or PAR4-mediated Gαq signaling, which was measured by intracellular calcium mobilization. RESULTS C4a failed to elicit platelet aggregation via PAR1- or PAR4-mediated manner. In addition, no PAR1- or PAR4-mediated calcium mobilization was observed upon C4a stimulation on HEK293 cells. CONCLUSIONS Complement factor C4a does not activate PAR1 or PAR4 on human platelets. These data show that PAR1 and PAR4 activation by C4a on microvascular cells likely requires a cofactor, which reinforces the concept of cell type-specific regulation of protease signaling.
Collapse
Affiliation(s)
- Xu Han
- Department of PharmacologyCase Western Reserve UniversityClevelandOHUSA
| | | | - Marvin T. Nieman
- Department of PharmacologyCase Western Reserve UniversityClevelandOHUSA
| |
Collapse
|
35
|
Acadesine suppresses TNF-α induced complement component 3 (C3), in retinal pigment epithelial (RPE) cells. PLoS One 2020; 15:e0244307. [PMID: 33362238 PMCID: PMC7757886 DOI: 10.1371/journal.pone.0244307] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 12/07/2020] [Indexed: 01/02/2023] Open
Abstract
Rationale Age-related macular degeneration (AMD) is the most prevalent form of irreversible blindness in the developed world. Aging, inflammation and complement dysregulation affecting the retinal pigment epithelium (RPE), are considered significant contributors in its pathogenesis and several evidences have linked tumor necrosis factor alpha (TNF-α) and complement component 3 (C3) with AMD. Acadesine, an analog of AMP and an AMP-activated protein kinase (AMPK) activator, has been shown to have cytoprotective effects in human clinical trials as well as having anti-inflammatory and anti-vascular exudative effects in animals. The purpose of this study was to evaluate if acadesine is able to suppress TNF-α induced C3 in RPE cells. Methods ARPE-19 and human primary RPE cells were cultured and allowed to grow to confluence. TNF-α was used for C3 induction in the presence or absence of acadesine. Small molecule inhibitors and siRNA were used to determine if acadesine exerts its effect via the extracellular or intracellular pathway and to evaluate the importance of AMPK for these effects. The expression level of C3 was determined by immunoblot analysis. Results Acadesine suppresses TNF-α induced C3 in a dose dependent manner. When we utilized the adenosine receptor inhibitor dipyridamole (DPY) along with acadesine, acadesine’s effects were abolished, indicating the necessity of acadesine to enter the cell in order to exert it’s action. However, pretreatment with 5-iodotubericidin (5-Iodo), an adenosine kinase (AK) inhibitor, didn’t prevent acadesine from decreasing TNF-α induced C3 expression suggesting that acadesine does not exert its effect through AMP conversion and subsequent activation of AMPK. Consistent with this, knockdown of AMPK α catalytic subunit did not affect the inhibitory effect of acadesine on TNF-α upregulation of C3. Conclusions Our results suggest that acadesine suppresses TNF-α induced C3, likely through an AMPK-independent pathway, and could have potential use in complement over activation diseases.
Collapse
|
36
|
Little K, Llorián-Salvador M, Tang M, Du X, Marry S, Chen M, Xu H. Macrophage to myofibroblast transition contributes to subretinal fibrosis secondary to neovascular age-related macular degeneration. J Neuroinflammation 2020; 17:355. [PMID: 33239022 PMCID: PMC7690191 DOI: 10.1186/s12974-020-02033-7] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 11/11/2020] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Macular fibrosis causes irreparable vision loss in neovascular age-related macular degeneration (nAMD) even with anti-vascular endothelial growth factor (VEGF) therapy. Inflammation is known to play an important role in macular fibrosis although the underlying mechanism remains poorly defined. The aim of this study was to understand how infiltrating macrophages and complement proteins may contribute to macular fibrosis. METHODS Subretinal fibrosis was induced in C57BL/6J mice using the two-stage laser protocol developed by our group. The eyes were collected at 10, 20, 30 and 40 days after the second laser and processed for immunohistochemistry for infiltrating macrophages (F4/80 and Iba-1), complement components (C3a and C3aR) and fibrovascular lesions (collagen-1, Isolectin B4 and α-SMA). Human retinal sections with macular fibrosis were also used in the study. Bone marrow-derived macrophages (BMDMs) from C57BL/6J mice were treated with recombinant C3a, C5a or TGF-β for 48 and 96 h. qPCR, Western blot and immunohistochemistry were used to examine the expression of myofibroblast markers. The involvement of C3a-C3aR pathway in macrophage to myofibroblast transition (MMT) and subretinal fibrosis was further investigated using a C3aR antagonist (C3aRA) and a C3a blocking antibody in vitro and in vivo. RESULTS Approximately 20~30% of F4/80+ (or Iba-1+) infiltrating macrophages co-expressed α-SMA in subretinal fibrotic lesions both in human nAMD eyes and in the mouse model. TGF-β and C3a, but not C5a treatment, significantly upregulated expression of α-SMA, fibronectin and collagen-1 in BMDMs. C3a-induced upregulation of α-SMA, fibronectin and collagen-1 in BMDMs was prevented by C3aRA treatment. In the two-stage laser model of induced subretinal fibrosis, treatment with C3a blocking antibody but not C3aRA significantly reduced vascular leakage and Isolectin B4+ lesions. The treatment did not significantly alter collagen-1+ fibrotic lesions. CONCLUSIONS MMT plays a role in macular fibrosis secondary to nAMD. MMT can be induced by TGF-β and C3a but not C5a. Further research is required to fully understand the role of MMT in macular fibrosis. Macrophage to myofibroblast transition (MMT) contributes to subretinal fibrosis. Subretinal fibrosis lesions contain various cell types, including macrophages and myofibroblasts, and are fibrovascular. Myofibroblasts are key cells driving pathogenic fibrosis, and they do so by producing excessive amount of extracellular matrix proteins. We have found that infiltrating macrophages can transdifferentiate into myofibroblasts, a phenomenon termed macrophage to myofibroblast transition (MMT) in macular fibrosis. In addition to TGF-β1, C3a generated during complement activation in CNV can also induce MMT contributing to macular fibrosis. RPE = retinal pigment epithelium. BM = Bruch's membrane. MMT = macrophage to myofibroblast transition. TGFB = transforming growth factor β. a-SMA = alpha smooth muscle actin. C3a = complement C3a.
