1
|
Khan AH, Mulfaul K. Choroidal macrophages in homeostasis, aging and age-related macular degeneration. Exp Eye Res 2025; 250:110159. [PMID: 39577606 DOI: 10.1016/j.exer.2024.110159] [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: 07/30/2024] [Revised: 11/13/2024] [Accepted: 11/14/2024] [Indexed: 11/24/2024]
Abstract
With increasing age, the optimal functioning of the choroid is essential for efficient removal of waste products formed from photoreceptor renewal. A decline in regulatory elements of the immune system, termed immunosenescence, and the failure of para-inflammation to restore tissue homeostasis can result in the progression of healthy aging to sight-threatening inflammation of the choroid. Macrophages are uniquely situated between the innate and adaptive immune systems, with a high capacity for phagocytosis, recognition of complement components, as well as antigen presentation. In this review, we provide an overview of macrophages and their properties in the healthy choroid and cover the impact of aging, immunosenescence and inflammaging on the function of choroidal macrophages. We will discuss the impact of age on macrophage phenotype and behaviour in the pathophysiology of age-related macular degeneration.
Collapse
Affiliation(s)
- Adnan H Khan
- Institute for Vision Research, University of Iowa, Iowa City, IA, USA; Department of Ophthalmology & Visual Sciences, University of Iowa, Iowa City, IA, USA
| | - Kelly Mulfaul
- Institute for Vision Research, University of Iowa, Iowa City, IA, USA; Department of Ophthalmology & Visual Sciences, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
2
|
Wu M, Fletcher EL, Chinnery HR, Downie LE, Mueller SN. Redefining our vision: an updated guide to the ocular immune system. Nat Rev Immunol 2024; 24:896-911. [PMID: 39215057 DOI: 10.1038/s41577-024-01064-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/01/2024] [Indexed: 09/04/2024]
Abstract
Balanced immune responses in the eyes are crucial to preserve vision. The ocular immune system has long been considered distinct, owing to the so-called 'immune privilege' of its component tissues. More recently, intravital imaging and transcriptomic techniques have reshaped scientific understanding of the ocular immune landscape, such as revealing the specialization of immune cell populations in the various tissues of the eye. As knowledge of the phenotypes of corneal and retinal immune cells has evolved, links to both the systemic immune system, and the central and peripheral nervous systems, have been identified. Using intravital imaging, T cells have recently been found to reside in, and actively patrol, the healthy human cornea. Disease-associated retinal microglia with links to retinal degeneration have also been identified. This Review provides an updated guide to the ocular immune system, highlighting current knowledge of the immune cells that are present in steady-state and specific diseased ocular tissues, as well as evidence for their relationship to systemic disease. In addition, we discuss emerging intravital imaging techniques that can be used to visualize immune cell morphology and dynamics in living human eyes and how these could be applied to advance understanding of the human immune system.
Collapse
Affiliation(s)
- Mengliang Wu
- Department of Optometry and Vision Sciences, The University of Melbourne, Carlton, Victoria, Australia
- Department of Microbiology and Immunology, The University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Erica L Fletcher
- Department of Anatomy and Physiology, The University of Melbourne, Carlton, Victoria, Australia
| | - Holly R Chinnery
- Department of Optometry and Vision Sciences, The University of Melbourne, Carlton, Victoria, Australia.
- Lions Eye Institute, Nedlands, Western Australia, Australia.
- Optometry, The University of Western Australia, Crawley, Western Australia, Australia.
| | - Laura E Downie
- Department of Optometry and Vision Sciences, The University of Melbourne, Carlton, Victoria, Australia.
| | - Scott N Mueller
- Department of Microbiology and Immunology, The University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia.
| |
Collapse
|
3
|
Xu C, Fu X, Qin H, Yao K. Traversing the epigenetic landscape: DNA methylation from retina to brain in development and disease. Front Cell Neurosci 2024; 18:1499719. [PMID: 39678047 PMCID: PMC11637887 DOI: 10.3389/fncel.2024.1499719] [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: 09/21/2024] [Accepted: 11/18/2024] [Indexed: 12/17/2024] Open
Abstract
DNA methylation plays a crucial role in development, aging, degeneration of various tissues and dedifferentiated cells. This review explores the multifaceted impact of DNA methylation on the retina and brain during development and pathological processes. First, we investigate the role of DNA methylation in retinal development, and then focus on retinal diseases, detailing the changes in DNA methylation patterns in diseases such as diabetic retinopathy (DR), age-related macular degeneration (AMD), and glaucoma. Since the retina is considered an extension of the brain, its unique structure allows it to exhibit similar immune response mechanisms to the brain. We further extend our exploration from the retina to the brain, examining the role of DNA methylation in brain development and its associated diseases, such as Alzheimer's disease (AD) and Huntington's disease (HD) to better understand the mechanistic links between retinal and brain diseases, and explore the possibility of communication between the visual system and the central nervous system (CNS) from an epigenetic perspective. Additionally, we discuss neurodevelopmental brain diseases, including schizophrenia (SZ), autism spectrum disorder (ASD), and intellectual disability (ID), focus on how DNA methylation affects neuronal development, synaptic plasticity, and cognitive function, providing insights into the molecular mechanisms underlying neurodevelopmental disorders.
Collapse
Affiliation(s)
- Chunxiu Xu
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan, China
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Xuefei Fu
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan, China
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Huan Qin
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan, China
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Kai Yao
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan, China
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, China
| |
Collapse
|
4
|
Blasdel N, Bhattacharya S, Donaldson PC, Reh TA, Todd L. Monocyte Invasion into the Retina Restricts the Regeneration of Neurons from Müller Glia. J Neurosci 2024; 44:e0938242024. [PMID: 39353729 PMCID: PMC11561870 DOI: 10.1523/jneurosci.0938-24.2024] [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: 05/16/2024] [Revised: 09/20/2024] [Accepted: 09/24/2024] [Indexed: 10/04/2024] Open
Abstract
Endogenous reprogramming of glia into neurogenic progenitors holds great promise for neuron restoration therapies. Using lessons from regenerative species, we have developed strategies to stimulate mammalian Müller glia to regenerate neurons in vivo in the adult retina. We have demonstrated that the transcription factor Ascl1 can stimulate Müller glia neurogenesis. However, Ascl1 is only able to reprogram a subset of Müller glia into neurons. We have reported that neuroinflammation from microglia inhibits neurogenesis from Müller glia. Here we found that the peripheral immune response is a barrier to CNS regeneration. We show that monocytes from the peripheral immune system infiltrate the injured retina and negatively influence neurogenesis from Müller glia. Using CCR2 knock-out mice of both sexes, we found that preventing monocyte infiltration improves the neurogenic and proliferative capacity of Müller glia stimulated by Ascl1. Using scRNA-seq analysis, we identified a signaling axis wherein Osteopontin, a cytokine highly expressed by infiltrating immune cells is sufficient to suppress mammalian neurogenesis. This work implicates the response of the peripheral immune system as a barrier to regenerative strategies of the retina.
Collapse
Affiliation(s)
- Nicolai Blasdel
- Department of Biological Structure, University of Washington, Seattle, Washington 98195
| | - Sucheta Bhattacharya
- Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, Syracuse, New York 13210
| | - Phoebe C Donaldson
- Department of Biological Structure, University of Washington, Seattle, Washington 98195
| | - Thomas A Reh
- Department of Biological Structure, University of Washington, Seattle, Washington 98195
| | - Levi Todd
- Department of Biological Structure, University of Washington, Seattle, Washington 98195
- Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, Syracuse, New York 13210
| |
Collapse
|
5
|
Hyttinen JMT, Koskela A, Blasiak J, Kaarniranta K. Autophagy in drusen biogenesis secondary to age-related macular degeneration. Acta Ophthalmol 2024; 102:759-772. [PMID: 39087629 DOI: 10.1111/aos.16744] [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: 03/25/2024] [Accepted: 07/11/2024] [Indexed: 08/02/2024]
Abstract
Age-related macular degeneration (AMD) is an emerging cause of blindness in aged people worldwide. One of the key signs of AMD is the degeneration of the retinal pigment epithelium (RPE), which is indispensable for the maintenance of the adjacent photoreceptors. Because of impaired energy metabolism resulting from constant light exposure, hypoxia, and oxidative stress, accumulation of drusen in AMD-affected eyes is observed. Drusen contain damaged cellular proteins, lipoprotein particles, lipids and carbohydrates and they are related to impaired protein clearance, inflammation, and extracellular matrix modification. When autophagy, a major cellular proteostasis pathway, is impaired, the accumulations of intracellular lipofuscin and extracellular drusen are detected. As these aggregates grow over time, they finally cause the disorganisation and destruction of the RPE and photoreceptors leading to visual loss. In this review, the role of autophagy in drusen biogenesis is discussed since impairment in removing cellular waste in RPE cells plays a key role in AMD progression. In the future, means which improve intracellular clearance might be of use in AMD therapy to slow the progression of drusen formation.
Collapse
Affiliation(s)
- Juha M T Hyttinen
- Department of Ophthalmology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Ali Koskela
- Department of Ophthalmology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Janusz Blasiak
- Faculty of Medicine, Collegium Medicum, Mazovian Academy in Plock, Plock, Poland
| | - Kai Kaarniranta
- Department of Ophthalmology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
- Department of Molecular Genetics, University of Lodz, Lodz, Poland
- Department of Ophthalmology, Kuopio University Hospital, Kuopio, Finland
| |
Collapse
|
6
|
de Oliveira Figueiredo EC, Bucolo C, Eandi CM. Therapeutic innovations for geographic atrophy: A promising horizon. Curr Opin Pharmacol 2024; 78:102484. [PMID: 39243634 DOI: 10.1016/j.coph.2024.102484] [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: 11/28/2023] [Revised: 05/24/2024] [Accepted: 08/12/2024] [Indexed: 09/09/2024]
Abstract
This mini review spotlights the most promising treatments for geographic atrophy, the advanced form of age-related macular degeneration, often resulting in severe and irreversible vision loss. The pathophysiology is complex, and various therapeutic strategies, including anticomplement therapies, gene therapies, cell-based interventions, and artificial intelligence-driven diagnostics are discussed. Anticomplement therapies (antifactors C3 and C5) showed promise in reducing the inflammatory response and the progression of the atrophy. Gene therapies, targeting specific genetic mutations, are under development to correct underlying defects and potentially reverse disease progression. Cell-based therapies are gaining momentum, with early studies indicating encouraging results in the replacement of damaged retinal pigment epithelium cells.
Collapse
Affiliation(s)
| | - Claudio Bucolo
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Catania, Italy
| | - Chiara M Eandi
- Hôpital Ophtalmique Jules-Gonin, Fondation Asile des Aveugles, Lausanne, Switzerland; Department of Surgical Science, University of Torino, Torino, Italy.
| |
Collapse
|
7
|
Fradot V, Augustin S, Fontaine V, Marazova K, Guillonneau X, Sahel JA, Picaud S. Rodent Models of Retinal Degeneration: From Purified Cells in Culture to Living Animals. Cold Spring Harb Perspect Med 2024; 14:a041311. [PMID: 37848250 PMCID: PMC11444255 DOI: 10.1101/cshperspect.a041311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2023]
Abstract
Rodent models of retinal degeneration are essential for the development of therapeutic strategies. In addition to living animal models, we here also discuss models based on rodent cell cultures, such as purified retinal ganglion cells and retinal explants. These ex vivo models extend the possibilities for investigating pathological mechanisms and assessing the neuroprotective effect of pharmacological agents by eliminating questions on drug pharmacokinetics and bioavailability. The number of living rodent models has greatly increased with the possibilities to achieve transgenic modifications in animals for knocking in and out genes and mutations. The Cre-lox system has further enabled investigators to target specific genes or mutations in specific cells at specific stages. However, chemically or physically induced models can provide alternatives to such targeted gene modifications. The increased diversity of rodent models has widened our possibility to address most ocular pathologies for providing initial proof of concept of innovative therapeutic strategies.
Collapse
Affiliation(s)
- Valérie Fradot
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris F-75012, France
| | - Sébastien Augustin
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris F-75012, France
| | - Valérie Fontaine
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris F-75012, France
| | - Katia Marazova
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris F-75012, France
| | - Xavier Guillonneau
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris F-75012, France
| | - José A Sahel
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris F-75012, France
- Department of Ophthalmology, The University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213, USA
| | - Serge Picaud
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris F-75012, France
| |
Collapse
|
8
|
Connolly E, El-Farouki G, Brennan K, Cahill M, Doyle SL. Poor Response to Bevacizumab Correlates With Higher IL-6 and IL-8 Aqueous Cytokines in AMD. Invest Ophthalmol Vis Sci 2024; 65:37. [PMID: 39325472 PMCID: PMC11437685 DOI: 10.1167/iovs.65.11.37] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2024] Open
Abstract
Purpose To evaluate the effect of intravitreal bevacizumab on aqueous levels of a panel of 12 inflammatory cytokines in patients with neovascular age-related macular degeneration (nAMD) and correlate response to treatment, as measured by change in the central subfovea thickness (CST), with cytokine levels. Methods Thirty-three treatment-naïve patients with nAMD received a loading dose of intravitreal bevacizumab consisting of three injections at six weekly intervals. The aqueous samples prior to the first (baseline), second (week 6), and third (week 12) injections were analyzed for cytokine levels. Participants were subgrouped based on changes in CST on spectral-domain optical coherence tomography (SD-OCT) at 12 weeks. Group 1 included patients with a decrease in CST (responders; n = 27). Group 2 included patients who had no decrease in CST (poor responders; n = 6). Results Aqueous IL-8 was the only cytokine to demonstrate a significant difference in levels between responders and poor responders, with higher interleukin-8 (IL-8) at week 12 in the poor responder group. Aqueous IL-6 and IL-8 levels showed a positive correlation with CST on SD-OCT (Spearman r = 0.45 and 0.55, respectively). There was a temporal increase overall in cytokine concentration accompanying bevacizumab treatment. Conclusions Aqueous IL-6 and IL-8 may be important markers of treatment response or poor response in nAMD. Future therapeutic strategies may include targeted treatment against both vascular endothelial cell growth factor (VEGF) and IL-6 and/or IL-8 in patients who do not respond to anti-VEGF treatment alone.
