1
|
Sheibani N, Song YS, Farnoodian M, Inampudi S, Hanna B, Wang S, Darjatmoko SR, Sorenson CM. Bim Expression Influences Choroidal Endothelial Cell Characteristics and Their Response to Therapeutic Intervention. Int J Mol Sci 2024; 25:10254. [PMID: 39408582 PMCID: PMC11476819 DOI: 10.3390/ijms251910254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 09/17/2024] [Accepted: 09/20/2024] [Indexed: 10/19/2024] Open
Abstract
In the aging population, choroidal vessels grow through the Bruch's membrane, resulting in a loss of central vision due to choroidal neovascularization (CNV). During active neovascularization, CNV is associated with inappropriate levels of apoptosis in multiple cell types, including choroidal endothelial cells (ChECs). Bim is a pro-apoptotic member of the Bcl-2 family. It is essential for cell apoptosis due to exposure to drugs such as dexamethasone or decreased pro-survival factors, including vascular endothelial growth factor (VEGF). To better elucidate the cell autonomous contribution of Bim expression in the integrity and neovascularization of the choroidal vasculature, we isolated ChECs from wild-type and Bim-deficient (Bim-/-) mice. ChECs lacking Bim expression demonstrated increased expression of VEGF, osteopontin, and the inflammatory cytokines Rantes/Ccl5 and IL6. Bim-/- ChECs were more proliferative and demonstrated an increased capacity to undergo capillary morphogenesis. Anti-VEGF had a diminished capacity to disrupt capillary morphogenesis in Bim-/- ChECs. In vivo, utilizing the mouse laser photocoagulation model, anti-VEGF treatment mitigated CNV in wild-type but not Bim-/- mice. We also tested other modalities that are thought to not require the intrinsic death pathway for their function and showed that propranolol, anti-CTGF, and the TSP1-mimetic peptide ABT898 mitigated CNV in mice lacking Bim expression to varying degrees. Thus, in ChECs, Bim expression could impact the effectiveness of treatment modalities that require the intrinsic death pathway to mitigate CNV.
Collapse
Affiliation(s)
- Nader Sheibani
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (N.S.); (Y.-S.S.); (M.F.); (S.W.); (S.R.D.)
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- McPherson Eye Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Yong-Seok Song
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (N.S.); (Y.-S.S.); (M.F.); (S.W.); (S.R.D.)
- McPherson Eye Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Mitra Farnoodian
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (N.S.); (Y.-S.S.); (M.F.); (S.W.); (S.R.D.)
- McPherson Eye Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Samay Inampudi
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (S.I.); (B.H.)
| | - Barbara Hanna
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (S.I.); (B.H.)
| | - Shoujian Wang
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (N.S.); (Y.-S.S.); (M.F.); (S.W.); (S.R.D.)
- McPherson Eye Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Soesiawati R. Darjatmoko
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (N.S.); (Y.-S.S.); (M.F.); (S.W.); (S.R.D.)
- McPherson Eye Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Christine M. Sorenson
- McPherson Eye Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (S.I.); (B.H.)
| |
Collapse
|
2
|
Bloomfield CL, Gong J, Droho S, Makinde HM, Gurra MG, Stumpf CH, Kharel A, Gadhvi G, Winter DR, Cui W, Cuda CM, Lavine JA. Retinal microglia express more MHC class I and promote greater T-cell-driven inflammation than brain microglia. Front Immunol 2024; 15:1399989. [PMID: 38799448 PMCID: PMC11116593 DOI: 10.3389/fimmu.2024.1399989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 04/22/2024] [Indexed: 05/29/2024] Open
Abstract
Introduction Macrophage function is determined by microenvironment and origin. Brain and retinal microglia are both derived from yolk sac progenitors, yet their microenvironments differ. Utilizing single-cell RNA sequencing (scRNA-seq) data from mice, we tested the hypothesis that retinal and brain microglia exhibit distinct transcriptional profiles due to their unique microenvironments. Methods Eyes and brains from 2-4 month wildtype mice were combined (20 eyes; 3 brains) to yield one biologically diverse sample per organ. Each tissue was digested into single cell suspensions, enriched for immune cells, and sorted for scRNA-seq. Analysis was performed in Seurat v3 including clustering, integration, and differential expression. Multi-parameter flow cytometry was used for validation of scRNA-seq results. Lymphocytic choriomeningitis virus (LCMV) Clone 13, which produces a systemic, chronic, and neurotropic infection, was used to validate scRNA-seq and flow cytometry results in vivo. Results Cluster analysis of integrated gene expression data from eye and brain identified 6 Tmem119 + P2ry12 + microglial clusters. Differential expression analysis revealed that eye microglia were enriched for more pro-inflammatory processes including antigen processing via MHC class I (14.0-fold, H2-D1 and H2-K1) and positive regulation of T-cell immunity (8.4-fold) compared to brain microglia. Multi-parameter flow cytometry confirmed that retinal microglia expressed 3.2-fold greater H2-Db and 263.3-fold more H2-Kb than brain microglia. On Day 13 and 29 after LCMV infection, CD8+ T-cell density was greater in the retina than the brain. Discussion Our data demonstrate that the microenvironment of retina and brain differs, resulting in microglia-specific gene expression changes. Specifically, retinal microglia express greater MHC class I by scRNA-seq and multi-parameter flow cytometry, resulting in a possibly enhanced capability to stimulate CD8+ T-cell inflammation during LCMV infection. These results may explain tissue-specific differences between retina and brain during systemic viral infections and CD8+ T-cell driven autoimmune disease.