Collapse
Affiliation(s)
- Karis Little
- The Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Sciences, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK
| | - Maria Llorián-Salvador
- The Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Sciences, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK
| | - Miao Tang
- The Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Sciences, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK
| | - Xuan Du
- The Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Sciences, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK
| | - Stephen Marry
- The Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Sciences, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK
| | - Mei Chen
- The Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Sciences, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK
| | - Heping Xu
- The Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Sciences, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK.
| |
Collapse
|
37
|
Propson NE, Gedam M, Zheng H. Complement in Neurologic Disease. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2020; 16:277-298. [PMID: 33234021 DOI: 10.1146/annurev-pathol-031620-113409] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Classic innate immune signaling pathways provide most of the immune response in the brain. This response activates many of the canonical signaling mechanisms identified in peripheral immune cells, despite their relative absence in this immune-privileged tissue. Studies over the past decade have strongly linked complement protein production and activation to age-related functional changes and neurodegeneration. The reactivation of the complement signaling pathway in aging and disease has opened new avenues for understanding brain aging and neurological disease pathogenesis and has implicated cell types such as astrocytes, microglia, endothelial cells, oligodendrocytes, neurons, and even peripheral immune cells in these processes. In this review, we aim to unravel the past decade of research related to complement activation and its numerous consequences in aging and neurological disease.
Collapse
Affiliation(s)
- Nicholas E Propson
- Huffington Center on Aging, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Manasee Gedam
- Huffington Center on Aging, Baylor College of Medicine, Houston, Texas 77030, USA.,Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Hui Zheng
- Huffington Center on Aging, Baylor College of Medicine, Houston, Texas 77030, USA.,Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas 77030, USA.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA;
| |
Collapse
|
38
|
Chen M, Yang N, Lechner J, Toth L, Hogg R, Silvestri G, Chakravarthy U, Xu H. Plasma level of lipocalin 2 is increased in neovascular age-related macular degeneration patients, particularly those with macular fibrosis. IMMUNITY & AGEING 2020; 17:35. [PMID: 33292361 PMCID: PMC7666483 DOI: 10.1186/s12979-020-00205-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 11/01/2020] [Indexed: 02/07/2023]
Abstract
Background Previously, we and others have reported higher populations of circulating neutrophils in patients with neovascular age-related macular degeneration (nAMD). Neutrophil gelatinase-associated lipocalin (NGAL, also known as lipocalin-2, LCN2), an important innate immune mediator, is known to be critically involved in sterile inflammation-mediated organ failure, fibrosis, cancer progression and retinal degeneration. This study investigated the plasma levels of LCN2, matrix metalloproteinase 9 (MMP9) and LCN2/MMP9 complex in different types of nAMD and examined whether the levels were related to patients’ responsiveness to anti-VEGF therapy. Results One hundred and seventy-four nAMD patients, including 108 with choroidal neovascularisation (CNV), 32 with retinal angiomatous proliferation (RAP), 23 with polypoidal choroidal vasculopathy (PCV) and 11 unclassified patients, and 43 healthy controls were recruited to this case-control study. Fifty-eight nAMD patients had macular fibrosis and 110 patients did not. Out of the 174 nAMD patients, 80 patients responded completely, 90 responded partially, and 4 did not respond to the anti-VEGF therapy. The plasma levels of LCN2 in nAMD patients (181.46 ± 73.62 ng/ml) was significantly higher than that in healthy controls (152.24 ± 49.55 ng/ml, P = 0.047). However, the difference disappeared after adjusting for age. A positive correlation between plasma level of LCN2 and age was observed in nAMD patients (r = 0.29, P = 0.0002) but not in healthy controls. The plasma level of LCN2 was also positively correlated with circulating neutrophils in nAMD patients (r = 0.34, p = 0.0007) but not in healthy controls (r = 0.057, p = 0.77). No correlation was observed between age and circulating neutrophils. Further analysis of nAMD subtypes uncovered a significantly higher level of LCN2 in patients with macular fibrosis even after adjusting for age. No relationship was observed between plasma levels of LCN2 and patients’ responsiveness to anti-VEGF therapy. The plasma levels of MMP9 and LCN2/MMP9 complex were comparable between nAMD and controls. Conclusions Our results suggest that higher plasma levels of LCN2 in nAMD are related to ageing and increased population of circulating neutrophils. Our results also suggest that higher levels of LCN2 may increase the risk of macular fibrosis in nAMD.