Collapse
Affiliation(s)
- Emma Connolly
- Department of Clinical Medicine, School of Medicine, Trinity College Dublin, Dublin, Ireland
- Trinity College Institute of Neuroscience, Trinity College Dublin, Ireland
| | - Ghaleb El-Farouki
- Department of Clinical Medicine, School of Medicine, Trinity College Dublin, Dublin, Ireland
- Research Foundation, Royal Victoria Eye and Ear Hospital, Dublin, Ireland
| | - Kiva Brennan
- Department of Clinical Medicine, School of Medicine, Trinity College Dublin, Dublin, Ireland
- Trinity College Institute of Neuroscience, Trinity College Dublin, Ireland
| | - Mark Cahill
- Research Foundation, Royal Victoria Eye and Ear Hospital, Dublin, Ireland
- Progressive Vision Research, Sandyford, Dublin, Ireland
| | - Sarah L Doyle
- Department of Clinical Medicine, School of Medicine, Trinity College Dublin, Dublin, Ireland
- Trinity College Institute of Neuroscience, Trinity College Dublin, Ireland
| |
Collapse
|
9
|
Rui Y, Zhang M, Lee DM, Snyder VC, Raghuraman R, Gofas-Salas E, Mecê P, Yadav S, Tiruveedhula P, Grieve K, Sahel JA, Errera MH, Rossi EA. Label-Free Imaging of Inflammation at the Level of Single Cells in the Living Human Eye. OPHTHALMOLOGY SCIENCE 2024; 4:100475. [PMID: 38881602 PMCID: PMC11179426 DOI: 10.1016/j.xops.2024.100475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 12/22/2023] [Accepted: 01/16/2024] [Indexed: 06/18/2024]
Abstract
Purpose Putative microglia were recently detected using adaptive optics ophthalmoscopy in healthy eyes. Here we evaluate the use of nonconfocal adaptive optics scanning light ophthalmoscopy (AOSLO) for quantifying the morphology and motility of presumed microglia and other immune cells in eyes with retinal inflammation from uveitis and healthy eyes. Design Observational exploratory study. Participants Twelve participants were imaged, including 8 healthy participants and 4 posterior uveitis patients recruited from the clinic of 1 of the authors (M.H.E.). Methods The Pittsburgh AOSLO imaging system was used with a custom-designed 7-fiber optical fiber bundle for simultaneous confocal and nonconfocal multioffset detection. The inner retina was imaged at several locations at multiple timepoints in healthy participants and uveitis patients to generate time-lapse images. Main Outcome Measures Microglia and macrophages were manually segmented from nonconfocal AOSLO images, and their morphological characteristics quantified (including soma size, diameter, and circularity). Cell soma motion was quantified across time for periods of up to 30 minutes and their speeds were calculated by measuring their displacement over time. Results A spectrum of cell morphologies was detected in healthy eyes from circular amoeboid cells to elongated cells with visible processes, resembling activated and ramified microglia, respectively. Average soma diameter was 16.1 ± 0.9 μm. Cell movement was slow in healthy eyes (0.02 μm/sec on average), but macrophage-like cells moved rapidly in some uveitis patients (up to 3 μm/sec). In an eye with infectious uveitis, many macrophage-like cells were detected; during treatment their quantity and motility decreased as vision improved. Conclusions In vivo adaptive optics ophthalmoscopy offers promise as a potentially powerful tool for detecting and monitoring inflammation and response to treatment at a cellular level in the living eye. Financial Disclosures Proprietary or commercial disclosure may be found in the Footnotes and Disclosures at the end of this article.
Collapse
Affiliation(s)
- Yuhua Rui
- Department of Ophthalmology, University of Pittsburgh School of Medicine Pittsburgh, Pennsylvania
- Eye Center of Xiangya Hospital, Central South University Hunan Key Laboratory of Ophthalmology Changsha, Hunan, China
| | - Min Zhang
- Department of Ophthalmology, University of Pittsburgh School of Medicine Pittsburgh, Pennsylvania
| | - Daniel M.W. Lee
- Department of Bioengineering, University of Pittsburgh Swanson School of Engineering Pittsburgh, Pennsylvania
| | - Valerie C. Snyder
- Department of Ophthalmology, University of Pittsburgh School of Medicine Pittsburgh, Pennsylvania
| | - Rashmi Raghuraman
- Department of Ophthalmology, University of Pittsburgh School of Medicine Pittsburgh, Pennsylvania
| | - Elena Gofas-Salas
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
- CHNO des Quinze-Vingts, INSERM-DGOS CIC 1423, Paris, France
| | - Pedro Mecê
- Institut Langevin, ESPCI Paris, Université PSL, CNRS, Paris, France
| | - Sanya Yadav
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | | | - Kate Grieve
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
- CHNO des Quinze-Vingts, INSERM-DGOS CIC 1423, Paris, France
| | - José-Alain Sahel
- Department of Ophthalmology, University of Pittsburgh School of Medicine Pittsburgh, Pennsylvania
| | - Marie-Hélène Errera
- Department of Ophthalmology, University of Pittsburgh School of Medicine Pittsburgh, Pennsylvania
| | - Ethan A. Rossi
- Department of Ophthalmology, University of Pittsburgh School of Medicine Pittsburgh, Pennsylvania
- Department of Bioengineering, University of Pittsburgh Swanson School of Engineering Pittsburgh, Pennsylvania
- McGowan Institute for Regenerative Medicine Pittsburgh, Pennsylvania
| |
Collapse
|
10
|
Zhou L, Xu Z, Lu H, Cho H, Xie Y, Lee G, Ri K, Duh EJ. Suppression of inner blood-retinal barrier breakdown and pathogenic Müller glia activation in ischemia retinopathy by myeloid cell depletion. J Neuroinflammation 2024; 21:210. [PMID: 39182142 PMCID: PMC11344463 DOI: 10.1186/s12974-024-03190-9] [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: 05/16/2024] [Accepted: 07/29/2024] [Indexed: 08/27/2024] Open
Abstract
Ischemic retinopathies including diabetic retinopathy are major causes of vision loss. Inner blood-retinal barrier (BRB) breakdown with retinal vascular hyperpermeability results in macular edema. Although dysfunction of the neurovascular unit including neurons, glia, and vascular cells is now understood to underlie this process, there is a need for fuller elucidation of the underlying events in BRB dysfunction in ischemic disease, including a systematic analysis of myeloid cells and exploration of cellular cross-talk. We used an approach for microglia depletion with the CSF-1R inhibitor PLX5622 (PLX) in the retinal ischemia-reperfusion (IR) model. Under non-IR conditions, PLX treatment successfully depleted microglia in the retina. PLX suppressed the microglial activation response following IR as well as infiltration of monocyte-derived macrophages. This occurred in association with reduction of retinal expression of chemokines including CCL2 and the inflammatory adhesion molecule ICAM-1. In addition, there was a marked suppression of retinal neuroinflammation with reduction in expression of IL-1b, IL-6, Ptgs2, TNF-a, and Angpt2, a protein that regulates BRB permeability. PLX treatment significantly suppressed inner BRB breakdown following IR, without an appreciable effect on neuronal dysfunction. A translatomic analysis of Müller glial-specific gene expression in vivo using the Ribotag approach demonstrated a strong suppression of Müller cell expression of multiple pro-inflammatory genes following PLX treatment. Co-culture studies of Müller cells and microglia demonstrated that activated microglia directly upregulates Müller cell-expression of these inflammatory genes, indicating Müller cells as a downstream effector of myeloid cells in retinal IR. Co-culture studies of these two cell types with endothelial cells demonstrated the ability of both activated microglia and Müller cells to compromise EC barrier function. Interestingly, quiescent Müller cells enhanced EC barrier function in this co-culture system. Together this demonstrates a pivotal role for myeloid cells in inner BRB breakdown in the setting of ischemia-associated disease and indicates that myeloid cells play a major role in iBRB dysregulation, through direct and indirect effects, while Müller glia participate in amplifying the neuroinflammatory effect of myeloid cells.
Collapse
Affiliation(s)
- Lingli Zhou
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou, China
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Zhenhua Xu
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Haining Lu
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hongkwan Cho
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yangyiran Xie
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Grace Lee
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kaoru Ri
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Elia J Duh
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
11
|
Song SY, Park DH, Lee SH, Lim HK, Park JW, Seo JW, Cho SS. Protective Effects of 7S,15R-Dihydroxy-16S,17S-Epoxy-Docosapentaenoic Acid (diHEP-DPA) against Blue Light-Induced Retinal Damages in A2E-Laden ARPE-19 Cells. Antioxidants (Basel) 2024; 13:982. [PMID: 39199228 PMCID: PMC11351242 DOI: 10.3390/antiox13080982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 08/09/2024] [Accepted: 08/11/2024] [Indexed: 09/01/2024] Open
Abstract
The purpose of this study was to investigate the protective effects of 7S,15R-dihydroxy-16S,17S-epoxy-docosapentaenoic acid (diHEP-DPA) in retinal pigment epithelial (RPE) cell damage. ARPE-19 cells, a human RPE cell line, were cultured with diHEP-DPA and Bis-retinoid N-retinyl-N-retinylidene ethanolamine (A2E), followed by exposure to BL. Cell viability and cell death rates were determined. Western blotting was performed to determine changes in apoptotic factors, mitogen-activated protein kinase (MAPK) family proteins, inflammatory proteins, and oxidative and carbonyl stresses. The levels of pro-inflammatory cytokines in the culture medium supernatants were also measured. Exposure to A2E and BL increased the ARPE-19 cell death rate, which was alleviated by diHEP-DPA in a concentration-dependent manner. A2E and BL treatments induced apoptosis in ARPE-19 cells, which was also alleviated by diHEP-DPA. Analysis of the relationship with MAPK proteins revealed that the expression of p-JNK and p-P38 increased after A2E and BL treatments and decreased with exposure to diHEP-DPA in a concentration-dependent manner. DiHEP-DPA also affected the inflammatory response by suppressing the expression of inflammatory proteins and the production of pro-inflammatory cytokines. Furthermore, it was shown that diHEP-DPA regulated the proteins related to oxidative and carbonyl stresses. Taken together, our results provide evidence that diHEP-DPA can inhibit cell damage caused by A2E and BL exposure at the cellular level by controlling various pathways involved in apoptosis and inflammatory responses.
Collapse
Affiliation(s)
- Seung-Yub Song
- Department of Pharmacy, College of Pharmacy, Mokpo National University, Muan 58554, Jeonnam, Republic of Korea; (S.-Y.S.); (S.-H.L.); (J.-W.P.)
- Biomedicine, Health & Life Convergence Sciences, BK21 Four, College of Pharmacy, Mokpo National University, Muan 58554, Jeonnam, Republic of Korea;
| | - Dae-Hun Park
- College of Oriental Medicine, Dongshin University, Naju-si 58245, Jeonnam, Republic of Korea;
| | - Sung-Ho Lee
- Department of Pharmacy, College of Pharmacy, Mokpo National University, Muan 58554, Jeonnam, Republic of Korea; (S.-Y.S.); (S.-H.L.); (J.-W.P.)
- Biomedicine, Health & Life Convergence Sciences, BK21 Four, College of Pharmacy, Mokpo National University, Muan 58554, Jeonnam, Republic of Korea;
| | - Han-Kyu Lim
- Biomedicine, Health & Life Convergence Sciences, BK21 Four, College of Pharmacy, Mokpo National University, Muan 58554, Jeonnam, Republic of Korea;
- Department of Marine and Fisheries Resources, Mokpo National University, Muan 58554, Jeonnam, Republic of Korea
| | - Jin-Woo Park
- Department of Pharmacy, College of Pharmacy, Mokpo National University, Muan 58554, Jeonnam, Republic of Korea; (S.-Y.S.); (S.-H.L.); (J.-W.P.)
- Biomedicine, Health & Life Convergence Sciences, BK21 Four, College of Pharmacy, Mokpo National University, Muan 58554, Jeonnam, Republic of Korea;
| | - Jeong-Woo Seo
- Microbial Biotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeongeup-si 56212, Jeollabuk-do, Republic of Korea;
| | - Seung-Sik Cho
- Department of Pharmacy, College of Pharmacy, Mokpo National University, Muan 58554, Jeonnam, Republic of Korea; (S.-Y.S.); (S.-H.L.); (J.-W.P.)
- Biomedicine, Health & Life Convergence Sciences, BK21 Four, College of Pharmacy, Mokpo National University, Muan 58554, Jeonnam, Republic of Korea;
| |
Collapse
|
12
|
Borella Y, Danielsen N, Markle EM, Snyder VC, Lee DMW, Zhang M, Eller AW, Chhablani J, Paques M, Rossi EA. Are the Hypo-Reflective Clumps Associated With Age-Related Macular Degeneration in Adaptive Optics Ophthalmoscopy Autofluorescent? Invest Ophthalmol Vis Sci 2024; 65:28. [PMID: 39167400 PMCID: PMC11343010 DOI: 10.1167/iovs.65.10.28] [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: 05/31/2024] [Accepted: 08/03/2024] [Indexed: 08/23/2024] Open
Abstract
Purpose Hypo-reflective clumps (HRCs) are structures associated with age-related macular degeneration (AMD) that were identified using flood-illumination adaptive optics ophthalmoscopy (FIAO) and hypothesized to be either macrophages that have accumulated melanin through the phagocytosis of retinal pigmented epithelial (RPE) cell organelles or transdifferentiated RPE cells. HRCs may be autofluorescent (AF) in the near infrared (NIR) but clinical NIR autofluorescence imaging lacks the resolution to answer this question definitively. Here, we used near infrared autofluorescence (NIRAF) imaging in fluorescence adaptive optics scanning laser ophthalmoscopy (AOSLO) to determine whether HRCs are AF. Methods Patients with AMD and HRCs underwent imaging with FIAO, optical coherence tomography (OCT), and multi-modal AOSLO (confocal, NIRAF, and non-confocal multi-offset detection using a fiber bundle). HRCs were segmented on FIAO and images, co-registered across modalities, and HRC morphometry and AF were quantified. Results Eight patients participated (mean age = 79 years, standard deviation [SD] = 5.7, range = 69-89 years, and 5 female patients). Most HRCs (86%, n = 153/178) were autofluorescent on AOSLO. HRC AF signal varied but most uniformly dark HRCs on FIAO showed corresponding AF on AOSLO, whereas heterogeneous HRCs showed a smaller AF area or no AF. Conclusions These findings are consistent with the hypothesis that HRCs contain AF RPE organelles. A small proportion of HRCs were not AF; these may represent macrophages that have not yet accumulated enough organelles to become AF. HRCs may have clinical significance but further study is needed to understand the interplay among HRCs, RPE cells, and macrophages, and their relationship to geographic atrophy (GA) progression in AMD.
Collapse
Affiliation(s)
- Ysé Borella
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
- Vision Institute, 15-20 National Ophthalmology Hospital, Clinical Investigation Center 1423 and Sorbonne University, Paris, France
| | - Natalie Danielsen
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
- Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, Pennsylvania, United States
| | - Evelyn M. Markle
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | - Valerie C. Snyder
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | - Daniel M. W. Lee
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
- Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, Pennsylvania, United States
| | - Min Zhang
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | - Andrew W. Eller
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | - Jay Chhablani
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | - Michel Paques
- Vision Institute, 15-20 National Ophthalmology Hospital, Clinical Investigation Center 1423 and Sorbonne University, Paris, France
| | - Ethan A. Rossi
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
- Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, Pennsylvania, United States
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| |
Collapse
|
13
|
Song MS, Kim YH, Oh J. Spatial Distribution of Hyperreflective Choroidal Foci in the Macula of Normal Eyes. Transl Vis Sci Technol 2024; 13:35. [PMID: 39172482 PMCID: PMC11346144 DOI: 10.1167/tvst.13.8.35] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 07/09/2024] [Indexed: 08/23/2024] Open
Abstract
Purpose To investigate variations in the spatial distribution of hyperreflective foci in the choroid of the macula in normal eyes. Methods We included eyes with a normal fundus from patients who had undergone optical coherence tomography angiography, covering a 6-mm × 6-mm area centered on the fovea. The macular area was divided into nine sectors according to the modified Early Treatment of Diabetic Retinopathy Study grid. Hyperreflective choroidal foci (HCF) distribution, choriocapillaris vascular density, and choroidal stromal density were determined on en face images of the choroid in each sector. Results We included 35 eyes from 35 participants, with a mean age of 52.7 ± 16.8 years. The mean number and area fraction of HCF at the 5-mm macular area were 35.6 ± 7.8 foci/mm2 and 3.0% ± 0.7%, respectively. The number of HCF in the central circle (50.7 ± 20.9 foci/mm2) was greater than that in the inner (35.1 ± 13.0 foci/mm2) or outer rings (35.6 ± 6.5 foci/mm2) (P < 0.001, P < 0.001, respectively). The area fraction of HCF in the central circle (4.84% ± 3.36%) was greater than that in the inner (2.62% ± 1.17%; P < 0.001) or outer rings (3.12% ± 0.67%; P = 0.004). The HCF distribution did not significantly correlate with the choriocapillaris vascular density or choroidal stromal density in each sector. Conclusions HCF were more densely distributed in the macular center than in the pericentral or peripheral macular areas. Translational Relevance HCF measurement and spatial distribution could provide additional information for evaluating choroidal stromal characteristics.