Collapse
Affiliation(s)
- Christina L. Bloomfield
- Department of Medicine, University of Illinois College of Medicine, Rockford, IL, United States
| | - Joyce Gong
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Steven Droho
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Hadijat M. Makinde
- Division of Rheumatology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Miranda G. Gurra
- Division of Biostatistics, Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Cecilia H. Stumpf
- Division of Rheumatology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Arjun Kharel
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Gaurav Gadhvi
- Division of Rheumatology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Deborah R. Winter
- Division of Rheumatology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Weiguo Cui
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Carla M. Cuda
- Division of Rheumatology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Jeremy A. Lavine
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
3
|
Zuo W, Sun R, Ji Z, Ma G. Macrophage-driven cardiac inflammation and healing: insights from homeostasis and myocardial infarction. Cell Mol Biol Lett 2023; 28:81. [PMID: 37858035 PMCID: PMC10585879 DOI: 10.1186/s11658-023-00491-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 09/19/2023] [Indexed: 10/21/2023] Open
Abstract
Early and prompt reperfusion therapy has markedly improved the survival rates among patients enduring myocardial infarction (MI). Nonetheless, the resulting adverse remodeling and the subsequent onset of heart failure remain formidable clinical management challenges and represent a primary cause of disability in MI patients worldwide. Macrophages play a crucial role in immune system regulation and wield a profound influence over the inflammatory repair process following MI, thereby dictating the degree of myocardial injury and the subsequent pathological remodeling. Despite numerous previous biological studies that established the classical polarization model for macrophages, classifying them as either M1 pro-inflammatory or M2 pro-reparative macrophages, this simplistic categorization falls short of meeting the precision medicine standards, hindering the translational advancement of clinical research. Recently, advances in single-cell sequencing technology have facilitated a more profound exploration of macrophage heterogeneity and plasticity, opening avenues for the development of targeted interventions to address macrophage-related factors in the aftermath of MI. In this review, we provide a summary of macrophage origins, tissue distribution, classification, and surface markers. Furthermore, we delve into the multifaceted roles of macrophages in maintaining cardiac homeostasis and regulating inflammation during the post-MI period.
Collapse
Affiliation(s)
- Wenjie Zuo
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, No. 87, Dingjiaqiao, Nanjing, 210009, China
| | - Renhua Sun
- Department of Cardiology, Yancheng No. 1 People's Hospital, No. 66 South Renmin Road, Yancheng, 224000, China
| | - Zhenjun Ji
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, No. 87, Dingjiaqiao, Nanjing, 210009, China
| | - Genshan Ma
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, No. 87, Dingjiaqiao, Nanjing, 210009, China.
| |
Collapse
|
4
|
Droho S, Voigt AP, Sterling JK, Rajesh A, Chan KS, Cuda CM, Perlman H, Lavine JA. NR4A1 deletion promotes pro-angiogenic polarization of macrophages derived from classical monocytes in a mouse model of neovascular age-related macular degeneration. J Neuroinflammation 2023; 20:238. [PMID: 37858232 PMCID: PMC10588116 DOI: 10.1186/s12974-023-02928-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 10/10/2023] [Indexed: 10/21/2023] Open
Abstract
BACKGROUND Neovascular age-related macular degeneration causes vision loss from destructive angiogenesis, termed choroidal neovascularization (CNV). Cx3cr1-/- mice display alterations in non-classical monocytes and microglia with increased CNV size, suggesting that non-classical monocytes may inhibit CNV formation. NR4A1 is a transcription factor that is necessary for maturation of non-classical monocytes from classical monocytes. While Nr4a1-/- mice are deficient in non-classical monocytes, results are confounded by macrophage hyper-activation. Nr4a1se2/se2 mice lack a transcriptional activator, resulting in non-classical monocyte loss without macrophage hyper-activation. MAIN BODY We subjected Nr4a1-/- and Nr4a1se2/se2 mice to the laser-induced CNV model and performed multi-parameter flow cytometry. We found that both models lack non-classical monocytes, but only Nr4a1-/- mice displayed increased CNV area. Additionally, CD11c+ macrophages were increased in Nr4a1-/- mice. Single-cell transcriptomic analysis uncovered that CD11c+ macrophages were enriched from Nr4a1-/- mice and expressed a pro-angiogenic transcriptomic profile that was disparate from prior reports of macrophage hyper-activation. CONCLUSIONS These results suggest that non-classical monocytes are dispensable during CNV, and NR4A1 deficiency results in increased recruitment of pro-angiogenic macrophages.