Collapse
Affiliation(s)
- Mei Chen
- Centre for Experimental Medicine, The Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK
| | - Nan Yang
- Centre for Experimental Medicine, The Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK
| | - Judith Lechner
- Centre for Experimental Medicine, The Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK
| | - Levente Toth
- Centre for Experimental Medicine, The Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK
| | - Ruth Hogg
- Centre for Public Health, School of Medicine, Dentistry & Biomedical Science, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK
| | | | - Usha Chakravarthy
- Centre for Public Health, School of Medicine, Dentistry & Biomedical Science, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK
| | - Heping Xu
- Centre for Experimental Medicine, The Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK.
| |
Collapse
|
39
|
Tzoumas N, Hallam D, Harris CL, Lako M, Kavanagh D, Steel DHW. Revisiting the role of factor H in age-related macular degeneration: Insights from complement-mediated renal disease and rare genetic variants. Surv Ophthalmol 2020; 66:378-401. [PMID: 33157112 DOI: 10.1016/j.survophthal.2020.10.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 10/27/2020] [Accepted: 10/29/2020] [Indexed: 12/14/2022]
Abstract
Ophthalmologists are long familiar with the eye showing signs of systemic disease, but the association between age-related macular degeneration and abnormal complement activation, common to several renal disorders, has only recently been elucidated. Although complement activation products were identified in drusen almost three decades ago, it was not until the early 21st century that a single-nucleotide polymorphism in the complement factor H gene was identified as a major heritable determinant of age-related macular degeneration, galvanizing global efforts to unravel the pathogenesis of this common disease. Advances in proteomic analyses and familial aggregation studies have revealed distinctive clinical phenotypes segregated by the functional effects of common and rare genetic variants on the mature protein and its splice variant, factor H-like protein 1. The predominance of loss-of-function, N-terminal mutations implicate age-related macular degeneration as a disease of general complement dysregulation, offering several therapeutic avenues for its modulation. Here, we explore the molecular impact of these mutations/polymorphisms on the ability of variant factor H/factor H-like protein 1 to localize to polyanions, pentraxins, proinflammatory triggers, and cell surfaces across ocular and renal tissues and exert its multimodal regulatory functions and their clinical implications. Finally, we critically evaluate key therapeutic and diagnostic efforts in this rapidly evolving field.
Collapse
Affiliation(s)
- Nikolaos Tzoumas
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom.
| | - Dean Hallam
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Claire L Harris
- Complement Therapeutics Research Group, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom; National Renal Complement Therapeutics Centre, Royal Victoria Infirmary, Newcastle upon Tyne, United Kingdom
| | - Majlinda Lako
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - David Kavanagh
- Complement Therapeutics Research Group, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom; National Renal Complement Therapeutics Centre, Royal Victoria Infirmary, Newcastle upon Tyne, United Kingdom
| | - David H W Steel
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom; Sunderland Eye Infirmary, Sunderland, United Kingdom
| |
Collapse
|
40
|
Lynch AM, Wagner BD, Palestine AG, Janjic N, Patnaik JL, Mathias MT, Siringo FS, Mandava N. Plasma Biomarkers of Reticular Pseudodrusen and the Risk of Progression to Advanced Age-Related Macular Degeneration. Transl Vis Sci Technol 2020; 9:12. [PMID: 32974084 PMCID: PMC7488626 DOI: 10.1167/tvst.9.10.12] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 08/03/2020] [Indexed: 12/16/2022] Open
Abstract
Purpose To determine, using an aptamer-based technology in patients with intermediate age-related macular degeneration (AMD), (1) if there is a difference in plasma levels of 4979 proteins in patients with and without reticular pseudodrusen (RPD), and (2) if plasma levels of proteins are related to time to conversion to advanced AMD. Methods Patients with intermediate AMD and RPD were identified from an AMD registry. Relative concentrations of each protein were log (base 2) transformed and compared between patients with and without RPD using linear regression. A Cox proportional hazards survival model was fit to each aptamer to quantify associations with time to conversion. A pathway analysis was conducted in converters versus non-converters using the Reactome database. Results Of the 109 intermediate AMD patients, 39 had bilateral RPD (36%). Two proteins, TCL1A and CNDP1, were lower in patients in the intermediate AMD group with RPD. Twenty-one patients converted to advanced AMD with a median time to conversion of 25.2 months (range, 2.3-48.5 months) and median follow-up time in non-converters of 26.4 months (range, 0.03-49.7 months). Several proteins (lysozyme C, TFF3, RNAS6, and SAP3) distinguished patients who converted from those who did not convert to advanced AMD. The top conversion pathways included tumor necrosis factors bind their physiological receptors, digestion and absorption, signaling by activin, and signaling by TGF-β family members. Conclusions We identified a protein signature related to RPD, as well as to conversion to advanced AMD. The pathway analysis suggests that dysfunction of critical systemic pathways may have links to conversion to advanced AMD. Translational Relevance Biomarkers identified in plasma likely reflect systemic alterations in protein expression in patients with intermediate AMD.