Collapse
Affiliation(s)
- Myung-Sun Song
- Department of Ophthalmology, Korea University College of Medicine, Seoul, Korea
| | - Young Ho Kim
- Department of Ophthalmology, Korea University College of Medicine, Seoul, Korea
| | - Jaeryung Oh
- Department of Ophthalmology, Korea University College of Medicine, Seoul, Korea
| |
Collapse
|
14
|
Hallak JA, Abbasi A, Goldberg RA, Modi Y, Zhao C, Jing Y, Chen N, Mercer D, Sahu S, Alobaidi A, López FJ, Luhrs K, Waring JF, den Hollander AI, Smaoui N. Janus Kinase Inhibitor Therapy and Risk of Age-Related Macular Degeneration in Autoimmune Disease. JAMA Ophthalmol 2024; 142:750-758. [PMID: 38990568 PMCID: PMC11240228 DOI: 10.1001/jamaophthalmol.2024.2376] [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: 02/02/2024] [Accepted: 05/08/2024] [Indexed: 07/12/2024]
Abstract
Importance The involvement of chronic inflammation in the pathogenesis of age-related macular degeneration (AMD) opens therapeutic possibilities to AMD management. Objective To determine whether Janus kinase inhibitors (JAKis) are associated with a reduced risk of AMD development in patients with autoimmune diseases. Design, Setting, and Participants This retrospective observational cohort study used administrative claims data from Merative MarketScan research databases (Commercial and Medicare Supplemental) and Optum Clinformatics Data Mart databases between January 1, 2010, and January 31, 2022. Patients with autoimmune diseases satisfying study eligibility criteria and who received JAKi treatment (9126 in MarketScan and 5667 in Optum) were propensity score matched (1:1) to identical numbers of study-eligible patients who received non-JAKi-based immunotherapy. Exposure Treatment duration of 6 months or longer. Main Outcomes and Measures Incidence rates of AMD (exudative and nonexudative) over the first 6 to 18 months of treatment were determined, and bayesian Poisson regression models were used to estimate incidence rate ratios, 95% CIs, and posterior probabilities of AMD. Results After matching, female sex represented the majority of the patient population in both MarketScan and Optum (14 019/18 252 [76.6%] and 8563/3364 [75.2%], respectively in the JAKi patient population). More than 60% of the patient population was older than 55 years of age in both cohorts. Over the specified treatment period, a 49% relative reduction in incidence of AMD was observed among patients who received JAKi therapy (10/9126 events; adjusted incidence rate ratio [AIRR], 0.51; 95% CI, 0.19-0.90) vs those who received non-JAKi therapy (43/9126 events; AIRR, 1 [reference]) in MarketScan, and a 73% relative reduction in incidence of AMD was observed among patients who received JAKi therapy (3/5667 events; AIRR, 0.27; 95% CI, 0.03-0.74) vs those who received non-JAKi therapy (21/5667 events; AIRR, 1 [reference]) in Optum. The absolute percentage reductions were 0.36% (MarketScan) and 0.32% (Optum), favoring patients who received JAKi therapy. Posterior probabilities of the adjusted risk being less than unity were 97.6% (MarketScan) and 98.9% (Optum) for those who received JAKi therapy vs those who received non-JAKi therapy in MarketScan and Optum, respectively. Conclusions and Relevance JAKi use may be associated with a reduced risk of incident AMD in US adults with major autoimmune diseases. The absolute percentage reduction is consistent with a potential role for JAKi in this population. Future studies with long-term follow-up are recommended to investigate the association between JAKi use and incident AMD in other disease indications. Investigation into the role of systemic inflammation and JAK-signal transducers and activators of transcription signaling in AMD may improve understanding of the pathophysiology of AMD and lead to new treatment options.
Collapse
Affiliation(s)
- Joelle A. Hallak
- Health Economics and Outcomes Research, AbbVie, North Chicago, Illinois
| | - Ali Abbasi
- Genomics Research Center, AbbVie, North Chicago, Illinois
- Currently with University of Groningen, Groningen, the Netherlands
| | | | - Yasha Modi
- New York University Langone Health, New York
| | - Changgeng Zhao
- Health Economics and Outcomes Research, AbbVie, North Chicago, Illinois
| | - Yonghua Jing
- Health Economics and Outcomes Research, AbbVie, North Chicago, Illinois
| | - Naijun Chen
- Health Economics and Outcomes Research, AbbVie, North Chicago, Illinois
| | - Daniel Mercer
- Genesis Research Group, Hoboken, New Jersey
- Currently with Genesis Research Group, Hoboken, New Jersey
| | - Soumya Sahu
- Health Economics and Outcomes Research, AbbVie, North Chicago, Illinois
| | - Ali Alobaidi
- Health Economics and Outcomes Research, AbbVie, North Chicago, Illinois
| | | | - Keith Luhrs
- Ophthalmology Discovery Research, AbbVie, Irvine, California
- Currently with Bausch + Lomb, Irvine, California
| | | | | | - Nizar Smaoui
- Genomics Research Center, AbbVie, North Chicago, Illinois
| |
Collapse
|
15
|
Tarallo V, Magliacane Trotta S, Panico S, D'Orsi L, Mercadante G, Cicatiello V, De Falco S. PlGF and VEGF-A/PlGF Heterodimer are Crucial for Recruitment and Activation of Immune Cells During Choroid Neovascularization. Invest Ophthalmol Vis Sci 2024; 65:12. [PMID: 38967942 PMCID: PMC11232896 DOI: 10.1167/iovs.65.8.12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 06/06/2024] [Indexed: 07/06/2024] Open
Abstract
Purpose Recruitment and activation of inflammatory cells, such as retinal microglia/macrophages, in the subretinal space contribute significantly to the pathogenesis of age-related macular degeneration (AMD). This study aims to explore the functional role of vascular endothelial growth factor (VEGF-A), placental growth factor (PlGF) and VEGF-A/PlGF heterodimer in immune homeostasis and activation during pathological laser-induced choroidal neovascularization (CNV). Methods To investigate these roles, we utilized the PlGF-DE knockin (KI) mouse model, which is the full functional knockout (KO) of PlGF. In this model, mice express a variant of PlGF, named PlGF-DE, that is unable to bind and activate VEGFR-1 but can still form heterodimer with VEGF-A. Results Our findings demonstrate that, although there is no difference in healthy conditions, PlGF-DE-KI mice exhibit decreased microglia reactivity and reduced recruitment of both microglia and monocyte-macrophages, compared to wild-type mice during laser-induced CNV. This impairment is associated with a reduction in VEGF receptor 1 (VEGFR-1) phosphorylation in the retinae of PlGF-DE-KI mice compared to C57Bl6/J mice. Corroborating these data, intravitreal delivery of PlGF or VEGF-A/PlGF heterodimer in PlGF-DE-KI mice rescued the immune cell response at the early phase of CNV compared to VEGF-A delivery. Conclusions In summary, our study suggests that targeting PlGF and the VEGF-A/PlGF heterodimer, thereby preventing VEGFR-1 activation, could represent a potential therapeutic approach for the management of inflammatory processes in diseases such as AMD.
Collapse
Affiliation(s)
- Valeria Tarallo
- Angiogenesis Lab, Institute of Genetics and Biophysics ‘Adriano Buzzati-Traverso’ - CNR, Naples, Italy
| | - Sara Magliacane Trotta
- Angiogenesis Lab, Institute of Genetics and Biophysics ‘Adriano Buzzati-Traverso’ - CNR, Naples, Italy
| | - Sonia Panico
- Angiogenesis Lab, Institute of Genetics and Biophysics ‘Adriano Buzzati-Traverso’ - CNR, Naples, Italy
| | - Luca D'Orsi
- Angiogenesis Lab, Institute of Genetics and Biophysics ‘Adriano Buzzati-Traverso’ - CNR, Naples, Italy
- BIOVIIIx srl, Via Alessandro Manzoni 1, Napoli, Italy
| | - Grazia Mercadante
- Angiogenesis Lab, Institute of Genetics and Biophysics ‘Adriano Buzzati-Traverso’ - CNR, Naples, Italy
| | - Valeria Cicatiello
- Angiogenesis Lab, Institute of Genetics and Biophysics ‘Adriano Buzzati-Traverso’ - CNR, Naples, Italy
| | - Sandro De Falco
- Angiogenesis Lab, Institute of Genetics and Biophysics ‘Adriano Buzzati-Traverso’ - CNR, Naples, Italy
- BIOVIIIx srl, Via Alessandro Manzoni 1, Napoli, Italy
- AnBition srl, Via Alessandro Manzoni 1, Napoli, Italy
| |
Collapse
|
16
|
Tong M, Bai Y, Han X, Kong L, Ren L, Zhang L, Li X, Yao J, Yan B. Single-cell profiling transcriptomic reveals cellular heterogeneity and cellular crosstalk in choroidal neovascularization model. Exp Eye Res 2024; 242:109877. [PMID: 38537669 DOI: 10.1016/j.exer.2024.109877] [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: 10/14/2023] [Revised: 03/18/2024] [Accepted: 03/21/2024] [Indexed: 04/01/2024]
Abstract
Choroidal neovascularization (CNV) is a hallmark of neovascular age-related macular degeneration (nAMD) and a major contributor to vision loss in nAMD cases. However, the identification of specific cell types associated with nAMD remains challenging. Herein, we performed single-cell sequencing to comprehensively explore the cellular diversity and understand the foundational components of the retinal pigment epithelium (RPE)/choroid complex. We unveiled 10 distinct cell types within the RPE/choroid complex. Notably, we observed significant heterogeneity within endothelial cells (ECs), fibroblasts, and macrophages, underscoring the intricate nature of the cellular composition in the RPE/choroid complex. Within the EC category, four distinct clusters were identified and EC cluster 0 was tightly associated with choroidal neovascularization. We identified five clusters of fibroblasts actively involved in the pathogenesis of nAMD, influencing fibrotic responses, angiogenic effects, and photoreceptor function. Additionally, three clusters of macrophages were identified, suggesting their potential roles in regulating the progression of nAMD through immunomodulation and inflammation regulation. Through CellChat analysis, we constructed a complex cell-cell communication network, revealing the role of EC clusters in interacting with fibroblasts and macrophages in the context of nAMD. These interactions were found to govern angiogenic effects, fibrotic responses, and inflammatory processes. In summary, this study reveals noteworthy cellular heterogeneity in the RPE/choroid complex and provides valuable insights into the pathogenesis of CNV. These findings will open up potential avenues for deep understanding and targeted therapeutic interventions in nAMD.
Collapse
Affiliation(s)
- Ming Tong
- Eye Institute and Department of Ophthalmology, Eye and ENT Hospital, Fudan University, Shanghai, 200031, China
| | - Yun Bai
- College of Information Science, Shanghai Ocean University, Shanghai, 201306, China
| | - Xiaoyan Han
- Eye Institute and Department of Ophthalmology, Eye and ENT Hospital, Fudan University, Shanghai, 200031, China
| | - Lingjie Kong
- Eye Institute and Department of Ophthalmology, Eye and ENT Hospital, Fudan University, Shanghai, 200031, China
| | - Ling Ren
- Eye Institute and Department of Ophthalmology, Eye and ENT Hospital, Fudan University, Shanghai, 200031, China
| | - Linyu Zhang
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, 210000, China; The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, 210000, China
| | - Xiumiao Li
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, 210000, China
| | - Jin Yao
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, 210000, China; The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, 210000, China.
| | - Biao Yan
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China.
| |
Collapse
|
17
|
Keenan TDL, Bailey C, Abraham M, Orndahl C, Menezes S, Bellur S, Arunachalam T, Kangale-Whitney C, Srinivas S, Karamat A, Nittala M, Cunningham D, Jeffrey BG, Wiley HE, Thavikulwat AT, Sadda S, Cukras CA, Chew EY, Wong WT. Phase 2 Trial Evaluating Minocycline for Geographic Atrophy in Age-Related Macular Degeneration: A Nonrandomized Controlled Trial. JAMA Ophthalmol 2024; 142:345-355. [PMID: 38483382 PMCID: PMC10941022 DOI: 10.1001/jamaophthalmol.2024.0118] [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: 10/26/2023] [Accepted: 12/20/2023] [Indexed: 03/17/2024]
Abstract
Importance Existing therapies to slow geographic atrophy (GA) enlargement in age-related macular degeneration (AMD) have relatively modest anatomic efficacy, require intravitreal administration, and increase the risk of neovascular AMD. Additional therapeutic approaches are desirable. Objective To evaluate the safety and possible anatomic efficacy of oral minocycline, a microglial inhibitor, for the treatment of GA in AMD. Design, Setting, and Participants This was a phase 2, prospective, single-arm, 45-month, nonrandomized controlled trial conducted from December 2016 to April 2023. Patients with GA from AMD in 1 or both eyes were recruited from the National Institutes of Health (Bethesda, Maryland) and Bristol Eye Hospital (Bristol, UK). Study data were analyzed from September 2022 to May 2023. Intervention After a 9-month run-in phase, participants began oral minocycline, 100 mg, twice daily for 3 years. Main Outcomes and Measures The primary outcome measure was the difference in rate of change of square root GA area on fundus autofluorescence between the 24-month treatment phase and 9-month run-in phase. Results Of the 37 participants enrolled (mean [SD] age, 74.3 [7.6] years; 21 female [57%]), 36 initiated the treatment phase. Of these participants, 21 (58%) completed at least 33 months, whereas 15 discontinued treatment (8 by request, 6 for adverse events/illness, and 1 death). Mean (SE) square root GA enlargement rate in study eyes was 0.31 (0.03) mm per year during the run-in phase and 0.28 (0.02) mm per year during the treatment phase. The primary outcome measure of mean (SE) difference in enlargement rates between the 2 phases was -0.03 (0.03) mm per year (P = .39). Similarly, secondary outcome measures of GA enlargement rate showed no differences between the 2 phases. The secondary outcome measures of mean difference in rate of change between 2 phases were 0.2 letter score per month (95% CI, -0.4 to 0.9; P = .44) for visual acuity and 0.7 μm per month (-0.4 to 1.8; P = .20) for subfoveal retinal thickness. Of the 129 treatment-emergent adverse events among 32 participants, 49 (38%) were related to minocycline (with no severe or ocular events), including elevated thyrotropin level (15 participants) and skin hyperpigmentation/discoloration (8 participants). Conclusions and Relevance In this phase 2 nonrandomized controlled trial, oral minocycline was not associated with a decrease in GA enlargement over 24 months, compared with the run-in phase. This observation was consistent across primary and secondary outcome measures. Oral minocycline at this dose is likely not associated with slower rate of enlargement of GA in AMD.