Collapse
Affiliation(s)
- Steven Droho
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Andrew P Voigt
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Jacob K Sterling
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Amrita Rajesh
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Kyle S Chan
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Carla M Cuda
- Division of Rheumatology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Harris Perlman
- Division of Rheumatology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Jeremy A Lavine
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA.
| |
Collapse
|
5
|
Tan Y, Huang J, Li D, Zou C, Liu D, Qin B. Single-cell RNA sequencing in dissecting microenvironment of age-related macular degeneration: Challenges and perspectives. Ageing Res Rev 2023; 90:102030. [PMID: 37549871 DOI: 10.1016/j.arr.2023.102030] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 04/29/2023] [Accepted: 08/04/2023] [Indexed: 08/09/2023]
Abstract
Age-related macular degeneration (AMD) is the leading cause of blindness in individuals over the age of 50 years, yet its etiology and pathogenesis largely remain uncovered. Single-cell RNA sequencing (scRNA-seq) technologies are recently developed and have a number of advantages over conventional bulk RNA sequencing techniques in uncovering the heterogeneity of complex microenvironments containing numerous cell types and cell communications during various biological processes. In this review, we summarize the latest discovered cellular components and regulatory mechanisms during AMD development revealed by scRNA-seq. In addition, we discuss the main challenges and future directions in exploring the pathophysiology of AMD equipped with single-cell technologies. Our review underscores the importance of multimodal single-cell platforms (such as single-cell spatiotemporal multi-omics and single-cell exosome omics) as new approaches for basic and clinical AMD research in identifying biomarker, characterizing cellular responses to drug treatment and environmental stimulation.
Collapse
Affiliation(s)
- Yao Tan
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen, China
| | - Jianguo Huang
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen, China
| | - Deshuang Li
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen, China
| | - Chang Zou
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen, China; Shenzhen Aier Ophthalmic Technology Institute, Shenzhen, China; School of Life and Health Sciences, The Chinese University of Kong Hong, Shenzhen 518000, Guangdong, China.
| | - Dongcheng Liu
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen, China; Shenzhen Aier Ophthalmic Technology Institute, Shenzhen, China.
| | - Bo Qin
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen, China; Shenzhen Aier Ophthalmic Technology Institute, Shenzhen, China; Aier School of Ophthalmology, Central South University, Changsha, China.
| |
Collapse
|
6
|
Huang K, Liu X, Lv Z, Zhang D, Zhou Y, Lin Z, Guo J. MMP9-Responsive Graphene Oxide Quantum Dot-Based Nano-in-Micro Drug Delivery System for Combinatorial Therapy of Choroidal Neovascularization. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2207335. [PMID: 36871144 DOI: 10.1002/smll.202207335] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 01/29/2023] [Indexed: 06/18/2023]
Abstract
Age-related macular degeneration (AMD), especially wet AMD with choroidal neovascularization (CNV), commonly causes blindness in older patients and disruption of the choroid followed by second-wave injuries, including chronic inflammation, oxidative stress, and excessive matrix metalloproteinase 9 (MMP9) expression. Increased macrophage infiltrate in parallel with microglial activation and MMP9 overexpression on CNV lesions is shown to contribute to the inflammatory process and then enhance pathological ocular angiogenesis. Graphene oxide quantum dots (GOQDs), as natural antioxidants, exert anti-inflammatory effects and minocycline is a specific macrophage/microglial inhibitor that can suppress both macrophage/microglial activation and MMP9 activity. Herein, an MMP9-responsive GOQD-based minocycline-loaded nano-in-micro drug delivery system (C18PGM) is developed by chemically bonding GOQDs to an octadecyl-modified peptide sequence (C18-GVFHQTVS, C18P) that can be specifically cleaved by MMP9. Using a laser-induced CNV mouse model, the prepared C18PGM shows significant MMP9 inhibitory activity and anti-inflammatory action followed by antiangiogenic effects. Moreover, C18PGM combined with antivascular endothelial growth factor antibody bevacizumab markedly increases the antiangiogenesis effect by interfering with the "inflammation-MMP9-angiogenesis" cascade. The prepared C18PGM shows a good safety profile and no obvious ophthalmic or systemic side effects. The results taken together suggest that C18PGM is an effective and novel strategy for combinatorial therapy of CNV.