Collapse
Affiliation(s)
- Anne M Lynch
- Department of Ophthalmology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Brandie D Wagner
- Department of Biostatistics and Informatics, University of Colorado School of Public Health, Aurora, CO, USA
| | - Alan G Palestine
- Department of Ophthalmology, University of Colorado School of Medicine, Aurora, CO, USA
| | | | - Jennifer L Patnaik
- Department of Ophthalmology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Marc T Mathias
- Department of Ophthalmology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Frank S Siringo
- Department of Ophthalmology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Naresh Mandava
- Department of Ophthalmology, University of Colorado School of Medicine, Aurora, CO, USA
| |
Collapse
|
41
|
Somasundaran S, Constable IJ, Mellough CB, Carvalho LS. Retinal pigment epithelium and age-related macular degeneration: A review of major disease mechanisms. Clin Exp Ophthalmol 2020; 48:1043-1056. [PMID: 32710488 PMCID: PMC7754492 DOI: 10.1111/ceo.13834] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 07/05/2020] [Accepted: 07/10/2020] [Indexed: 01/18/2023]
Abstract
Age‐related macular degeneration (AMD) is a progressive degenerative disease that is the leading cause of vision loss in the elderly population. Degeneration/dysregulation of the retinal pigment epithelium (RPE), a supportive monolayer of cells underlying the photoreceptors, is commonly seen in patients with AMD. While treatment exists for the neovascular/wet form of AMD, there is currently no cure for the non‐exudative/dry form of AMD, making it imperative to understand the pathogenesis of this disease. Although our understanding of the aetiology of AMD has increased over the years, the underlying disease mechanism has not yet been identified, mainly due to the multifactorial nature of this disease. Herein, we review some of the commonly proposed degeneration pathways of RPE cells and their role in the pathogenesis of AMD; including activation of the complement cascade, oxidative stress‐induced cell death mechanisms, dysfunctional mitochondria and the role of crystallins in AMD disease progression.
Collapse
Affiliation(s)
- Shreya Somasundaran
- Centre for Ophthalmology and Visual Science/Lions Eye Institute, University of Western Australia, Nedlands, Western Australia, Australia
| | - Ian J Constable
- Centre for Ophthalmology and Visual Science/Lions Eye Institute, University of Western Australia, Nedlands, Western Australia, Australia
| | - Carla B Mellough
- Centre for Ophthalmology and Visual Science/Lions Eye Institute, University of Western Australia, Nedlands, Western Australia, Australia
| | - Livia S Carvalho
- Centre for Ophthalmology and Visual Science/Lions Eye Institute, University of Western Australia, Nedlands, Western Australia, Australia
| |
Collapse
|
42
|
Shivshankar P, Fekry B, Eckel-Mahan K, Wetsel RA. Circadian Clock and Complement Immune System-Complementary Control of Physiology and Pathology? Front Cell Infect Microbiol 2020; 10:418. [PMID: 32923410 PMCID: PMC7456827 DOI: 10.3389/fcimb.2020.00418] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 07/08/2020] [Indexed: 12/11/2022] Open
Abstract
Mammalian species contain an internal circadian (i.e., 24-h) clock that is synchronized to the day and night cycles. Large epidemiological studies, which are supported by carefully controlled studies in numerous species, support the idea that chronic disruption of our circadian cycles results in a number of health issues, including obesity and diabetes, defective immune response, and cancer. Here we focus specifically on the role of the complement immune system and its relationship to the internal circadian clock system. While still an incompletely understood area, there is evidence that dysregulated proinflammatory cytokines, complement factors, and oxidative stress can be induced by circadian disruption and that these may feed back into the oscillator at the level of circadian gene regulation. Such a feedback cycle may contribute to impaired host immune response against pathogenic insults. The complement immune system including its activated anaphylatoxins, C3a and C5a, not only facilitate innate and adaptive immune response in chemotaxis and phagocytosis, but they can also amplify chronic inflammation in the host organism. Consequent development of autoimmune disorders, and metabolic diseases associated with additional environmental insults that activate complement can in severe cases, lead to accelerated tissue dysfunction, fibrosis, and ultimately organ failure. Because several promising complement-targeted therapeutics to block uncontrolled complement activation and treat autoimmune diseases are in various phases of clinical trials, understanding fully the circadian properties of the complement system, and the reciprocal regulation by these two systems could greatly improve patient treatment in the long term.