Collapse
Affiliation(s)
| | | | | | | | | | - Sunil Bellur
- National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | | | | | | | | | | | - Denise Cunningham
- National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Brett G. Jeffrey
- National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Henry E. Wiley
- National Eye Institute, National Institutes of Health, Bethesda, Maryland
- Now with Genentech Inc, South San Francisco, California
| | | | - SriniVas Sadda
- Doheny Eye Institute, Pasadena, California
- University of California, Los Angeles, Los Angeles
| | | | - Emily Y. Chew
- National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Wai T. Wong
- National Eye Institute, National Institutes of Health, Bethesda, Maryland
- Now with Janssen Research and Development LLC, Brisbane, California
| |
Collapse
|
18
|
Brandli A, Vessey KA, Fletcher EL. The contribution of pattern recognition receptor signalling in the development of age related macular degeneration: the role of toll-like-receptors and the NLRP3-inflammasome. J Neuroinflammation 2024; 21:64. [PMID: 38443987 PMCID: PMC10913318 DOI: 10.1186/s12974-024-03055-1] [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: 11/16/2023] [Accepted: 02/26/2024] [Indexed: 03/07/2024] Open
Abstract
Age-related macular degeneration (AMD) is a leading cause of irreversible vision loss, characterised by the dysfunction and death of the photoreceptors and retinal pigment epithelium (RPE). Innate immune cell activation and accompanying para-inflammation have been suggested to contribute to the pathogenesis of AMD, although the exact mechanism(s) and signalling pathways remain elusive. Pattern recognition receptors (PRRs) are essential activators of the innate immune system and drivers of para-inflammation. Of these PRRs, the two most prominent are (1) Toll-like receptors (TLR) and (2) NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3)-inflammasome have been found to modulate the progression of AMD. Mutations in TLR2 have been found to be associated with an increased risk of developing AMD. In animal models of AMD, inhibition of TLR and NLRP3 has been shown to reduce RPE cell death, inflammation and angiogenesis signalling, offering potential novel treatments for advanced AMD. Here, we examine the evidence for PRRs, TLRs2/3/4, and NLRP3-inflammasome pathways in macular degeneration pathogenesis.
Collapse
Affiliation(s)
- Alice Brandli
- Department of Anatomy and Physiology, The University of Melbourne, Grattan St, Parkville, Victoria, 3010, Australia
- Roche Pharma Research and Early Development, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Kirstan A Vessey
- Department of Anatomy and Physiology, The University of Melbourne, Grattan St, Parkville, Victoria, 3010, Australia
| | - Erica L Fletcher
- Department of Anatomy and Physiology, The University of Melbourne, Grattan St, Parkville, Victoria, 3010, Australia.
| |
Collapse
|
19
|
Lam S, Lindsey J, Carranza Leon BG, Takkouche S. Shedding light on eye disease in obesity: A review. Clin Obes 2024; 14:e12616. [PMID: 37532290 DOI: 10.1111/cob.12616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 06/24/2023] [Accepted: 07/16/2023] [Indexed: 08/04/2023]
Abstract
Obesity is known to be associated with numerous ocular manifestations, including but not limited to, diabetic retinopathy (DR), age-related macular degeneration (AMD), cataracts, glaucoma, and dry eye disease. This review aims to provide an overview of the ophthalmological findings in obesity. A literature search was conducted using PubMed and Cochrane databases for studies describing randomized clinical trials, meta-analyses, systematic reviews, and observational studies published from 1 January 2017 to 1 April 2023. The search terms used included relevant keywords such as 'obesity', 'body mass index', 'waist-to-hip ratio', 'bariatric', 'ophthalmology', 'eye disease', 'myopia', 'retinopathy', 'glaucoma', and 'cataract'. This literature search was performed on 1 April 2023. Obesity is associated with increased risk of developing DR, a sight-threatening complication of diabetes mellitus. Similarly, obesity has been shown to increase risk of AMD, cataracts, glaucoma, and ocular surface disease. Multiple mechanisms linking obesity to ophthalmic disease have been proposed. Adipose tissue produces various inflammatory cytokines that can affect ocular tissues, leading to disease progression. Additionally, obesity is associated with systemic metabolic changes that can influence ocular health. Bariatric surgery has been shown to be protective against development of ophthalmic disease. Obesity is a significant risk factor for several ophthalmological diseases. Healthcare providers should encourage weight loss in patients with overweight or obesity to prevent or delay the onset of ocular complications. Further research is needed to better understand the underlying mechanisms of this association, and to identify effective strategies for preventing or managing ophthalmic disease in patients with obesity.
Collapse
Affiliation(s)
- Shravika Lam
- Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Jennifer Lindsey
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute, Nashville, Tennessee, USA
| | | | - Sahar Takkouche
- Division of Diabetes, Endocrinology and Metabolism, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
20
|
Desmettre T, Baillif S, Mathis T, Gatinel D, Mainster M. [Blue light and intraocular lenses (IOLs): Beliefs and realities]. J Fr Ophtalmol 2024; 47:104043. [PMID: 38241770 DOI: 10.1016/j.jfo.2023.104043] [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: 09/26/2023] [Revised: 10/18/2023] [Accepted: 10/22/2023] [Indexed: 01/21/2024]
Abstract
The first intraocular lenses (IOLs) used for cataract surgery transmitted both ultraviolet (UV) radiation and visible light to the retina. Colorless UV-blocking IOLs were introduced and rapidly adopted in the 1980s. Yellow-tinted blue-blocking (also known as blue-filtering) IOLs were marketed in the early 1990s. Blue-blocking IOLs were intended to simulate age-related crystalline lens yellowing to reduce the cyanopsia that some patients experienced after cataract surgery. When blue-filtering IOLs were introduced in North America, however, blue-blocking chromophores were advocated as a way to protect patients from age-related macular degeneration (AMD) despite the lack of evidence that normal environmental light exposure causes AMD. The "blue light hazard" is a term that describes the experimental finding that acute, abnormally intense light exposures are potentially more phototoxic to the retina when short rather than long wavelengths are used. Thus, in brief exposures to intense light sources such as welding arcs, ultraviolet radiation is more hazardous than blue light, which is more hazardous than longer wavelength green or red light. International commissions have cautioned that the blue light hazard does not apply to normal indoor or outdoor light exposures. Nonetheless, the hazard is used for commercial purposes to suggest misleadingly that ambient environmental light can cause acute retinal phototoxicity and increase the risk of AMD. Very large epidemiological studies show that blue-blocking IOLs do not reduce the risk or progression of AMD. Additionally, blue-filtering IOLs or spectacles cannot decrease glare disability, because they decrease image and glare illuminance in the same proportion. Blue light is essential for older adults' scotopic photoreception needed to reduce the risk of nighttime falling and related injuries. It is also critical for circadian photoreception that is essential for good health, sleep and cognitive performance. Unfortunately, age-related pupillary miosis, retinal rod and ganglion cell photoreceptor degeneration and decreased outdoor activity all reduce the amount of healthful blue light available to older adults. Blue-restricting IOLs further reduce the available blue light at a time when older adults need it most. Patients and ophthalmologists are exposed to hypothesis-based advertisements for blue-filtering optical devices that suppress short wavelength light critical for vision in dim lighting and for good physical and mental health. Spectacle and intraocular lens selections should be based on scientific fact, not conjecture. Ideal IOLs should improve photoreception rather than limit it permanently. Practice efficiency, surgical convenience and physician-manufacturer relationships may eliminate a patient's opportunity to choose between colorless blue-transmitting IOLs and yellow-tinted, blue-restricting IOLs. Cataract surgeons ultimately determine whether their patients have the opportunity to make an informed choice about their future photoreception.
Collapse
Affiliation(s)
- T Desmettre
- Centre de rétine médicale, 187, rue de Menin, 59520 Marquette-Lez-Lille, France.
| | - S Baillif
- Département d'ophtalmologie, hôpital Pasteur, 30, voie Romaine, 06000 Nice cedex 1, France
| | - T Mathis
- Service d'ophtalmologie, hôpital de la Croix-Rousse, hospices civils de Lyon, 69004 Lyon, France
| | - D Gatinel
- Service d'ophtalmologie, fondation A.-de-Rothschild, 25, rue Manin, 75940 Paris cedex 19, France
| | - M Mainster
- Department of Ophthalmology, University of Kansas School of Medicine, Prairie Village, Kansas, États-Unis
| |
Collapse
|
21
|
Fernández-Albarral JA, Ramírez AI, de Hoz R, Matamoros JA, Salobrar-García E, Elvira-Hurtado L, López-Cuenca I, Sánchez-Puebla L, Salazar JJ, Ramírez JM. Glaucoma: from pathogenic mechanisms to retinal glial cell response to damage. Front Cell Neurosci 2024; 18:1354569. [PMID: 38333055 PMCID: PMC10850296 DOI: 10.3389/fncel.2024.1354569] [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: 12/12/2023] [Accepted: 01/10/2024] [Indexed: 02/10/2024] Open
Abstract
Glaucoma is a neurodegenerative disease of the retina characterized by the irreversible loss of retinal ganglion cells (RGCs) leading to visual loss. Degeneration of RGCs and loss of their axons, as well as damage and remodeling of the lamina cribrosa are the main events in the pathogenesis of glaucoma. Different molecular pathways are involved in RGC death, which are triggered and exacerbated as a consequence of a number of risk factors such as elevated intraocular pressure (IOP), age, ocular biomechanics, or low ocular perfusion pressure. Increased IOP is one of the most important risk factors associated with this pathology and the only one for which treatment is currently available, nevertheless, on many cases the progression of the disease continues, despite IOP control. Thus, the IOP elevation is not the only trigger of glaucomatous damage, showing the evidence that other factors can induce RGCs death in this pathology, would be involved in the advance of glaucomatous neurodegeneration. The underlying mechanisms driving the neurodegenerative process in glaucoma include ischemia/hypoxia, mitochondrial dysfunction, oxidative stress and neuroinflammation. In glaucoma, like as other neurodegenerative disorders, the immune system is involved and immunoregulation is conducted mainly by glial cells, microglia, astrocytes, and Müller cells. The increase in IOP produces the activation of glial cells in the retinal tissue. Chronic activation of glial cells in glaucoma may provoke a proinflammatory state at the retinal level inducing blood retinal barrier disruption and RGCs death. The modulation of the immune response in glaucoma as well as the activation of glial cells constitute an interesting new approach in the treatment of glaucoma.
Collapse
Affiliation(s)
- Jose A. Fernández-Albarral
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid (UCM), Grupo UCM 920105, IdISSC, Madrid, Spain
| | - Ana I. Ramírez
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid (UCM), Grupo UCM 920105, IdISSC, Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, Madrid, Spain
| | - Rosa de Hoz
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid (UCM), Grupo UCM 920105, IdISSC, Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, Madrid, Spain
| | - José A. Matamoros
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid (UCM), Grupo UCM 920105, IdISSC, Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, Madrid, Spain
| | - Elena Salobrar-García
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid (UCM), Grupo UCM 920105, IdISSC, Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, Madrid, Spain
| | - Lorena Elvira-Hurtado
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid (UCM), Grupo UCM 920105, IdISSC, Madrid, Spain
| | - Inés López-Cuenca
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid (UCM), Grupo UCM 920105, IdISSC, Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, Madrid, Spain
| | - Lidia Sánchez-Puebla
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid (UCM), Grupo UCM 920105, IdISSC, Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University of Madrid, Madrid, Spain
| | - Juan J. Salazar
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid (UCM), Grupo UCM 920105, IdISSC, Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, Madrid, Spain
| | - José M. Ramírez
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid (UCM), Grupo UCM 920105, IdISSC, Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University of Madrid, Madrid, Spain
| |
Collapse
|
22
|
Roubeix C, Nous C, Augustin S, Ronning KE, Mathis T, Blond F, Lagouge-Roussey P, Crespo-Garcia S, Sullivan PM, Gautier EL, Reichhart N, Sahel JA, Burns ME, Paques M, Sørensen TL, Strauss O, Guillonneau X, Delarasse C, Sennlaub F. Splenic monocytes drive pathogenic subretinal inflammation in age-related macular degeneration. J Neuroinflammation 2024; 21:22. [PMID: 38233865 PMCID: PMC10792815 DOI: 10.1186/s12974-024-03011-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 01/02/2024] [Indexed: 01/19/2024] Open
Abstract
Age-related macular degeneration (AMD) is invariably associated with the chronic accumulation of activated mononuclear phagocytes in the subretinal space. The mononuclear phagocytes are composed of microglial cells but also of monocyte-derived cells, which promote photoreceptor degeneration and choroidal neovascularization. Infiltrating blood monocytes can originate directly from bone marrow, but also from a splenic reservoir, where bone marrow monocytes develop into angiotensin II receptor (ATR1)+ splenic monocytes. The involvement of splenic monocytes in neurodegenerative diseases such as AMD is not well understood. Using acute inflammatory and well-phenotyped AMD models, we demonstrate that angiotensin II mobilizes ATR1+ splenic monocytes, which we show are defined by a transcriptional signature using single-cell RNA sequencing and differ functionally from bone marrow monocytes. Splenic monocytes participate in the chorio-retinal infiltration and their inhibition by ATR1 antagonist and splenectomy reduces the subretinal mononuclear phagocyte accumulation and pathological choroidal neovascularization formation. In aged AMD-risk ApoE2-expressing mice, a chronic AMD model, ATR1 antagonist and splenectomy also inhibit the chronic retinal inflammation and associated cone degeneration that characterizes these mice. Our observation of elevated levels of plasma angiotensin II in AMD patients, suggests that similar events take place in clinical disease and argue for the therapeutic potential of ATR1 antagonists to inhibit splenic monocytes for the treatment of blinding AMD.