Collapse
Affiliation(s)
- Keke Huang
- Institute of Advanced Materials for Nano-Bio Applications, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, P. R. China
| | - Xin Liu
- Institute of Advanced Materials for Nano-Bio Applications, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, P. R. China
| | - Ziru Lv
- Institute of Advanced Materials for Nano-Bio Applications, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, P. R. China
| | - Di Zhang
- Institute of Advanced Materials for Nano-Bio Applications, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, P. R. China
| | - Yuling Zhou
- Department of ophthalmology, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Huangshi, Hubei, 435000, P. R. China
| | - Zhiqing Lin
- Institute of Advanced Materials for Nano-Bio Applications, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, P. R. China
| | - Juan Guo
- Department of Ophthalmology, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, Sichuan, 610031, P. R. China
| |
Collapse
|
7
|
Wang X, Wang T, Lam E, Alvarez D, Sun Y. Ocular Vascular Diseases: From Retinal Immune Privilege to Inflammation. Int J Mol Sci 2023; 24:12090. [PMID: 37569464 PMCID: PMC10418793 DOI: 10.3390/ijms241512090] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 07/21/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023] Open
Abstract
The eye is an immune privileged tissue that insulates the visual system from local and systemic immune provocation to preserve homeostatic functions of highly specialized retinal neural cells. If immune privilege is breached, immune stimuli will invade the eye and subsequently trigger acute inflammatory responses. Local resident microglia become active and release numerous immunological factors to protect the integrity of retinal neural cells. Although acute inflammatory responses are necessary to control and eradicate insults to the eye, chronic inflammation can cause retinal tissue damage and cell dysfunction, leading to ocular disease and vision loss. In this review, we summarized features of immune privilege in the retina and the key inflammatory responses, factors, and intracellular pathways activated when retinal immune privilege fails, as well as a highlight of the recent clinical and research advances in ocular immunity and ocular vascular diseases including retinopathy of prematurity, age-related macular degeneration, and diabetic retinopathy.
Collapse
Affiliation(s)
- Xudong Wang
- Department of Ophthalmology, Harvard Medical School, Boston Children’s Hospital, Boston, MA 02115, USA; (X.W.)
| | - Tianxi Wang
- Department of Ophthalmology, Harvard Medical School, Boston Children’s Hospital, Boston, MA 02115, USA; (X.W.)
| | - Enton Lam
- Department of Ophthalmology, Harvard Medical School, Boston Children’s Hospital, Boston, MA 02115, USA; (X.W.)
| | - David Alvarez
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Ye Sun
- Department of Ophthalmology, Harvard Medical School, Boston Children’s Hospital, Boston, MA 02115, USA; (X.W.)
| |
Collapse
|
8
|
Zhou ZY, Chang TF, Lin ZB, Jing YT, Wen LS, Niu YL, Bai Q, Guo CM, Sun JX, Wang YS, Dou GR. Microglial Galectin3 enhances endothelial metabolism and promotes pathological angiogenesis via Notch inhibition by competitively binding to Jag1. Cell Death Dis 2023; 14:380. [PMID: 37369647 DOI: 10.1038/s41419-023-05897-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 05/26/2023] [Accepted: 06/16/2023] [Indexed: 06/29/2023]
Abstract
Microglia were considered as immune cells in inflammation until their angiogenic role was widely understood. Although the pro-inflammatory role of microglia in retinal angiogenesis has been explored, little is known about its role in pro-angiogenesis and the microglia-endothelia interaction. Here, we report that galectin-3 (Gal3) released by activated microglia functions as a communicator between microglia and endothelia and competitively binds to Jag1, thus inhibiting the Notch signaling pathway and enhancing endothelial angiogenic metabolism to promote angiogenesis. These results suggest that Gal3 may be a novel and effective target in the treatment of retinal angiogenesis.
Collapse
Affiliation(s)
- Zi-Yi Zhou
- Department of Ophthalmology, Eye Institute of Chinese PLA, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Tian-Fang Chang
- Department of Ophthalmology, Eye Institute of Chinese PLA, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Zhi-Bin Lin
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Yu-Tong Jing
- Department of Ophthalmology, Eye Institute of Chinese PLA, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Li-Shi Wen
- Department of Ophthalmology, Eye Institute of Chinese PLA, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Ya-Li Niu
- Department of Ophthalmology, Eye Institute of Chinese PLA, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Qian Bai
- Department of Ophthalmology, Eye Institute of Chinese PLA, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Chang-Mei Guo
- Department of Ophthalmology, Eye Institute of Chinese PLA, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Jia-Xing Sun
- Department of Ophthalmology, Eye Institute of Chinese PLA, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| | - Yu-Sheng Wang
- Department of Ophthalmology, Eye Institute of Chinese PLA, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| | - Guo-Rui Dou
- Department of Ophthalmology, Eye Institute of Chinese PLA, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
9
|
Kantor AB, Akassoglou K, Stavenhagen JB. Fibrin-Targeting Immunotherapy for Dementia. J Prev Alzheimers Dis 2023; 10:647-660. [PMID: 37874085 PMCID: PMC11227370 DOI: 10.14283/jpad.2023.105] [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] [Indexed: 10/25/2023]
Abstract
Blood-brain barrier (BBB) disruption is an early event in the development of Alzheimer's disease. It precedes extracellular deposition of amyloid-β in senile plaques and blood vessel walls, the intracellular accumulation of neurofibrillary tangles containing phosphorylated tau protein, microglial activation, and neuronal cell death. BBB disruption allows the coagulation protein fibrinogen to leak from the blood into the brain, where it is converted by thrombin cleavage into fibrin and deposits in the parenchyma and CNS vessels. Fibrinogen cleavage by thrombin exposes a cryptic epitope termed P2 which can bind CD11b and CD11c on microglia, macrophages and dendritic cells and trigger an inflammatory response toxic to neurons. Indeed, genetic and pharmacological evidence demonstrates a causal role for fibrin in innate immune cell activation and the development of neurodegenerative diseases. The P2 inflammatory epitope is spatially and compositionally distinct from the coagulation epitope on fibrin. Mouse monoclonal antibody 5B8, which targets the P2 epitope without interfering with the clotting process, has been shown to reduce neurodegeneration and neuroinflammation in animal models of Alzheimer's disease and multiple sclerosis. The selectivity and efficacy of this anti-human fibrin-P2 antibody in animal models supports the development of a monoclonal antibody drug targeting fibrin P2 for the treatment of neurodegenerative diseases. THN391 is a humanized, affinity-matured antibody which has a 100-fold greater affinity for fibrin P2 and improved development properties compared to the parental 5B8 antibody. It is currently in a Phase 1 clinical trial.