Collapse
Affiliation(s)
- Pooja Shivshankar
- Research Center for Immunology and Autoimmune Diseases, Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Baharan Fekry
- Center for Metabolic and Degenerative Diseases, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Kristin Eckel-Mahan
- Center for Metabolic and Degenerative Diseases, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Rick A Wetsel
- Research Center for Immunology and Autoimmune Diseases, Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
43
|
Johnson AA, Shokhirev MN, Wyss-Coray T, Lehallier B. Systematic review and analysis of human proteomics aging studies unveils a novel proteomic aging clock and identifies key processes that change with age. Ageing Res Rev 2020; 60:101070. [PMID: 32311500 DOI: 10.1016/j.arr.2020.101070] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 03/23/2020] [Accepted: 04/07/2020] [Indexed: 12/14/2022]
Abstract
The development of clinical interventions that significantly improve human healthspan requires robust markers of biological age as well as thoughtful therapeutic targets. To promote these goals, we performed a systematic review and analysis of human aging and proteomics studies. The systematic review includes 36 different proteomics analyses, each of which identified proteins that significantly changed with age. We discovered 1,128 proteins that had been reported by at least two or more analyses and 32 proteins that had been reported by five or more analyses. Each of these 32 proteins has known connections relevant to aging and age-related disease. GDF15, for example, extends both lifespan and healthspan when overexpressed in mice and is additionally required for the anti-diabetic drug metformin to exert beneficial effects on body weight and energy balance. Bioinformatic enrichment analyses of our 1,128 commonly identified proteins heavily implicated processes relevant to inflammation, the extracellular matrix, and gene regulation. We additionally propose a novel proteomic aging clock comprised of proteins that were reported to change with age in plasma in three or more different studies. Using a large patient cohort comprised of 3,301 subjects (aged 18-76 years), we demonstrate that this clock is able to accurately predict human age.
Collapse
|
44
|
Paraoan L, Sharif U, Carlsson E, Supharattanasitthi W, Mahmud NM, Kamalden TA, Hiscott P, Jackson M, Grierson I. Secretory proteostasis of the retinal pigmented epithelium: Impairment links to age-related macular degeneration. Prog Retin Eye Res 2020; 79:100859. [PMID: 32278708 DOI: 10.1016/j.preteyeres.2020.100859] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 03/24/2020] [Accepted: 03/26/2020] [Indexed: 12/19/2022]
Abstract
Secretory proteostasis integrates protein synthesis, processing, folding and trafficking pathways that are essential for efficient cellular secretion. For the retinal pigment epithelium (RPE), secretory proteostasis is of vital importance for the maintenance of the structural and functional integrity of apical (photoreceptors) and basal (Bruch's membrane/choroidal blood supply) sides of the environment it resides in. This integrity is achieved through functions governed by RPE secreted proteins, which include extracellular matrix modelling/remodelling, angiogenesis and immune response modulation. Impaired RPE secretory proteostasis affects not only the extracellular environment, but leads to intracellular protein aggregation and ER-stress with subsequent cell death. Ample recent evidence implicates dysregulated proteostasis as a key factor in the development of age-related macular degeneration (AMD), the leading cause of blindness in the developed world, and research aiming to characterise the roles of various proteins implicated in AMD-associated dysregulated proteostasis unveiled unexpected facets of the mechanisms involved in degenerative pathogenesis. This review analyses cellular processes unveiled by the study of the top 200 transcripts most abundantly expressed by the RPE/choroid in the light of the specialised secretory nature of the RPE. Functional roles of these proteins and the mechanisms of their impaired secretion, due to age and genetic-related causes, are analysed in relation to AMD development. Understanding the importance of RPE secretory proteostasis in relation to maintaining retinal health and how it becomes impaired in disease is of paramount importance for the development and assessment of future therapeutic advancements involving gene and cell therapies.
Collapse
Affiliation(s)
- Luminita Paraoan
- Department of Eye and Vision Science, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom.