Collapse
Affiliation(s)
- Christophe Roubeix
- Sorbonne Université, INSERM, CNRS, UMR_S 968, Institut de la Vision, 75012, Paris, France
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Experimental Ophthalmology, Department of Ophthalmology, Charitéplatz 1, 10117, Berlin, Germany
| | - Caroline Nous
- Sorbonne Université, INSERM, CNRS, UMR_S 968, Institut de la Vision, 75012, Paris, France
| | - Sébastien Augustin
- Sorbonne Université, INSERM, CNRS, UMR_S 968, Institut de la Vision, 75012, Paris, France
| | - Kaitryn E Ronning
- Sorbonne Université, INSERM, CNRS, UMR_S 968, Institut de la Vision, 75012, Paris, France
| | - Thibaud Mathis
- Service d'Ophtalmologie, Centre Hospitalier Universitaire de la Croix-Rousse, Hospices Civils de Lyon, Université Claude Bernard Lyon 1, 69004, Lyon, France
| | - Frédéric Blond
- Sorbonne Université, INSERM, CNRS, UMR_S 968, Institut de la Vision, 75012, Paris, France
| | | | - Sergio Crespo-Garcia
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Experimental Ophthalmology, Department of Ophthalmology, Charitéplatz 1, 10117, Berlin, Germany
| | - Patrick M Sullivan
- Department of Medicine, Centers for Aging and Geriatric Research Education and Clinical Center, Durham Veteran Affairs Medical Center, Duke University, Durham, NC, 27710, USA
| | - Emmanuel L Gautier
- Sorbonne Université, INSERM, UMR_S 1166, Hôpital de la Pitié-Salpêtrière, 75013, Paris, France
| | - Nadine Reichhart
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Experimental Ophthalmology, Department of Ophthalmology, Charitéplatz 1, 10117, Berlin, Germany
| | - José-Alain Sahel
- Sorbonne Université, INSERM, CNRS, UMR_S 968, Institut de la Vision, 75012, Paris, France
| | - Marie E Burns
- Center for Neuroscience, Department of Cell Biology and Human Anatomy, Department of Ophthalmology and Vision Science, University of California, Davis, CA, 95616, USA
| | - Michel Paques
- Sorbonne Université, INSERM, CNRS, UMR_S 968, Institut de la Vision, 75012, Paris, France
- Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, INSERM-DHOS Clinical Investigation Center 1423, Paris, France
| | - Torben Lykke Sørensen
- Clinical Eye Research Division, Department of Ophthalmology, Zealand University Hospital Roskilde, Roskilde, Denmark
- Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark
| | - Olaf Strauss
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Experimental Ophthalmology, Department of Ophthalmology, Charitéplatz 1, 10117, Berlin, Germany
| | - Xavier Guillonneau
- Sorbonne Université, INSERM, CNRS, UMR_S 968, Institut de la Vision, 75012, Paris, France
| | - Cécile Delarasse
- Sorbonne Université, INSERM, CNRS, UMR_S 968, Institut de la Vision, 75012, Paris, France.
| | - Florian Sennlaub
- Sorbonne Université, INSERM, CNRS, UMR_S 968, Institut de la Vision, 75012, Paris, France.
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Experimental Ophthalmology, Department of Ophthalmology, Charitéplatz 1, 10117, Berlin, Germany.
| |
Collapse
|
23
|
Laudenberg N, Kinuthia UM, Langmann T. Microglia depletion/repopulation does not affect light-induced retinal degeneration in mice. Front Immunol 2024; 14:1345382. [PMID: 38288111 PMCID: PMC10822957 DOI: 10.3389/fimmu.2023.1345382] [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: 11/27/2023] [Accepted: 12/28/2023] [Indexed: 01/31/2024] Open
Abstract
Reactive microglia are a hallmark of age-related retinal degenerative diseases including age-related macular degeneration (AMD). These cells are capable of secreting neurotoxic substances that may aggravate inflammation that leads to loss of photoreceptors and impaired vision. Despite their role in driving detrimental inflammation, microglia also play supporting roles in the retina as they are a crucial cellular component of the regulatory innate immune system. In this study, we used the colony stimulating factor 1 receptor (CSF1R)-antagonist PLX3397 to investigate the effects of microglia depletion and repopulation in a mouse model of acute retinal degeneration that mimics some aspects of dry AMD. Our main goal was to investigate whether microglia depletion and repopulation affects the outcome of light-induced retinal degeneration. We found that microglia depletion effectively decreased the expression of several key pro-inflammatory factors but was unable to influence the extent of retinal degeneration as determined by optical coherence tomography (OCT) and histology. Interestingly, we found prominent cell debris accumulation in the outer retina under conditions of microglia depletion, presumably due to the lack of efficient phagocytosis that could not be compensated by the retinal pigment epithelium. Moreover, our in vivo experiments showed that renewal of retinal microglia by repopulation did also not prevent rapid microglia activation or preserve photoreceptor death under conditions of light damage. We conclude that microglia ablation strongly reduces the expression of pro-inflammatory factors but cannot prevent photoreceptor loss in the light-damage paradigm of retinal degeneration.
Collapse
Affiliation(s)
- Nils Laudenberg
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Urbanus Muthai Kinuthia
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Thomas Langmann
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| |
Collapse
|
24
|
Blot G, Karadayi R, Przegralek L, Sartoris TM, Charles-Messance H, Augustin S, Negrier P, Blond F, Muñiz-Ruvalcaba FP, Rivera-de la Parra D, Vignaud L, Couturier A, Sahel JA, Acar N, Jimenez-Corona A, Delarasse C, Garfias Y, Sennlaub F, Guillonneau X. Perilipin 2-positive mononuclear phagocytes accumulate in the diabetic retina and promote PPARγ-dependent vasodegeneration. J Clin Invest 2023; 133:e161348. [PMID: 37781924 PMCID: PMC10702478 DOI: 10.1172/jci161348] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 08/01/2023] [Indexed: 10/03/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM), characterized by hyperglycemia and dyslipidemia, leads to nonproliferative diabetic retinopathy (NPDR). NPDR is associated with blood-retina barrier disruption, plasma exudates, microvascular degeneration, elevated inflammatory cytokine levels, and monocyte (Mo) infiltration. Whether and how the diabetes-associated changes in plasma lipid and carbohydrate levels modify Mo differentiation remains unknown. Here, we show that mononuclear phagocytes (MPs) in areas of vascular leakage in DR donor retinas expressed perilipin 2 (PLIN2), a marker of intracellular lipid load. Strong upregulation of PLIN2 was also observed when healthy donor Mos were treated with plasma from patients with T2DM or with palmitate concentrations typical of those found in T2DM plasma, but not under high-glucose conditions. PLIN2 expression correlated with the expression of other key genes involved in lipid metabolism (ACADVL, PDK4) and the DR biomarkers ANGPTL4 and CXCL8. Mechanistically, we show that lipid-exposed MPs induced capillary degeneration in ex vivo explants that was inhibited by pharmaceutical inhibition of PPARγ signaling. Our study reveals a mechanism linking dyslipidemia-induced MP polarization to the increased inflammatory cytokine levels and microvascular degeneration that characterize NPDR. This study provides comprehensive insights into the glycemia-independent activation of Mos in T2DM and identifies MP PPARγ as a target for inhibition of lipid-activated MPs in DR.
Collapse
Affiliation(s)
- Guillaume Blot
- Institute of Vision, Sorbonne University, INSERM, CNRS, Paris, France
- ED394 Physiology and Physiopathology Doctoral School, Sorbonne University, Paris, France
| | - Rémi Karadayi
- Institute of Vision, Sorbonne University, INSERM, CNRS, Paris, France
| | | | | | - Hugo Charles-Messance
- Institute of Vision, Sorbonne University, INSERM, CNRS, Paris, France
- ED394 Physiology and Physiopathology Doctoral School, Sorbonne University, Paris, France
| | | | - Pierre Negrier
- Institute of Vision, Sorbonne University, INSERM, CNRS, Paris, France
- A. de Rothschild Foundation Hospital, Paris, France
| | - Frédéric Blond
- Institute of Vision, Sorbonne University, INSERM, CNRS, Paris, France
| | | | - David Rivera-de la Parra
- Comprehensive Care Center for Diabetes Patients, Salvador Zubrian National Institute of Health Sciences and Nutrition, Mexico City, Mexico
- Institute of Ophthalmology “Fundación Conde de Valenciana” I.A.P., Mexico City, Mexico
| | - Lucile Vignaud
- Institute of Vision, Sorbonne University, INSERM, CNRS, Paris, France
| | - Aude Couturier
- Institute of Vision, Sorbonne University, INSERM, CNRS, Paris, France
- ED394 Physiology and Physiopathology Doctoral School, Sorbonne University, Paris, France
- Department of Ophthalmology, Hôpital Lariboisière, AP-HP, University of Paris, Paris, France
| | - José-Alain Sahel
- Institute of Vision, Sorbonne University, INSERM, CNRS, Paris, France
- A. de Rothschild Foundation Hospital, Paris, France
- Department of Ophthalmology, The University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- CHNO des Quinze-Vingts, Institut Hospitalo-Universitaire FOReSIGHT, INSERM-DGOS CIC 1423, Paris, France
| | - Niyazi Acar
- Eye and Nutrition Research Group, Center for Taste and Food Sciences, CNRS, INRAE, Institut Agro, Bourgogne Franche-Comté University, Dijon, France
| | - Aida Jimenez-Corona
- Department of Epidemiology and Visual Health, Instituto de Oftalmología Fundación Conde de Valenciana, Mexico City, Mexico
- General Directorate of Epidemiology, Secretariat of Health, Mexico City, Mexico
| | - Cécile Delarasse
- Institute of Vision, Sorbonne University, INSERM, CNRS, Paris, France
| | - Yonathan Garfias
- Department of Biochemistry, School of Medicine, National Autonomous University, Mexico City, Mexico
- Cell and Tissue Biology, Research Unit, Instituto de Oftalmología Fundación Conde de Valenciana”, Mexico City, Mexico
| | - Florian Sennlaub
- Institute of Vision, Sorbonne University, INSERM, CNRS, Paris, France
| | | |
Collapse
|
25
|
Twarog M, Schustak J, Xu Y, Coble M, Dolan K, Esterberg R, Huang Q, Saint-Geniez M, Bao Y. TNFα induced by DNA-sensing in macrophage compromises retinal pigment epithelial (RPE) barrier function. Sci Rep 2023; 13:14451. [PMID: 37660150 PMCID: PMC10475136 DOI: 10.1038/s41598-023-41610-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 08/29/2023] [Indexed: 09/04/2023] Open
Abstract
Increasing evidence suggests that chronic inflammation plays an important role in the pathogenesis of age-related macular degeneration (AMD); however, the precise pathogenic stressors and sensors, and their impact on disease progression remain unclear. Several studies have demonstrated that type I interferon (IFN) response is activated in the retinal pigment epithelium (RPE) of AMD patients. Previously, we demonstrated that human RPE cells can initiate RNA-mediated type I IFN responses through RIG-I, yet are unable to directly sense and respond to DNA. In this study, we utilized a co-culture system combining primary human macrophage and iPS-derived RPE to study how each cell type responds to nucleic acids challenges and their effect on RPE barrier function in a homotypic and heterotypic manner. We find that DNA-induced macrophage activation induces an IFN response in the RPE, and compromises RPE barrier function via tight-junction remodeling. Investigation of the secreted cytokines responsible for RPE dysfunction following DNA-induced macrophages activation indicates that neutralization of macrophage-secreted TNFα, but not IFNβ, is sufficient to rescue RPE morphology and barrier function. Our data reveals a novel mechanism of intercellular communication by which DNA induces RPE dysfunction via macrophage-secreted TNFa, highlighting the complexity and potential pathological relevance of RPE and macrophage interactions.
Collapse
Affiliation(s)
- Michael Twarog
- Department of Ophthalmology, Novartis Institutes for BioMedical Research, 22 Windsor Street, Cambridge, MA, USA
| | - Joshua Schustak
- Department of Ophthalmology, Novartis Institutes for BioMedical Research, 22 Windsor Street, Cambridge, MA, USA
| | - YongYao Xu
- Department of Ophthalmology, Novartis Institutes for BioMedical Research, 22 Windsor Street, Cambridge, MA, USA
| | - Matthew Coble
- Department of Ophthalmology, Novartis Institutes for BioMedical Research, 22 Windsor Street, Cambridge, MA, USA
| | - Katie Dolan
- Department of Ophthalmology, Novartis Institutes for BioMedical Research, 22 Windsor Street, Cambridge, MA, USA
| | - Robert Esterberg
- Department of Ophthalmology, Novartis Institutes for BioMedical Research, 22 Windsor Street, Cambridge, MA, USA
| | - Qian Huang
- Department of Ophthalmology, Novartis Institutes for BioMedical Research, 22 Windsor Street, Cambridge, MA, USA
| | - Magali Saint-Geniez
- Department of Ophthalmology, Novartis Institutes for BioMedical Research, 22 Windsor Street, Cambridge, MA, USA
| | - Yi Bao
- Department of Ophthalmology, Novartis Institutes for BioMedical Research, 22 Windsor Street, Cambridge, MA, USA.
| |
Collapse
|
26
|
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: 1.5] [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
|
27
|
Khan AH, Chowers I, Lotery AJ. Beyond the Complement Cascade: Insights into Systemic Immunosenescence and Inflammaging in Age-Related Macular Degeneration and Current Barriers to Treatment. Cells 2023; 12:1708. [PMID: 37443742 PMCID: PMC10340338 DOI: 10.3390/cells12131708] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 06/22/2023] [Accepted: 06/22/2023] [Indexed: 07/15/2023] Open
Abstract
Landmark genetic studies have revealed the effect of complement biology and its regulation on the pathogenesis of age-related macular degeneration (AMD). Limited phase 3 clinical trial data showing a benefit of complement inhibition in AMD raises the prospect of more complex mediators at play. Substantial evidence supports the role of para-inflammation in maintaining homeostasis in the retina and choroid. With increasing age, a decline in immune system regulation, known as immunosenescence, has been shown to alter the equilibrium maintained by para-inflammation. The altered equilibrium results in chronic, sterile inflammation with aging, termed 'inflammaging', including in the retina and choroid. The chronic inflammatory state in AMD is complex, with contributions from cells of the innate and adaptive branches of the immune system, sometimes with overlapping features, and the interaction of their secretory products with retinal cells such as microglia and retinal pigment epithelium (RPE), extracellular matrix and choroidal vascular endothelial cells. In this review, the chronic inflammatory state in AMD will be explored by immune cell type, with a discussion of factors that will need to be overcome in the development of curative therapies.