Collapse
Affiliation(s)
- A B Kantor
- Jeffrey Stavenhagen, PhD, Therini Bio, Inc, Sacramento, CA, USA,
| | | | | |
Collapse
|
10
|
Rajesh A, Droho S, Lavine JA. Macrophages in close proximity to the vitreoretinal interface are potential biomarkers of inflammation during retinal vascular disease. J Neuroinflammation 2022; 19:203. [PMID: 35941655 PMCID: PMC9361599 DOI: 10.1186/s12974-022-02562-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 07/29/2022] [Indexed: 12/21/2022] Open
Abstract
Background Diabetic retinopathy and retinal vein occlusion are vision threatening retinal vascular diseases. Current first-line therapy targets the vascular component, but many patients are treatment-resistant due to unchecked inflammation. Non-invasive inflammatory imaging biomarkers are a significant unmet clinical need for patients. Imaging of macrophage-like cells on the surface of the retina using clinical optical coherence tomography (OCT) is an emerging field. These cells are increased in patients with retinal vascular disease, and could be a potential inflammatory biomarker. However, since OCT is limited by an axial resolution of 5–10 microns, the exact location and identity of these retinal cells is currently unknown. Methods We performed OCT followed by confocal immunofluorescence in wild-type mice to identify macrophages within 5–10 microns of the vitreoretinal interface. Next, we used Cx3cr1CreER/+; Rosa26zsGreen/+ mice to fate map retinal surface macrophages. Using confocal immunofluorescence of retinal sections and flatmounts, we quantified IBA1+Tmem119+CD169neg microglia, IBA1+Tmem119negCD169neg perivascular macrophages, and IBA1+Tmem119negCD169+ vitreal hyalocytes. Finally, we modeled neuroinflammation with CCL2 treatment and characterized retinal surface macrophages using flow cytometry, OCT, and confocal immunofluorescence. Results We were able to detect IBA1+ macrophages within 5–10 microns of the vitreoretinal interface in wild-type mice using OCT followed by confirmatory confocal immunofluorescence. Retinal surface macrophages were 83.5% GFP+ at Week 1 and 82.4% GFP+ at Week 4 using fate mapping mice. At steady state, these macrophages included 82% IBA1+Tmem119+CD169neg microglia, 9% IBA1+Tmem119negCD169+ vitreal hyalocytes, and 9% IBA1+Tmem119negCD169neg perivascular macrophages. After CCL2-driven neuroinflammation, many Ly6C+ cells were detectable on the retinal surface using OCT followed by confocal immunofluorescence. Conclusions Macrophages within close proximity to the vitreoretinal interface are self-renewing cells, and predominantly microglia with minor populations of perivascular macrophages and vitreal hyalocytes at steady state. In the context of neuroinflammation, monocytes and monocyte-derived macrophages are a significant component of retinal surface macrophages. Human OCT-based imaging of retinal surface macrophages is a potential biomarker for inflammation during retinal vascular disease. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02562-3.