| | - Umar Sharif
- Department of Eye and Vision Science, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| | - Emil Carlsson
- Department of Eye and Vision Science, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| | - Wasu Supharattanasitthi
- Department of Eye and Vision Science, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom; Department of Physiology, Faculty of Pharmacy, Mahidol University, Bangkok, Thailand
| | - Nur Musfirah Mahmud
- Department of Eye and Vision Science, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| | - Tengku Ain Kamalden
- Eye Research Centre, Department of Ophthalmology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Paul Hiscott
- Department of Eye and Vision Science, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| | - Malcolm Jackson
- Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| | - Ian Grierson
- Department of Eye and Vision Science, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
45
|
Lynch AM, Palestine AG, Wagner BD, Patnaik JL, Frazier-Abel AA, Mathias MT, Siringo FS, Holers VM, Mandava N. Complement factors and reticular pseudodrusen in intermediate age-related macular degeneration staged by multimodal imaging. BMJ Open Ophthalmol 2020; 5:e000361. [PMID: 32509962 PMCID: PMC7254108 DOI: 10.1136/bmjophth-2019-000361] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 11/11/2019] [Accepted: 12/17/2019] [Indexed: 12/31/2022] Open
Abstract
Objective Systemic activation of the complement system in intermediate age-related macular degeneration (AMD) is understudied. Moreover, links between the presence of reticular pseudodrusen (RPD) and systemic complement dysregulation have not been studied. The aim of this study was to determine if there is a difference in plasma complement factor levels in intermediate AMD compared with controls, and if complement levels are related to the presence of RPD. Methods and analysis Levels of complement factors C1q (µg/mL), C4 (µg/mL), C2 (µg/mL), Mannose Binding Lectin (ng/mL), C4b (µg/mL), C3 (µg/mL), factor B (µg/mL), factor D (µg/mL), properdin (µg/mL), C3a (ng/mL), iC3b/C3b (ng/mL), Ba (ng/mL), factor H (µg/mL), factor I (µg/mL), C5 (µg/mL), C5a (pg/mL) and SC5b-9 (ng/mL) were measured in plasma. Results 109 cases and 65 controls were included in the study. Thirty-nine (36%) cases had RPD. Significantly lower systemic levels of: C1q (OR 0.96, 95% CI 0.94 to 0.98), factor B (OR 0.98, 95% CI 0.96 to 0.99), iC3b/C3b (OR 0.97, 95% CI 0.95 to 0.98), factor H (OR 0.99, 95% CI 0.98 to 0.99), factor I (OR 0.83, 95% CI 0.77 to 0.89) and C5 (OR 0.94, 95% CI 0.90 to 0.98) were found in cases versus controls. Significantly elevated levels of: C2 (OR 1.29, 95% CI 1.07 to 1.59), C3a (OR 1.03, 95% CI 1.01 to 1.05) Ba (OR 1.03, 95% CI 1.01 to 1.05) and C5a (OR 1.04, 95% CI 1.02 to 1.07) were found in cases versus controls. Systemic levels of complement factors measured were not related to the presence of RPD. Conclusions Levels of several systemic complement pathway factors were found to be altered in intermediate AMD. Systemic levels of complement factors were not related to RPD.
Collapse
Affiliation(s)
- Anne M Lynch
- Department of Ophthalmology, University of Colorado Denver School of Medicine, Aurora, Colorado, USA
| | - Alan G Palestine
- Department of Ophthalmology, University of Colorado Denver School of Medicine, Aurora, Colorado, USA
| | - Brandie D Wagner
- Department of Biostatistics and Informatics, University of Colorado School of Public Health, Aurora, Colorado, USA
| | - Jennifer L Patnaik
- Department of Ophthalmology, University of Colorado Denver School of Medicine, Aurora, Colorado, USA
| | | | - Marc T Mathias
- Department of Ophthalmology, University of Colorado Denver School of Medicine, Aurora, Colorado, USA
| | - Frank S Siringo
- Department of Ophthalmology, University of Colorado Denver School of Medicine, Aurora, Colorado, USA
| | - Vernon Michael Holers
- Departments of Medicine and Immunology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Naresh Mandava
- Department of Ophthalmology, University of Colorado Denver School of Medicine, Aurora, Colorado, USA
| |
Collapse
|
46
|
Subhi Y, Krogh Nielsen M, Molbech CR, Oishi A, Singh A, Nissen MH, Sørensen TL. Polypoidal Choroidal Vasculopathy Associate With Diminished Regulatory T Cells That Are Polarized Into a T Helper 2-Like Phenotype. Invest Ophthalmol Vis Sci 2019; 60:2583-2590. [PMID: 31219532 DOI: 10.1167/iovs.19-26882] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose To investigate possible roles of T helper (Th) cells, regulatory T cells (Tregs), and the recently mapped Th-like Tregs in patients with polypoidal choroidal vasculopathy (PCV). Methods In this prospective case-control study, we obtained fresh venous blood from patients with PCV (n = 24), age-matched healthy controls (n = 32), and patients with neovascular AMD (n = 45). All participants underwent a comprehensive ocular examination including fluorescein and indocyanine green angiography for where retinal disease was suspected. Using flow cytometry, we identified Th subsets, Tregs, and Th-like Tregs. Plasma samples were stored at -80°C to investigate plasma cytokines of interest. Results Compared to healthy controls, patients with PCV had lower percentages of Tregs (8.7% ± 2.8% vs. 7.3% ± 1.7%, P = 0.027), which were significantly more Th2-like polarized (42.6% ± 13.3% vs. 50.5% ± 13.0%, P = 0.029). These changes differed from that observed in neovascular AMD, which compared to healthy controls had fewer Th1/Th17 cells (3.6% ± 2.7% vs. 2.4% ± 2.5%, P = 0.049), comparable Treg levels, and no distinct polarization of Th-like Tregs. Because of these findings, we measured plasma IL-4 and IL-33 levels. Plasma IL-33 in patients with PCV (median 0.30 pg/mL) was twice as high compared to healthy controls (median 0.16 pg/mL; P = 0.037). Conclusions PCV associate with diminished Tregs that are polarized more into a Th2-like phenotype. This is correlated to IL-33 levels, which we also find increased in patients with PCV. Our findings suggest a possible role for Th2-like Tregs and IL-33 in PCV.