Collapse
Affiliation(s)
- Adnan H. Khan
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO17 1BJ, UK
- Southampton Eye Unit, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| | - Itay Chowers
- Department of Ophthalmology, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 91121, Israel
| | - Andrew J. Lotery
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO17 1BJ, UK
- Southampton Eye Unit, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| |
Collapse
|
28
|
Tzoumas N, Riding G, Williams MA, Steel DH. Complement inhibitors for age-related macular degeneration. Cochrane Database Syst Rev 2023; 6:CD009300. [PMID: 37314061 PMCID: PMC10266126 DOI: 10.1002/14651858.cd009300.pub3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
BACKGROUND Age-related macular degeneration (AMD) is a common eye disease and leading cause of sight loss worldwide. Despite its high prevalence and increasing incidence as populations age, AMD remains incurable and there are no treatments for most patients. Mounting genetic and molecular evidence implicates complement system overactivity as a key driver of AMD development and progression. The last decade has seen the development of several novel therapeutics targeting complement in the eye for the treatment of AMD. This review update encompasses the results of the first randomised controlled trials in this field. OBJECTIVES To assess the effects and safety of complement inhibitors in the prevention or treatment of AMD. SEARCH METHODS We searched CENTRAL on the Cochrane Library, MEDLINE, Embase, LILACS, Web of Science, ISRCTN registry, ClinicalTrials.gov, and the WHO ICTRP to 29 June 2022 with no language restrictions. We also contacted companies running clinical trials for unpublished data. SELECTION CRITERIA We included randomised controlled trials (RCTs) with parallel groups and comparator arms that studied complement inhibition for advanced AMD prevention/treatment. DATA COLLECTION AND ANALYSIS Two authors independently assessed search results and resolved discrepancies through discussion. Outcome measures evaluated at one year included change in best-corrected visual acuity (BCVA), untransformed and square root-transformed geographic atrophy (GA) lesion size progression, development of macular neovascularisation (MNV) or exudative AMD, development of endophthalmitis, loss of ≥ 15 letters of BCVA, change in low luminance visual acuity, and change in quality of life. We assessed risk of bias and evidence certainty using Cochrane risk of bias and GRADE tools. MAIN RESULTS Ten RCTs with 4052 participants and eyes with GA were included. Nine evaluated intravitreal (IVT) administrations against sham, and one investigated an intravenous agent against placebo. Seven studies excluded patients with prior MNV in the non-study eye, whereas the three pegcetacoplan studies did not. The risk of bias in the included studies was low overall. We also synthesised results of two intravitreal agents (lampalizumab, pegcetacoplan) at monthly and every-other-month (EOM) dosing intervals. Efficacy and safety of IVT lampalizumab versus sham for GA For 1932 participants in three studies, lampalizumab did not meaningfully change BCVA given monthly (+1.03 letters, 95% confidence interval (CI) -0.19 to 2.25) or EOM (+0.22 letters, 95% CI -1.00 to 1.44) (high-certainty evidence). For 1920 participants, lampalizumab did not meaningfully change GA lesion growth given monthly (+0.07 mm², 95% CI -0.09 to 0.23; moderate-certainty due to imprecision) or EOM (+0.07 mm², 95% CI -0.05 to 0.19; high-certainty). For 2000 participants, lampalizumab may have also increased MNV risk given monthly (RR 1.77, 95% CI 0.73 to 4.30) and EOM (RR 1.70, 95% CI 0.67 to 4.28), based on low-certainty evidence. The incidence of endophthalmitis in patients treated with monthly and EOM lampalizumab was 4 per 1000 (0 to 87) and 3 per 1000 (0 to 62), respectively, based on moderate-certainty evidence. Efficacy and safety of IVT pegcetacoplan versus sham for GA For 242 participants in one study, pegcetacoplan probably did not meaningfully change BCVA given monthly (+1.05 letters, 95% CI -2.71 to 4.81) or EOM (-1.42 letters, 95% CI -5.25 to 2.41), as supported by moderate-certainty evidence. In contrast, for 1208 participants across three studies, pegcetacoplan meaningfully reduced GA lesion growth when given monthly (-0.38 mm², 95% CI -0.57 to -0.19) and EOM (-0.29 mm², 95% CI -0.44 to -0.13), with high certainty. These reductions correspond to 19.2% and 14.8% versus sham, respectively. A post hoc analysis showed possibly greater benefits in 446 participants with extrafoveal GA given monthly (-0.67 mm², 95% CI -0.98 to -0.36) and EOM (-0.60 mm², 95% CI -0.91 to -0.30), representing 26.1% and 23.3% reductions, respectively. However, we did not have data on subfoveal GA growth to undertake a formal subgroup analysis. In 1502 participants, there is low-certainty evidence that pegcetacoplan may have increased MNV risk when given monthly (RR 4.47, 95% CI 0.41 to 48.98) or EOM (RR 2.29, 95% CI 0.46 to 11.35). The incidence of endophthalmitis in patients treated with monthly and EOM pegcetacoplan was 6 per 1000 (1 to 53) and 8 per 1000 (1 to 70) respectively, based on moderate-certainty evidence. Efficacy and safety of IVT avacincaptad pegol versus sham for GA In a study of 260 participants with extrafoveal or juxtafoveal GA, monthly avacincaptad pegol probably did not result in a clinically meaningful change in BCVA at 2 mg (+1.39 letters, 95% CI -5.89 to 8.67) or 4 mg (-0.28 letters, 95% CI -8.74 to 8.18), based on moderate-certainty evidence. Despite this, the drug was still found to have probably reduced GA lesion growth, with estimates of 30.5% reduction at 2 mg (-0.70 mm², 95% CI -1.99 to 0.59) and 25.6% reduction at 4 mg (-0.71 mm², 95% CI -1.92 to 0.51), based on moderate-certainty evidence. Avacincaptad pegol may have also increased the risk of developing MNV (RR 3.13, 95% CI 0.93 to 10.55), although this evidence is of low certainty. There were no cases of endophthalmitis reported in this study. AUTHORS' CONCLUSIONS Despite confirmation of the negative findings of intravitreal lampalizumab across all endpoints, local complement inhibition with intravitreal pegcetacoplan meaningfully reduces GA lesion growth relative to sham at one year. Inhibition of complement C5 with intravitreal avacincaptad pegol is also an emerging therapy with probable benefits on anatomical endpoints in the extrafoveal or juxtafoveal GA population. However, there is currently no evidence that complement inhibition with any agent improves functional endpoints in advanced AMD; further results from the phase 3 studies of pegcetacoplan and avacincaptad pegol are eagerly awaited. Progression to MNV or exudative AMD is a possible emergent adverse event of complement inhibition, requiring careful consideration should these agents be used clinically. Intravitreal administration of complement inhibitors is probably associated with a small risk of endophthalmitis, which may be higher than that of other intravitreal therapies. Further research is likely to have an important impact on our confidence in the estimates of adverse effects and may change these. The optimal dosing regimens, treatment duration, and cost-effectiveness of such therapies are yet to be established.
Collapse
Affiliation(s)
- Nikolaos Tzoumas
- Biosciences Institute, Newcastle University, Newcastle-upon-Tyne, UK
- Sunderland Eye Infirmary, Sunderland, UK
| | - George Riding
- Biosciences Institute, Newcastle University, Newcastle-upon-Tyne, UK
- North Middlesex University Hospital NHS Trust, London, UK
| | - Michael A Williams
- School of Medicine, Dentistry and Biomedical Science, Queen's University of Belfast, Belfast, UK
| | - David Hw Steel
- Biosciences Institute, Newcastle University, Newcastle-upon-Tyne, UK
- Sunderland Eye Infirmary, Sunderland, UK
| |
Collapse
|
29
|
Kuchroo M, DiStasio M, Song E, Calapkulu E, Zhang L, Ige M, Sheth AH, Majdoubi A, Menon M, Tong A, Godavarthi A, Xing Y, Gigante S, Steach H, Huang J, Huguet G, Narain J, You K, Mourgkos G, Dhodapkar RM, Hirn MJ, Rieck B, Wolf G, Krishnaswamy S, Hafler BP. Single-cell analysis reveals inflammatory interactions driving macular degeneration. Nat Commun 2023; 14:2589. [PMID: 37147305 PMCID: PMC10162998 DOI: 10.1038/s41467-023-37025-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 02/27/2023] [Indexed: 05/07/2023] Open
Abstract
Due to commonalities in pathophysiology, age-related macular degeneration (AMD) represents a uniquely accessible model to investigate therapies for neurodegenerative diseases, leading us to examine whether pathways of disease progression are shared across neurodegenerative conditions. Here we use single-nucleus RNA sequencing to profile lesions from 11 postmortem human retinas with age-related macular degeneration and 6 control retinas with no history of retinal disease. We create a machine-learning pipeline based on recent advances in data geometry and topology and identify activated glial populations enriched in the early phase of disease. Examining single-cell data from Alzheimer's disease and progressive multiple sclerosis with our pipeline, we find a similar glial activation profile enriched in the early phase of these neurodegenerative diseases. In late-stage age-related macular degeneration, we identify a microglia-to-astrocyte signaling axis mediated by interleukin-1β which drives angiogenesis characteristic of disease pathogenesis. We validated this mechanism using in vitro and in vivo assays in mouse, identifying a possible new therapeutic target for AMD and possibly other neurodegenerative conditions. Thus, due to shared glial states, the retina provides a potential system for investigating therapeutic approaches in neurodegenerative diseases.
Collapse
Affiliation(s)
- Manik Kuchroo
- Department of Neuroscience, Yale University, New Haven, CT, USA
| | | | - Eric Song
- Department of Ophthalmology and Visual Science, Yale University, New Haven, CT, USA
| | - Eda Calapkulu
- Department of Ophthalmology and Visual Science, Yale University, New Haven, CT, USA
| | - Le Zhang
- Department of Neuroscience, Yale University, New Haven, CT, USA
- Department of Neurology, Yale University, New Haven, CT, USA
| | - Maryam Ige
- Yale School of Medicine, New Haven, CT, USA
| | | | - Abdelilah Majdoubi
- Department of Ophthalmology and Visual Science, Yale University, New Haven, CT, USA
| | - Madhvi Menon
- Division of Infection, Immunity and Respiratory Medicine, University of Manchester, Manchester, UK
| | - Alexander Tong
- Department of Computer Science, Yale University, New Haven, CT, USA
| | | | - Yu Xing
- Department of Ophthalmology and Visual Science, Yale University, New Haven, CT, USA
| | - Scott Gigante
- Computational Biology, Bioinformatics Program, Yale University, New Haven, CT, USA
| | - Holly Steach
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Jessie Huang
- Department of Computer Science, Yale University, New Haven, CT, USA
| | - Guillaume Huguet
- Mila-Quebec AI institute, Montréal, QC, Canada
- Department of Mathematics and Statistics, Université de Montréal, Montréal, QC, Canada
| | - Janhavi Narain
- Department of Computer Science, Rutgers University, New Brunswick, NJ, USA
| | - Kisung You
- Department of Genetics, Yale University, New Haven, CT, USA
| | - George Mourgkos
- Department of Ophthalmology and Visual Science, Yale University, New Haven, CT, USA
| | | | - Matthew J Hirn
- Department of Computational Mathematics, Science and Engineering, Michigan State University, East Lansing, MI, USA
- Department of Mathematics, Michigan State University, East Lansing, MI, USA
| | - Bastian Rieck
- Department of Biosystems Science and Engineering, ETH Zurich, Zurich, Switzerland
| | - Guy Wolf
- Mila-Quebec AI institute, Montréal, QC, Canada
- Department of Mathematics and Statistics, Université de Montréal, Montréal, QC, Canada
| | - Smita Krishnaswamy
- Department of Computer Science, Yale University, New Haven, CT, USA.
- Department of Genetics, Yale University, New Haven, CT, USA.
| | - Brian P Hafler
- Department of Pathology, Yale University, New Haven, CT, USA.
- Department of Ophthalmology and Visual Science, Yale University, New Haven, CT, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
30
|
Hammadi S, Tzoumas N, Ferrara M, Meschede IP, Lo K, Harris C, Lako M, Steel DH. Bruch's Membrane: A Key Consideration with Complement-Based Therapies for Age-Related Macular Degeneration. J Clin Med 2023; 12:2870. [PMID: 37109207 PMCID: PMC10145879 DOI: 10.3390/jcm12082870] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 03/29/2023] [Accepted: 04/03/2023] [Indexed: 04/29/2023] Open
Abstract
The complement system is crucial for immune surveillance, providing the body's first line of defence against pathogens. However, an imbalance in its regulators can lead to inappropriate overactivation, resulting in diseases such as age-related macular degeneration (AMD), a leading cause of irreversible blindness globally affecting around 200 million people. Complement activation in AMD is believed to begin in the choriocapillaris, but it also plays a critical role in the subretinal and retinal pigment epithelium (RPE) spaces. Bruch's membrane (BrM) acts as a barrier between the retina/RPE and choroid, hindering complement protein diffusion. This impediment increases with age and AMD, leading to compartmentalisation of complement activation. In this review, we comprehensively examine the structure and function of BrM, including its age-related changes visible through in vivo imaging, and the consequences of complement dysfunction on AMD pathogenesis. We also explore the potential and limitations of various delivery routes (systemic, intravitreal, subretinal, and suprachoroidal) for safe and effective delivery of conventional and gene therapy-based complement inhibitors to treat AMD. Further research is needed to understand the diffusion of complement proteins across BrM and optimise therapeutic delivery to the retina.
Collapse
Affiliation(s)
- Sarah Hammadi
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Nikolaos Tzoumas
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- Sunderland Eye Infirmary, Queen Alexandra Rd., Sunderland SR2 9H, UK
| | | | - Ingrid Porpino Meschede
- Gyroscope Therapeutics Limited, a Novartis Company, Rolling Stock Yard, 6th Floor, 188 York Way, London N7 9AS, UK
| | - Katharina Lo
- Gyroscope Therapeutics Limited, a Novartis Company, Rolling Stock Yard, 6th Floor, 188 York Way, London N7 9AS, UK
| | - Claire Harris
- Gyroscope Therapeutics Limited, a Novartis Company, Rolling Stock Yard, 6th Floor, 188 York Way, London N7 9AS, UK
- Clinical and Translational Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Majlinda Lako
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - David H. Steel
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- Sunderland Eye Infirmary, Queen Alexandra Rd., Sunderland SR2 9H, UK
| |
Collapse
|
31
|
Ramírez-Pardo I, Villarejo-Zori B, Jiménez-Loygorri JI, Sierra-Filardi E, Alonso-Gil S, Mariño G, de la Villa P, Fitze PS, Fuentes JM, García-Escudero R, Ferrington DA, Gomez-Sintes R, Boya P. Ambra1 haploinsufficiency in CD1 mice results in metabolic alterations and exacerbates age-associated retinal degeneration. Autophagy 2023; 19:784-804. [PMID: 35875981 PMCID: PMC9980615 DOI: 10.1080/15548627.2022.2103307] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Macroautophagy/autophagy is a key process in the maintenance of cellular homeostasis. The age-dependent decline in retinal autophagy has been associated with photoreceptor degeneration. Retinal dysfunction can also result from damage to the retinal pigment epithelium (RPE), as the RPE-retina constitutes an important metabolic ecosystem that must be finely tuned to preserve visual function. While studies of mice lacking essential autophagy genes have revealed a predisposition to retinal degeneration, the consequences of a moderate reduction in autophagy, similar to that which occurs during physiological aging, remain unclear. Here, we described a retinal phenotype consistent with accelerated aging in mice carrying a haploinsufficiency for Ambra1, a pro-autophagic gene. These mice showed protein aggregation in the retina and RPE, metabolic underperformance, and premature vision loss. Moreover, Ambra1+/gt mice were more prone to retinal degeneration after RPE stress. These findings indicate that autophagy provides crucial support to RPE-retinal metabolism and protects the retina against stress and physiological aging.Abbreviations : 4-HNE: 4-hydroxynonenal; AMBRA1: autophagy and beclin 1 regulator 1, AMD: age-related macular degeneration;; GCL: ganglion cell layer; GFAP: glial fibrillary acidic protein; GLUL: glutamine synthetase/glutamate-ammonia ligase; HCL: hierarchical clustering; INL: inner nuclear layer; IPL: inner plexiform layer; LC/GC-MS: liquid chromatography/gas chromatography-mass spectrometry; MA: middle-aged; MTDR: MitoTracker Deep Red; MFI: mean fluorescence intensity; NL: NH4Cl and leupeptin; Nqo: NAD(P)H quinone dehydrogenase; ONL: outer nuclear layer; OPL: outer plexiform layer; OP: oscillatory potentials; OXPHOS: oxidative phosphorylation; PCR: polymerase chain reaction; PRKC/PKCα: protein kinase C; POS: photoreceptor outer segment; RGC: retinal ganglion cells; RPE: retinal pigment epithelium; SI: sodium iodate; TCA: tricarboxylic acid.