Collapse
Affiliation(s)
- Amrita Rajesh
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, 240 E Huron St, McGaw M343, Chicago, IL, 60611, USA
| | - Steven Droho
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, 240 E Huron St, McGaw M343, Chicago, IL, 60611, USA
| | - Jeremy A Lavine
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, 240 E Huron St, McGaw M343, Chicago, IL, 60611, USA.
| |
Collapse
|
11
|
Voigt AP, Mullin NK, Mulfaul K, Lozano LP, Wiley LA, Flamme-Wiese MJ, Boese EA, Han IC, Scheetz TE, Stone EM, Tucker BA, Mullins RF. Choroidal endothelial and macrophage gene expression in atrophic and neovascular macular degeneration. Hum Mol Genet 2022; 31:2406-2423. [PMID: 35181781 PMCID: PMC9307320 DOI: 10.1093/hmg/ddac043] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 01/22/2022] [Accepted: 02/06/2022] [Indexed: 11/22/2022] Open
Abstract
The human choroid is a heterogeneous, highly vascular connective tissue that dysfunctions in age-related macular degeneration (AMD). In this study, we performed single-cell RNA sequencing on 21 human choroids, 11 of which were derived from donors with early atrophic or neovascular AMD. Using this large donor cohort, we identified new gene expression signatures and immunohistochemically characterized discrete populations of resident macrophages, monocytes/inflammatory macrophages and dendritic cells. These three immune populations demonstrated unique expression patterns for AMD genetic risk factors, with dendritic cells possessing the highest expression of the neovascular AMD-associated MMP9 gene. Additionally, we performed trajectory analysis to model transcriptomic changes across the choroidal vasculature, and we identified expression signatures for endothelial cells from choroidal arterioles and venules. Finally, we performed differential expression analysis between control, early atrophic AMD, and neovascular AMD samples, and we observed that early atrophic AMD samples had high expression of SPARCL1, a gene that has been shown to increase in response to endothelial damage. Likewise, neovascular endothelial cells harbored gene expression changes consistent with endothelial cell damage and demonstrated increased expression of the sialomucins CD34 and ENCM, which were also observed at the protein level within neovascular membranes. Overall, this study characterizes the molecular features of new populations of choroidal endothelial cells and mononuclear phagocytes in a large cohort of AMD and control human donors.
Collapse
Affiliation(s)
- Andrew P Voigt
- Department of Ophthalmology and Visual Sciences, The University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
- Institute for Vision Research, The University of Iowa, Iowa City, IA 52242, USA
| | - Nathaniel K Mullin
- Department of Ophthalmology and Visual Sciences, The University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
- Institute for Vision Research, The University of Iowa, Iowa City, IA 52242, USA
| | - Kelly Mulfaul
- Department of Ophthalmology and Visual Sciences, The University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
- Institute for Vision Research, The University of Iowa, Iowa City, IA 52242, USA
| | - Lola P Lozano
- Department of Ophthalmology and Visual Sciences, The University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
- Institute for Vision Research, The University of Iowa, Iowa City, IA 52242, USA
| | - Luke A Wiley
- Department of Ophthalmology and Visual Sciences, The University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
- Institute for Vision Research, The University of Iowa, Iowa City, IA 52242, USA
| | - Miles J Flamme-Wiese
- Department of Ophthalmology and Visual Sciences, The University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
- Institute for Vision Research, The University of Iowa, Iowa City, IA 52242, USA
| | - Erin A Boese
- Department of Ophthalmology and Visual Sciences, The University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
- Institute for Vision Research, The University of Iowa, Iowa City, IA 52242, USA
| | - Ian C Han
- Department of Ophthalmology and Visual Sciences, The University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
- Institute for Vision Research, The University of Iowa, Iowa City, IA 52242, USA
| | - Todd E Scheetz
- Department of Ophthalmology and Visual Sciences, The University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
- Institute for Vision Research, The University of Iowa, Iowa City, IA 52242, USA
| | - Edwin M Stone
- Department of Ophthalmology and Visual Sciences, The University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
- Institute for Vision Research, The University of Iowa, Iowa City, IA 52242, USA
| | - Budd A Tucker
- Department of Ophthalmology and Visual Sciences, The University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
- Institute for Vision Research, The University of Iowa, Iowa City, IA 52242, USA
| | - Robert F Mullins
- Department of Ophthalmology and Visual Sciences, The University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
- Institute for Vision Research, The University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
12
|
Qin X, Xiao L, Li N, Hou C, Li W, Li J, Yan N, Lin Y. Tetrahedral framework nucleic acids-based delivery of microRNA-155 inhibits choroidal neovascularization by regulating the polarization of macrophages. Bioact Mater 2021; 14:134-144. [PMID: 35310341 PMCID: PMC8892086 DOI: 10.1016/j.bioactmat.2021.11.031] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 10/11/2021] [Accepted: 11/23/2021] [Indexed: 01/10/2023] Open
Abstract
Choroidal neovascularization (CNV) is a common pathological feature of various eye diseases and an important cause of visual impairment in middle-aged and elderly patients. In previous studies, tetrahedral framework nucleic acids (tFNAs) showed good carrier performance. In this experiment, we developed microRNA-155-equipped tFNAs (T-155) and explored its biological effects on CNV. Based on the results of in-vitro experiments, T-155 could regulate macrophages into the antiangiogenic M1 type. Then, we injected T-155 into the vitreous of laser-induced CNV model mice and found that T-155 significantly reduced the size and area of CNV, inhibited blood vessel leakage. In summary, we prove that T-155 could regulate the inflammatory process of CNV by polarizing macrophages, thereby improving the symptoms of CNV. Thus, T-155 might become a new DNA-based drug with great potential for treating CNV. T-155 could regulate the inflammatory process of CNV by polarizing macrophages, thereby improving the symptoms of CNV. Thus, T-155 might become a new DNA-based drug with great potential for treating CNV.