Collapse
Affiliation(s)
- Yousif Subhi
- Clinical Eye Research Division, Department of Ophthalmology, Zealand University Hospital, Roskilde, Denmark.,Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark
| | - Marie Krogh Nielsen
- Clinical Eye Research Division, Department of Ophthalmology, Zealand University Hospital, Roskilde, Denmark.,Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark
| | - Christopher Rue Molbech
- Clinical Eye Research Division, Department of Ophthalmology, Zealand University Hospital, Roskilde, Denmark.,Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark
| | - Akio Oishi
- Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Amardeep Singh
- Clinical Eye Research Division, Department of Ophthalmology, Zealand University Hospital, Roskilde, Denmark.,Department of Ophthalmology, Skåne University Hospital Malmö-Lund, Lund, Sweden
| | - Mogens Holst Nissen
- Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark.,Eye Research Unit, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Torben Lykke Sørensen
- Clinical Eye Research Division, Department of Ophthalmology, Zealand University Hospital, Roskilde, Denmark.,Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
47
|
Mishra R, Rana S. A rational search for discovering potential neutraligands of human complement fragment 5a (hC5a). Bioorg Med Chem 2019; 27:115052. [DOI: 10.1016/j.bmc.2019.115052] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 08/15/2019] [Accepted: 08/17/2019] [Indexed: 12/13/2022]
|
48
|
Tsai IJ, Lin WC, Yang YH, Tseng YL, Lin YH, Chou CH, Tsau YK. High Concentration of C5a-Induced Mitochondria-Dependent Apoptosis in Murine Kidney Endothelial Cells. Int J Mol Sci 2019; 20:ijms20184465. [PMID: 31510052 PMCID: PMC6770645 DOI: 10.3390/ijms20184465] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Revised: 09/02/2019] [Accepted: 09/09/2019] [Indexed: 12/22/2022] Open
Abstract
Patients with a relapse of idiopathic nephrotic syndrome have significantly increased levels of serum complement component 5a (C5a), and proteinuria has been noted in mice treated with C5a via changes in permeability of kidney endothelial cells (KECs) in established animal models. However, the apoptosis of KECs treated with high concentrations of C5a has also been observed. As mitochondrial damage is known to be important in cell apoptosis, the aim of this study was to examine the association between C5a-induced mouse KEC apoptosis and mitochondrial damage. Mouse KECs were isolated and treated with different concentrations of C5a. Cell viability assays showed that a high-concentration mouse recombinant protein C5a (rmC5a) treatment reduced mouse KEC growth. Cell cycle phase analysis, including apoptosis (sub-G1 phase) showed an increased percentage of the subG1 phase with a high-concentration rmC5a treatment. Cytochrome c and caspase 3/9 activities were significantly induced in the mouse KECs after a high-dose rmC5a (50 ng/mL) treatment, and this was rescued by pretreatment with the C5a receptor (C5aR) inhibitor (W-54011) and N-acetylcysteine (NAC). Reactive oxygen species (ROS) formation was detected in C5a-treated mouse KECs; however, W-54011 or NAC pretreatment inhibited high-dose rmC5a-induced ROS formation and also reduced cytochrome c release, apoptotic cell formation, and apoptotic DNA fragmentation. These factors determined the apoptosis of mouse KECs treated with high-dose C5a through C5aR and subsequently led to apoptosis via ROS regeneration and cytochrome c release. The results showed that high concentrations of C5a induced mouse KEC apoptosis via a C5aR/ROS/mitochondria-dependent pathway. These findings may shed light on the potential mechanism of glomerular sclerosis, a process in idiopathic nephrotic syndrome causing renal function impairment.
Collapse
Affiliation(s)
- I-Jung Tsai
- Department of Pediatrics, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei 100, Taiwan.
| | - Wei-Chou Lin
- Department of Pathology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei 100, Taiwan.
| | - Yao-Hsu Yang
- Department of Pediatrics, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei 100, Taiwan.
| | - Yu-Lin Tseng
- Department of Pediatrics, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei 100, Taiwan.
| | - Yen-Hung Lin
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei 100, Taiwan.
| | - Chia-Hung Chou
- Department of Obstetrics and Gynecology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei 100, Taiwan.
| | - Yong-Kwei Tsau
- Department of Pediatrics, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei 100, Taiwan.