Collapse
Affiliation(s)
- Ignacio Ramírez-Pardo
- Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Madrid, Spain
| | - Beatriz Villarejo-Zori
- Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Madrid, Spain
| | - Juan Ignacio Jiménez-Loygorri
- Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Madrid, Spain
| | - Elena Sierra-Filardi
- Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Madrid, Spain
| | - Sandra Alonso-Gil
- Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Madrid, Spain
| | | | - Pedro de la Villa
- Department of Systems Biology, University of Alcalá, Alcalá de Henares, Madrid, Spain.,Vision neurophisiology group, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Patrick S Fitze
- Departament of Biodiversity and Evolutionary Biology, Museo Nacional de Ciencias Naturales, CSIC, Madrid, Spain
| | - José Manuel Fuentes
- Department of Biochemistry, Molecular Biology and Genetics, Faculty of Nursing and Occupational Therapy, University of Extremadura, Cáceres, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Nerodegenerative Diseases unit, Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), Cáceres, Spain
| | - Ramón García-Escudero
- Molecular Oncology Unit, CIEMAT, Madrid, Spain.,Biomedical Research Institute I+12, University Hospital 12 de Octubre, Madrid, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Deborah A Ferrington
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN, USA
| | - Raquel Gomez-Sintes
- Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Madrid, Spain
| | - Patricia Boya
- Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Madrid, Spain
| |
Collapse
|
32
|
Hata M, Hata M, Andriessen EM, Juneau R, Pilon F, Crespo-Garcia S, Diaz-Marin R, Guber V, Binet F, Fournier F, Buscarlet M, Grou C, Calderon V, Heckel E, Melichar HJ, Joyal JS, Wilson AM, Sapieha P. Early-life peripheral infections reprogram retinal microglia and aggravate neovascular age-related macular degeneration in later life. J Clin Invest 2023; 133:159757. [PMID: 36787231 PMCID: PMC9927938 DOI: 10.1172/jci159757] [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: 02/28/2022] [Accepted: 12/16/2022] [Indexed: 02/15/2023] Open
Abstract
Pathological neovascularization in age-related macular degeneration (nvAMD) drives the principal cause of blindness in the elderly. While there is a robust genetic association between genes of innate immunity and AMD, genome-to-phenome relationships are low, suggesting a critical contribution of environmental triggers of disease. Possible insight comes from the observation that a past history of infection with pathogens such as Chlamydia pneumoniae, or other systemic inflammation, can predispose to nvAMD in later life. Using a mouse model of nvAMD with prior C. pneumoniae infection, endotoxin exposure, and genetic ablation of distinct immune cell populations, we demonstrated that peripheral infections elicited epigenetic reprogramming that led to a persistent memory state in retinal CX3CR1+ mononuclear phagocytes (MNPs). The immune imprinting persisted long after the initial inflammation had subsided and ultimately exacerbated choroidal neovascularization in a model of nvAMD. Single-cell assay for transposase-accessible chromatin sequencing (scATAC-seq) identified activating transcription factor 3 (ATF3) as a central mediator of retina-resident MNP reprogramming following peripheral inflammation. ATF3 polarized MNPs toward a reparative phenotype biased toward production of proangiogenic factors in response to subsequent injury. Therefore, a past history of bacterial endotoxin-induced inflammation can lead to immunological reprograming within CNS-resident MNPs and aggravate pathological angiogenesis in the aging retina.
Collapse
Affiliation(s)
- Masayuki Hata
- Department of Ophthalmology,,Department of Biochemistry and Molecular Medicine, and
| | | | - Elisabeth M.M.A. Andriessen
- Department of Biomedical Sciences, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec, Canada
| | | | | | - Sergio Crespo-Garcia
- Department of Ophthalmology,,Department of Biochemistry and Molecular Medicine, and
| | | | | | | | | | | | - Caroline Grou
- Bioinformatics Core Facility, Institut de recherches cliniques de Montréal, Montreal, Quebec, Canada
| | - Virginie Calderon
- Bioinformatics Core Facility, Institut de recherches cliniques de Montréal, Montreal, Quebec, Canada
| | - Emilie Heckel
- Department of Pediatrics, Ophthalmology, and Pharmacology, Centre Hospitalier Universitaire Ste-Justine Research Center, Montreal, Quebec, Canada
| | - Heather J. Melichar
- Department of Medicine, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec, Canada
| | - Jean-Sebastien Joyal
- Department of Pediatrics, Ophthalmology, and Pharmacology, Centre Hospitalier Universitaire Ste-Justine Research Center, Montreal, Quebec, Canada
| | | | - Przemyslaw Sapieha
- Department of Ophthalmology,,Department of Biochemistry and Molecular Medicine, and,Department of Biomedical Sciences, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec, Canada
| |
Collapse
|
33
|
Augustin S, Lam M, Lavalette S, Verschueren A, Blond F, Forster V, Przegralek L, He Z, Lewandowski D, Bemelmans AP, Picaud S, Sahel JA, Mathis T, Paques M, Thuret G, Guillonneau X, Delarasse C, Sennlaub F. Melanophages give rise to hyperreflective foci in AMD, a disease-progression marker. J Neuroinflammation 2023; 20:28. [PMID: 36755326 PMCID: PMC9906876 DOI: 10.1186/s12974-023-02699-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 01/13/2023] [Indexed: 02/10/2023] Open
Abstract
Retinal melanosome/melanolipofuscin-containing cells (MCCs), clinically visible as hyperreflective foci (HRF) and a highly predictive imaging biomarker for the progression of age-related macular degeneration (AMD), are widely believed to be migrating retinal pigment epithelial (RPE) cells. Using human donor tissue, we identify the vast majority of MCCs as melanophages, melanosome/melanolipofuscin-laden mononuclear phagocytes (MPs). Using serial block-face scanning electron microscopy, RPE flatmounts, bone marrow transplantation and in vitro experiments, we show how retinal melanophages form by the transfer of melanosomes from the RPE to subretinal MPs when the "don't eat me" signal CD47 is blocked. These melanophages give rise to hyperreflective foci in Cd47-/--mice in vivo, and are associated with RPE dysmorphia similar to intermediate AMD. Finally, we show that Cd47 expression in human RPE declines with age and in AMD, which likely participates in melanophage formation and RPE decline. Boosting CD47 expression in AMD might protect RPE cells and delay AMD progression.
Collapse
Affiliation(s)
- Sebastien Augustin
- Sorbonne Université, INSERM, CNRS, UMR_S 968, Institut de la Vision, 17 rue Moreau, 75012 Paris, France
| | - Marion Lam
- Ophthalmology Department, Université de Paris, APHP, Hôpital Lariboisière, 75010 Paris, France
| | - Sophie Lavalette
- Sorbonne Université, INSERM, CNRS, UMR_S 968, Institut de la Vision, 17 rue Moreau, 75012 Paris, France
| | - Anna Verschueren
- grid.415610.70000 0001 0657 9752Centre Hospitalier National d’Ophtalmologie des Quinze-Vingts, INSERM-DHOS CIC 503, Paris, France
| | - Frédéric Blond
- Sorbonne Université, INSERM, CNRS, UMR_S 968, Institut de la Vision, 17 rue Moreau, 75012 Paris, France
| | - Valérie Forster
- Sorbonne Université, INSERM, CNRS, UMR_S 968, Institut de la Vision, 17 rue Moreau, 75012 Paris, France
| | - Lauriane Przegralek
- Sorbonne Université, INSERM, CNRS, UMR_S 968, Institut de la Vision, 17 rue Moreau, 75012 Paris, France
| | - Zhiguo He
- grid.6279.a0000 0001 2158 1682Laboratory of Biology, Engineering and Imaging for Ophthalmology, BiiO, EA2521, Faculty of Medicine, University of Saint Etienne, Saint Etienne, France
| | - Daniel Lewandowski
- grid.457349.80000 0004 0623 0579Cellules Souches et Radiations, Stabilité Génétique, Université de Paris, Université Paris-Saclay, Inserm, CEA, Fontenay-Aux-Roses, France
| | - Alexis-Pierre Bemelmans
- grid.457349.80000 0004 0623 0579Laboratoire des Maladies Neurodégénératives, Université Paris-Saclay, CEA, CNRS, MIRCen, Fontenay-Aux-Roses, France
| | - Serge Picaud
- Sorbonne Université, INSERM, CNRS, UMR_S 968, Institut de la Vision, 17 rue Moreau, 75012 Paris, France
| | - José-Alain Sahel
- Sorbonne Université, INSERM, CNRS, UMR_S 968, Institut de la Vision, 17 rue Moreau, 75012 Paris, France ,grid.415610.70000 0001 0657 9752Centre Hospitalier National d’Ophtalmologie des Quinze-Vingts, INSERM-DHOS CIC 503, Paris, France
| | - Thibaud Mathis
- grid.7849.20000 0001 2150 7757Service d’Ophtalmologie, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, UMR CNRS 5510 MATEIS, Université Lyon 1, 103 Grande rue de la Croix Rousse, 69317 Lyon Cedex 04, France
| | - Michel Paques
- Sorbonne Université, INSERM, CNRS, UMR_S 968, Institut de la Vision, 17 rue Moreau, 75012 Paris, France ,grid.415610.70000 0001 0657 9752Centre Hospitalier National d’Ophtalmologie des Quinze-Vingts, INSERM-DHOS CIC 503, Paris, France
| | - Gilles Thuret
- grid.6279.a0000 0001 2158 1682Laboratory of Biology, Engineering and Imaging for Ophthalmology, BiiO, EA2521, Faculty of Medicine, University of Saint Etienne, Saint Etienne, France
| | - Xavier Guillonneau
- Sorbonne Université, INSERM, CNRS, UMR_S 968, Institut de la Vision, 17 rue Moreau, 75012 Paris, France
| | - Cécile Delarasse
- Sorbonne Université, INSERM, CNRS, UMR_S 968, Institut de la Vision, 17 rue Moreau, 75012 Paris, France
| | - Florian Sennlaub
- Sorbonne Université, INSERM, CNRS, UMR_S 968, Institut de la Vision, 17 rue Moreau, 75012, Paris, France.
| |
Collapse
|
34
|
Su N, Hansen U, Plagemann T, Gäher K, Leclaire MD, König J, Höhn A, Grune T, Uhlig CE, Eter N, Heiduschka P. Sub-Retinal Injection of Human Lipofuscin in the Mouse - A Model of "Dry" Age-Related Macular Degeneration? Aging Dis 2023; 14:184-203. [PMID: 36818570 PMCID: PMC9937713 DOI: 10.14336/ad.2022.0626] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 06/26/2022] [Indexed: 11/18/2022] Open
Abstract
Lipofuscin (LF) accumulates during lifetime in the retinal pigment epithelium (RPE) and is thought to play a crucial role in intermediate and late age-related macular degeneration (AMD). In an attemt to simulate aged retina and to study response of retinal microglia and RPE cells to LF, we injected a suspension of LF into the subretinal space of adult mice. LF suspension was obtained from human donor eyes. Subretinal injection of PBS or sham injection served as a control. Eyes were inspected by autofluorescence and optical coherence tomography, by electroretinography and on histological and ultrastructural levels. Levels of cytokine mRNA were determined by quantitative PCR separately in the RPE/choroid complex and in the retina. After injection of LF, microglial cells migrated quickly into the subretinal space to close proximity to RPE cells and phagocytosed LF particles. Retinal function was affected only slightly by LF within the first two weeks. After longer time, RPE cells showed clear signs of melanin loss and degradation. Levels of mRNA of inflammatory cytokines increased sharply after injection of both PBS and LF and were higher in the RPE/choroid complex than in the retina and were slightly higher after LF injection. In conclusion, subretinal injection of LF causes an activation of microglial cells and their migration into subretinal space, enhanced expression of inflammatory cytokines and a gradual degradation of RPE cells. These features are found also in an aging retina, and subretinal injection of LF could be a model for intermediate and late AMD.
Collapse
Affiliation(s)
- Nan Su
- Research Laboratory, Department of Ophthalmology, University
Medical Center, Münster, Germany.
- Department of Ophthalmology, The First Affiliated Hospital of
Zhengzhou University, Zhengzhou, China.
| | - Uwe Hansen
- Institute of Musculoskeletal Medicine, Medical Faculty,
University of Münster, Münster, Germany.
| | - Tanja Plagemann
- Research Laboratory, Department of Ophthalmology, University
Medical Center, Münster, Germany.
| | - Karin Gäher
- Institute of Musculoskeletal Medicine, Medical Faculty,
University of Münster, Münster, Germany.
| | - M. Dominik Leclaire
- Research Laboratory, Department of Ophthalmology, University
Medical Center, Münster, Germany.
| | - Jeannette König
- German Institute of Human Nutrition,
Potsdam-Rehbrücke, Germany.
| | - Annika Höhn
- German Institute of Human Nutrition,
Potsdam-Rehbrücke, Germany.
| | - Tilman Grune
- German Institute of Human Nutrition,
Potsdam-Rehbrücke, Germany.
| | - Constantin E. Uhlig
- Cornea Bank Münster, Department of Ophthalmology,
University Medical Center, Münster, Germany.
| | - Nicole Eter
- Department of Ophthalmology, University Medical Center,
Münster, Germany.
| | - Peter Heiduschka
- Research Laboratory, Department of Ophthalmology, University
Medical Center, Münster, Germany.
| |
Collapse
|
35
|
P2X7-dependent immune pathways in retinal diseases. Neuropharmacology 2023; 223:109332. [PMID: 36372269 DOI: 10.1016/j.neuropharm.2022.109332] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/28/2022] [Accepted: 11/09/2022] [Indexed: 11/13/2022]
Abstract
Adenosine triphosphate (ATP) is a signalling molecule acting as a neurotransmitter but also as a danger signal. The purinergic receptor P2X7 is the main sensor of high concentration of ATP released by damaged cells. In the eye, P2X7 is expressed by resident microglia and immune cells that infiltrate the retina in disease such as age-related macular degeneration (AMD), a degenerative retinal disease, and uveitis, an inflammatory eye disease. Activation of P2X7 is involved in several physiological and pathological processes: phagocytosis, activation of the inflammasome NLRP3, release of pro-inflammatory mediators and cell death. The aim of this review is to discuss the potential involvement of P2X7 in the development of AMD and uveitis.