Collapse
|
13
|
Droho S, Cuda CM, Perlman H, Lavine JA. Macrophage-derived interleukin-6 is necessary and sufficient for choroidal angiogenesis. Sci Rep 2021; 11:18084. [PMID: 34508129 PMCID: PMC8433398 DOI: 10.1038/s41598-021-97522-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 08/25/2021] [Indexed: 01/18/2023] Open
Abstract
Neovascular age-related macular degeneration (nAMD) commonly causes vision loss from aberrant angiogenesis, termed choroidal neovascularization (CNV). Interleukin-6 (IL6) is a pro-inflammatory and pro-angiogenic cytokine that is correlated with AMD progression and nAMD activity. We hypothesize that anti-IL6 therapy is a potential nAMD therapeutic. We found that IL6 levels were increased after laser injury and expressed by macrophages. Il6-deficiency decreased laser-induced CNV area and exogenous IL6 addition increased choroidal sprouting angiogenesis. Il6-null mice demonstrated equally increased macrophage numbers as wildtype mice. At steady state, IL6R expression was detected on peripheral blood and ocular monocytes. After laser injury, the number of IL6R+Ly6C+ monocytes in blood and IL6R+ macrophages in the eye were increased. In human choroid, macrophages expressed IL6, IL6R, and IL6ST. Furthermore, IL6R+ macrophages displayed a transcriptional profile consistent with STAT3 (signal transducer and activator of transcription 3) activation and angiogenesis. Our data show that IL6 is both necessary and sufficient for choroidal angiogenesis. Macrophage-derived IL6 may stimulate choroidal angiogenesis via classical activation of IL6R+ macrophages, which then stimulate angiogenesis. Targeting IL6 or the IL6R could be an effective adjunctive therapy for treatment-resistant nAMD patients.
Collapse
Affiliation(s)
- Steven Droho
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Carla M Cuda
- Department of Medicine, Division of Rheumatology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Harris Perlman
- Department of Medicine, Division of Rheumatology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Jeremy A Lavine
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
14
|
Droho S, Perlman H, Lavine JA. Dendritic cells play no significant role in the laser-induced choroidal neovascularization model. Sci Rep 2021; 11:17254. [PMID: 34446787 PMCID: PMC8390527 DOI: 10.1038/s41598-021-96704-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 08/12/2021] [Indexed: 11/25/2022] Open
Abstract
Age-related macular degeneration (AMD) is genetically associated with complement. Dendritic cells (DCs) play key roles during innate and adaptive immunity, and express complement components and their receptors. We investigated ocular DC heterogeneity and the role of DCs in the laser-induced choroidal neovascularization (CNV) model. In order to determine the function of DCs, we used two models of DC deficiency: the Flt3-/- and Flt3l-/- mouse. We identified three types of ocular DCs: plasmacytoid DC, classical DC-1, and classical DC-2. At steady-state, classical DCs were found in the iris and choroid but were not detectable in the retina. Plasmacytoid DCs existed at very low levels in iris, choroid, and retina. After laser injury, the number of each DC subset was up-regulated in the choroid and retina. In Flt3-/- mice, we found reduced numbers of classical DCs at steady-state, but each DC subset equally increased after laser injury between wildtype and Flt3-/- mice. In Flt3l-/- mice, each DC subsets was severely reduced after laser injury. Neither Flt3-/- or Flt3l-/- mice demonstrated reduced CNV area compared to wildtype mice. DCs do not play any significant role during the laser-induced CNV model of neovascular AMD.