| |
Collapse
|
49
|
Murakami Y, Ishikawa K, Nakao S, Sonoda KH. Innate immune response in retinal homeostasis and inflammatory disorders. Prog Retin Eye Res 2019; 74:100778. [PMID: 31505218 DOI: 10.1016/j.preteyeres.2019.100778] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 08/12/2019] [Accepted: 09/02/2019] [Indexed: 01/03/2023]
Abstract
Innate immune cells such as neutrophils, monocyte-macrophages and microglial cells are pivotal for the health and disease of the retina. For the maintenance of retinal homeostasis, these cells and immunosuppressive molecules in the eye actively regulate the induction and the expression of inflammation in order to prevent excessive activation and subsequent tissue damage. In the disease context, these regulatory mechanisms are modulated genetically and/or by environmental stimuli such as damage-associated molecular patterns (DAMPs), and a chronic innate immune response regulates or contributes to the formation of diverse retinal disorders such as uveitis, retinitis pigmentosa, retinal vascular diseases and retinal fibrosis. Here we summarize the recent knowledge regarding the innate immune response in both ocular immune regulation and inflammatory retinal diseases, and we describe the potential of the innate immune response as a biomarker and therapeutic target.
Collapse
Affiliation(s)
- Yusuke Murakami
- Department of Ophthalmology, Graduate School of Medical Science, Kyushu University, Fukuoka, 812-8582, Japan
| | - Keijiro Ishikawa
- Department of Ophthalmology, Graduate School of Medical Science, Kyushu University, Fukuoka, 812-8582, Japan
| | - Shintaro Nakao
- Department of Ophthalmology, Graduate School of Medical Science, Kyushu University, Fukuoka, 812-8582, Japan
| | - Koh-Hei Sonoda
- Department of Ophthalmology, Graduate School of Medical Science, Kyushu University, Fukuoka, 812-8582, Japan.
| |
Collapse
|
50
|
Subhi Y, Nielsen MK, Molbech CR, Liisborg C, Søndergaard HB, Sellebjerg F, Sørensen TL. The transcriptome of peripheral blood mononuclear cells in patients with clinical subtypes of late age-related macular degeneration. IMMUNITY & AGEING 2019; 16:20. [PMID: 31428180 PMCID: PMC6696679 DOI: 10.1186/s12979-019-0160-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Accepted: 08/08/2019] [Indexed: 12/29/2022]
Abstract
Background Peripheral blood mononuclear cells (PBMCs) are implicated in the pathogenesis of age-related macular degeneration (AMD). We here mapped the global gene transcriptome of PBMCs from patients with different clinical subtypes of late AMD. Results We sampled fresh venous blood from patients with geographic atrophy (GA) secondary to AMD without choroidal neovascularizations (n = 19), patients with neovascular AMD without GA (n = 38), patients with polypoidal choroidal vasculopathy (PCV) (n = 19), and aged control individuals with healthy retinae (n = 20). We isolated PBMCs, extracted RNA, and used microarray to investigate gene expression. Volcano plots identified statistically significant differentially expressed genes (P < 0.05) at a high magnitude (≥30% higher/lower) for GA (62 genes), neovascular AMD (41 genes), and PCV (41 genes). These clinical subtypes differed substantially across gene expression and the following pathways identified in enrichment analyses. In a subgroup analysis, we investigated presence vs. absence of subretinal fibrosis and found 826 differentially expressed genes (≥30% higher/lower, P < 0.05) with relation to mRNA splicing, endothelial migration, and interleukin-1 signaling. Conclusions We here map the global gene transcriptome of PBMCs related to clinical subtypes of late AMD and find evidence of subtype-specific immunological involvement. Our findings provide a transcriptomic insight into the systemic immunity associated with AMD. Electronic supplementary material The online version of this article (10.1186/s12979-019-0160-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yousif Subhi
- 1Clinical Eye Research Division, Department of Ophthalmology, Zealand University Hospital, Vestermarksvej 23, DK-4000 Roskilde, Denmark
| | - Marie Krogh Nielsen
- 1Clinical Eye Research Division, Department of Ophthalmology, Zealand University Hospital, Vestermarksvej 23, DK-4000 Roskilde, Denmark
| | - Christopher Rue Molbech
- 1Clinical Eye Research Division, Department of Ophthalmology, Zealand University Hospital, Vestermarksvej 23, DK-4000 Roskilde, Denmark.,2Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Charlotte Liisborg
- 1Clinical Eye Research Division, Department of Ophthalmology, Zealand University Hospital, Vestermarksvej 23, DK-4000 Roskilde, Denmark.,2Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Helle Bach Søndergaard
- 3Danish Multiple Sclerosis Center, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Finn Sellebjerg
- 1Clinical Eye Research Division, Department of Ophthalmology, Zealand University Hospital, Vestermarksvej 23, DK-4000 Roskilde, Denmark.,3Danish Multiple Sclerosis Center, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Torben Lykke Sørensen
- 1Clinical Eye Research Division, Department of Ophthalmology, Zealand University Hospital, Vestermarksvej 23, DK-4000 Roskilde, Denmark.,2Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|