Collapse
Affiliation(s)
- Steven Droho
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Harris Perlman
- Division of Rheumatology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Jeremy A Lavine
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
15
|
Schlecht A, Zhang P, Wolf J, Thien A, Rosmus DD, Boneva S, Schlunck G, Lange C, Wieghofer P. Secreted Phosphoprotein 1 Expression in Retinal Mononuclear Phagocytes Links Murine to Human Choroidal Neovascularization. Front Cell Dev Biol 2021; 8:618598. [PMID: 33585455 PMCID: PMC7876283 DOI: 10.3389/fcell.2020.618598] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 12/01/2020] [Indexed: 01/08/2023] Open
Abstract
Age-related macular degeneration (AMD) represents the most common cause of blindness in the elderly in the Western world. An impairment of the outer blood-retina barrier and a localized inflammatory microenvironment cause sprouting of choroidal neovascular membranes (CNV) in neovascular AMD that are in intimate contact with surrounding myeloid cells, such as retinal microglia, and ultimately lead to visual impairment. The discovery of novel target molecules to interfere with angiogenesis and inflammation is vital for future treatment approaches in AMD patients. To explore the transcriptional profile and the function of retinal microglia at sites of CNV, we performed a comprehensive RNA-seq analysis of retinal microglia in the mouse model of laser-induced choroidal neovascularization (mCNV). Here, we identified the angiogenic factor Osteopontin (Opn), also known as "secreted phosphoprotein 1" (Spp1), as one of the most highly expressed genes in retinal microglia in the course of CNV formation. We confirmed the presence of SPP1 at the lesion site in recruited retinal microglia in Cx3cr1 CreER:Rosa26-tdTomato reporter mice by confocal microscopy and in whole retinal tissue lysates by ELISA highlighting a massive local production of SPP1. Inhibition of SPP1 by intravitreal injection of an anti-SPP1 antibody significantly increased the lesion size compared to IgG-treated control eyes. In line with our results in rodents, we found an increased SPP1 mRNA expression in surgically extracted human choroidal neovascular (hCNV) membranes by the quantitative RNA-seq approach of massive analysis of cDNA ends (MACE). Numerous IBA1+SPP1+ myeloid cells were detected in human CNV membranes. Taken together, these results highlight the importance of SPP1 in the formation of CNV and potentially offer new opportunities for therapeutic intervention by modulating the SPP1 pathway.
Collapse
Affiliation(s)
- Anja Schlecht
- Eye Center, Medical Center, Medical Faculty, University of Freiburg, Freiburg, Germany
| | - Peipei Zhang
- Eye Center, Medical Center, Medical Faculty, University of Freiburg, Freiburg, Germany
| | - Julian Wolf
- Eye Center, Medical Center, Medical Faculty, University of Freiburg, Freiburg, Germany
| | - Adrian Thien
- Eye Center, Medical Center, Medical Faculty, University of Freiburg, Freiburg, Germany
| | | | - Stefaniya Boneva
- Eye Center, Medical Center, Medical Faculty, University of Freiburg, Freiburg, Germany
| | - Günther Schlunck
- Eye Center, Medical Center, Medical Faculty, University of Freiburg, Freiburg, Germany
| | - Clemens Lange
- Eye Center, Medical Center, Medical Faculty, University of Freiburg, Freiburg, Germany
| | | |
Collapse
|
16
|
Reekie IR, Sharma S, Foers A, Sherlock J, Coles MC, Dick AD, Denniston AK, Buckley CD. The Cellular Composition of the Uveal Immune Environment. Front Med (Lausanne) 2021; 8:721953. [PMID: 34778287 PMCID: PMC8586083 DOI: 10.3389/fmed.2021.721953] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 10/05/2021] [Indexed: 12/26/2022] Open
Abstract
The uveal tract consists of the iris, the ciliary body and the choroid; these three distinct tissues form a continuous layer within the eye. Uveitis refers to inflammation of any region of the uveal tract. Despite being grouped together anatomically, the iris, ciliary body and choroid are distinct functionally, and inflammatory diseases may affect only one part and not the others. Cellular structure of tissues direct their function, and understanding the cellular basis of the immune environment of a tissue in health, the "steady state" on which the perturbations of disease are superimposed, is vital to understanding the pathogenesis of those diseases. A contemporary understanding of the immune system accepts that haematopoietic and yolk sac derived leukocytes, though vital, are not the only players of importance. An array of stromal cells, connective tissue cells such as fibroblasts and endothelial cells, may also have a role in the inflammatory reaction seen in several immune-mediated diseases. In this review we summarise what is known about the cellular composition of the uveal tract and the roles these disparate cell types have to play in immune homeostasis. We also discuss some unanswered questions surrounding the constituents of the resident leukocyte population of the different uveal tissues, and we look ahead to the new understanding that modern investigative techniques such as single cell transcriptomics, multi-omic data integration and highly-multiplexed imaging techniques may bring to the study of the uvea and uveitis, as they already have to other immune mediated inflammatory diseases.
Collapse
Affiliation(s)
- Ian R. Reekie
- The Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| | - Srilakshmi Sharma
- The Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
- Oxford University Hospitals National Health Service (NHS) Foundation Trust, Oxford Eye Hospital, Oxford, United Kingdom
| | - Andrew Foers
- The Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| | - Jonathan Sherlock
- The Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| | - Mark C. Coles
- The Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| | - Andrew D. Dick
- School of Clinical Sciences, University of Bristol, Bristol, United Kingdom
- National Institute for Health Research Biomedical Research Centre, Institute of Ophthalmology, Moorfields Eye Hospital, University College London, London, United Kingdom
| | - Alastair K. Denniston
- Institute for Inflammation and Ageing, College of Medical and Dental Sciences, Queen Elizabeth Hospital, University of Birmingham, Birmingham, United Kingdom
| | - Christopher D. Buckley
- The Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
- Institute for Inflammation and Ageing, College of Medical and Dental Sciences, Queen Elizabeth Hospital, University of Birmingham, Birmingham, United Kingdom
- *Correspondence: Christopher D. Buckley
| |
Collapse
|