Collapse
|
36
|
Hata M, Andriessen EMMA, Hata M, Diaz-Marin R, Fournier F, Crespo-Garcia S, Blot G, Juneau R, Pilon F, Dejda A, Guber V, Heckel E, Daneault C, Calderon V, Des Rosiers C, Melichar HJ, Langmann T, Joyal JS, Wilson AM, Sapieha P. Past history of obesity triggers persistent epigenetic changes in innate immunity and exacerbates neuroinflammation. Science 2023; 379:45-62. [PMID: 36603072 DOI: 10.1126/science.abj8894] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Age-related macular degeneration is a prevalent neuroinflammatory condition and a major cause of blindness driven by genetic and environmental factors such as obesity. In diseases of aging, modifiable factors can be compounded over the life span. We report that diet-induced obesity earlier in life triggers persistent reprogramming of the innate immune system, lasting long after normalization of metabolic abnormalities. Stearic acid, acting through Toll-like receptor 4 (TLR4), is sufficient to remodel chromatin landscapes and selectively enhance accessibility at binding sites for activator protein-1 (AP-1). Myeloid cells show less oxidative phosphorylation and shift to glycolysis, ultimately leading to proinflammatory cytokine transcription, aggravation of pathological retinal angiogenesis, and neuronal degeneration associated with loss of visual function. Thus, a past history of obesity reprograms mononuclear phagocytes and predisposes to neuroinflammation.
Collapse
Affiliation(s)
- Masayuki Hata
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec H1T 2M4, Canada.,Department of Biochemistry and Molecular Medicine, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec H1T 2M4, Canada
| | - Elisabeth M M A Andriessen
- Department of Biomedical Sciences, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec H1T 2M4, Canada
| | - Maki Hata
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec H1T 2M4, Canada
| | - Roberto Diaz-Marin
- Department of Biochemistry and Molecular Medicine, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec H1T 2M4, Canada
| | - Frédérik Fournier
- Department of Biochemistry and Molecular Medicine, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec H1T 2M4, Canada
| | - Sergio Crespo-Garcia
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec H1T 2M4, Canada.,Department of Biochemistry and Molecular Medicine, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec H1T 2M4, Canada
| | - Guillaume Blot
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec H1T 2M4, Canada.,Department of Biochemistry and Molecular Medicine, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec H1T 2M4, Canada
| | - Rachel Juneau
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec H1T 2M4, Canada
| | - Frédérique Pilon
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec H1T 2M4, Canada
| | - Agnieszka Dejda
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec H1T 2M4, Canada
| | - Vera Guber
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec H1T 2M4, Canada
| | - Emilie Heckel
- Departments of Pediatrics, Ophthalmology, and Pharmacology, Centre Hospitalier Universitaire Ste-Justine Research Center, Montreal, Quebec H3T 1C5, Canada
| | - Caroline Daneault
- Department of Nutrition, University of Montreal, Montreal, Quebec, Plateforme métabolomique de l'Institut de Cardiologie de Montréal, Montreal, Quebec H3C 3J7, Canada
| | - Virginie Calderon
- Bioinformatics & Molecular Biology Core Facility, Institut de Recherches Cliniques de Montréal, Montreal, Quebec H2W 1R7, Canada
| | - Christine Des Rosiers
- Department of Nutrition, University of Montreal, Montreal, Quebec, Plateforme métabolomique de l'Institut de Cardiologie de Montréal, Montreal, Quebec H3C 3J7, Canada
| | - Heather J Melichar
- Department of Medicine, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec H1T 2M4, Canada
| | - Thomas Langmann
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Jean-Sebastien Joyal
- Departments of Pediatrics, Ophthalmology, and Pharmacology, Centre Hospitalier Universitaire Ste-Justine Research Center, Montreal, Quebec H3T 1C5, Canada
| | - Ariel M Wilson
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec H1T 2M4, Canada
| | - Przemyslaw Sapieha
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec H1T 2M4, Canada.,Department of Biochemistry and Molecular Medicine, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec H1T 2M4, Canada
| |
Collapse
|
37
|
Choudhary M, Malek G. CD68: Potential Contributor to Inflammation and RPE Cell Dystrophy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1415:207-213. [PMID: 37440035 DOI: 10.1007/978-3-031-27681-1_30] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Abstract
Age-related macular degeneration (AMD) is the leading cause of visual impairment in the elderly in developed countries. It is a complex, multifactorial, progressive disease with diverse molecular pathways, including inflammation, regulating its pathogenesis. The myeloid marker CD68 is a protein highly expressed in circulating and tissue macrophages. Recent observations of immune markers in human AMD tissues have varied with some finding ectopic RPE cells in advanced AMD and others noting negligible numbers of CD68-positive cells. Additionally, animal models of retinal degeneration have shown upregulation of CD68, in a protective population of retinal microglia. Herein, we review the potential role of CD68 in regulating RPE health and inflammation in the sub-retinal space and discuss observations on its localization in a mouse model that presents with AMD-like features.
Collapse
Affiliation(s)
- Mayur Choudhary
- Department of Ophthalmology, Albert Eye Research Institute, Duke University School of Medicine, Durham, NC, USA
| | - Goldis Malek
- Department of Ophthalmology, Albert Eye Research Institute, Duke University School of Medicine, Durham, NC, USA.
- Department of Pathology, Albert Eye Research Institute, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
38
|
Vienola KV, Lejoyeux R, Gofas-Salas E, Snyder VC, Zhang M, Dansingani KK, Sahel JA, Chhablani J, Rossi EA. Autofluorescent hyperreflective foci on infrared autofluorescence adaptive optics ophthalmoscopy in central serous chorioretinopathy. Am J Ophthalmol Case Rep 2022; 28:101741. [PMID: 36345414 PMCID: PMC9636439 DOI: 10.1016/j.ajoc.2022.101741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 10/12/2022] [Accepted: 10/22/2022] [Indexed: 11/06/2022] Open
Abstract
Purpose To test the hypothesis that hyperreflective foci in central serous chorioretinopathy (CSCR) are autofluorescent and may represent macrophages that have engulfed outer retinal fluorophores from the retinal pigment epithelium (RPE) and photoreceptors. Methods Enrolled subjects underwent spectral domain and swept-source optical coherence tomography, adaptive optics flood-illumination, and adaptive optics scanning laser ophthalmoscopy (AOSLO), including near-infrared autofluorescence (AO-IRAF). For the AO-IRAF imaging, retinal fluorophores were excited using 795 nm light and collected in an emission band from 814 to 850 nm. Results In 2 of 3 eyes, a hyperautofluorescent signal was detected with an elliptical shape and punctate, granular aspects surrounded by a hypoautofluorescent halo. The size of these structures in the active case was measured to be 17 ± 4 μm in diameter, with at least 45 individual hyperautofluorescent foci identified from the AO-IRAF montage in the active stage of patient 2. In the asymptomatic case there were fewer structures visible (∼10) and their size was smaller (11 ± 4 μm). These hyper-AF foci were colocalized with hyperreflective foci on OCT and visible in simultaneously acquired confocal AOSLO images in active stage. The hyperautofluorescent foci in the patient with active CSCR disappeared coincident with clinical resolution. Conclusion and importance We show here the first AO-IRAF images from patients with CSCR, demonstrating hyper-autofluorescent punctate foci, colocalized with hyper-reflective foci on confocal AOSLO images and in OCT. The autofluorescence of these foci may be driven by the accumulation of photoreceptor and RPE fluorophores within macrophages during the active stage of the disease.
Collapse
Affiliation(s)
- Kari V. Vienola
- University of Pittsburgh, Department of Ophthalmology, School of Medicine, 4200 Fifth Ave, Pittsburgh, PA, USA
- Corresponding author. Laboratory of Biophysics, Institute of Biomedicine University of Turku Tykistönkatu 6A, Turku, Finland.
| | - Raphael Lejoyeux
- University of Pittsburgh, Department of Ophthalmology, School of Medicine, 4200 Fifth Ave, Pittsburgh, PA, USA
- Rothschild Foundation Hospital, 29 rue Manin, Paris, France
| | - Elena Gofas-Salas
- University of Pittsburgh, Department of Ophthalmology, School of Medicine, 4200 Fifth Ave, Pittsburgh, PA, USA
| | - Valerie C. Snyder
- University of Pittsburgh, Department of Ophthalmology, School of Medicine, 4200 Fifth Ave, Pittsburgh, PA, USA
| | - Min Zhang
- University of Pittsburgh, Department of Ophthalmology, School of Medicine, 4200 Fifth Ave, Pittsburgh, PA, USA
| | - Kunal K. Dansingani
- University of Pittsburgh, Department of Ophthalmology, School of Medicine, 4200 Fifth Ave, Pittsburgh, PA, USA
| | - José-Alain Sahel
- University of Pittsburgh, Department of Ophthalmology, School of Medicine, 4200 Fifth Ave, Pittsburgh, PA, USA
| | - Jay Chhablani
- University of Pittsburgh, Department of Ophthalmology, School of Medicine, 4200 Fifth Ave, Pittsburgh, PA, USA
| | - Ethan A. Rossi
- University of Pittsburgh, Department of Ophthalmology, School of Medicine, 4200 Fifth Ave, Pittsburgh, PA, USA
- University of Pittsburgh, Department of Bioengineering, Swanson School of Engineering, Pittsburgh, PA, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
39
|
Innate immunity dysregulation in aging eye and therapeutic interventions. Ageing Res Rev 2022; 82:101768. [PMID: 36280210 DOI: 10.1016/j.arr.2022.101768] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 09/29/2022] [Accepted: 10/20/2022] [Indexed: 01/31/2023]
Abstract
The prevalence of eye diseases increases considerably with age, resulting in significant vision impairment. Although the pathobiology of age-related eye diseases has been studied extensively, the contribution of immune-related changes due to aging remains elusive. In the eye, tissue-resident cells and infiltrating immune cells regulate innate responses during injury or infection. But due to aging, these cells lose their protective functions and acquire pathological phenotypes. Thus, dysregulated ocular innate immunity in the elderly increases the susceptibility and severity of eye diseases. Herein, we emphasize the impact of aging on the ocular innate immune system in the pathogenesis of infectious and non-infectious eye diseases. We discuss the role of age-related alterations in cellular metabolism, epigenetics, and cellular senescence as mechanisms underlying altered innate immune functions. Finally, we describe approaches to restore protective innate immune functions in the aging eye. Overall, the review summarizes our current understanding of innate immune functions in eye diseases and their dysregulation during aging.
Collapse
|
40
|
Pameijer EM, Heus P, Damen JAA, Spijker R, Hooft L, Ringens PJ, Imhof SM, van Leeuwen R. What did we learn in 35 years of research on nutrition and supplements for age-related macular degeneration: a systematic review. Acta Ophthalmol 2022; 100:e1541-e1552. [PMID: 35695158 PMCID: PMC9796889 DOI: 10.1111/aos.15191] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 05/14/2022] [Indexed: 01/07/2023]
Abstract
The aim of this paper is to summarize all available evidence from systematic reviews, randomized controlled trials (RCTs) and comparative nonrandomized studies (NRS) on the association between nutrition and antioxidant, vitamin, and mineral supplements and the development or progression of age-related macular degeneration (AMD). The Cochrane Database of Systematic Reviews, Cochrane register CENTRAL, MEDLINE and Embase were searched and studies published between January 2015 and May 2021 were included. The certainty of evidence was assessed according to the GRADE methodology. The main outcome measures were development of AMD, progression of AMD, and side effects. We included 7 systematic reviews, 7 RCTs, and 13 NRS. A high consumption of specific nutrients, i.e. β-carotene, lutein and zeaxanthin, copper, folate, magnesium, vitamin A, niacin, vitamin B6, vitamin C, docosahexaenoic acid, and eicosapentaenoic acid, was associated with a lower risk of progression of early to late AMD (high certainty of evidence). Use of antioxidant supplements and adherence to a Mediterranean diet, characterized by a high consumption of vegetables, whole grains, and nuts and a low consumption of red meat, were associated with a decreased risk of progression of early to late AMD (moderate certainty of evidence). A high consumption of alcohol was associated with a higher risk of developing AMD (moderate certainty of evidence). Supplementary vitamin C, vitamin E, or β-carotene were not associated with the development of AMD, and supplementary omega-3 fatty acids were not associated with progression to late AMD (high certainty of evidence). Research in the last 35 years included in our overview supports that a high intake of specific nutrients, the use of antioxidant supplements and adherence to a Mediterranean diet decrease the risk of progression of early to late AMD.
Collapse
Affiliation(s)
| | - Pauline Heus
- Cochrane Netherlands and Julius Center for Health Sciences and Primary Care, UMC UtrechtUtrecht UniversityThe Netherlands
| | - Johanna A. A. Damen
- Cochrane Netherlands and Julius Center for Health Sciences and Primary Care, UMC UtrechtUtrecht UniversityThe Netherlands
| | - René Spijker
- Cochrane Netherlands and Julius Center for Health Sciences and Primary Care, UMC UtrechtUtrecht UniversityThe Netherlands
| | - Lotty Hooft
- Cochrane Netherlands and Julius Center for Health Sciences and Primary Care, UMC UtrechtUtrecht UniversityThe Netherlands
| | - Peter J. Ringens
- Department of OphthalmologyMaastricht University Medical CenterMaastrichtThe Netherlands
| | | | | |
Collapse
|
41
|
Wong JHC, Ma JYW, Jobling AI, Brandli A, Greferath U, Fletcher EL, Vessey KA. Exploring the pathogenesis of age-related macular degeneration: A review of the interplay between retinal pigment epithelium dysfunction and the innate immune system. Front Neurosci 2022; 16:1009599. [PMID: 36408381 PMCID: PMC9670140 DOI: 10.3389/fnins.2022.1009599] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 10/12/2022] [Indexed: 07/30/2023] Open
Abstract
Age-related macular degeneration (AMD) is a leading cause of irreversible vision loss in the older population. Classical hallmarks of early and intermediate AMD are accumulation of drusen, a waste deposit formed under the retina, and pigmentary abnormalities in the retinal pigment epithelium (RPE). When the disease progresses into late AMD, vision is affected due to death of the RPE and the light-sensitive photoreceptors. The RPE is essential to the health of the retina as it forms the outer blood retinal barrier, which establishes ocular immune regulation, and provides support for the photoreceptors. Due to its unique anatomical position, the RPE can communicate with the retinal environment and the systemic immune environment. In AMD, RPE dysfunction and the accumulation of drusen drive the infiltration of retinal and systemic innate immune cells into the outer retina. While recruited endogenous or systemic mononuclear phagocytes (MPs) contribute to the removal of noxious debris, the accumulation of MPs can also result in chronic inflammation and contribute to AMD progression. In addition, direct communication and indirect molecular signaling between MPs and the RPE may promote RPE cell death, choroidal neovascularization and fibrotic scarring that occur in late AMD. In this review, we explore how the RPE and innate immune cells maintain retinal homeostasis, and detail how RPE dysfunction and aberrant immune cell recruitment contribute to AMD pathogenesis. Evidence from AMD patients will be discussed in conjunction with data from preclinical models, to shed light on future therapeutic targets for the treatment of AMD.
Collapse
|
42
|
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: 0.7] [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
|
43
|
Ren C, Yu J. Potential gene identification and pathway crosstalk analysis of age-related macular degeneration. Front Genet 2022; 13:992328. [PMID: 36147504 PMCID: PMC9486309 DOI: 10.3389/fgene.2022.992328] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 08/08/2022] [Indexed: 11/28/2022] Open
Abstract
Age-related macular degeneration (AMD), the most prevalent visual disorder among the elderly, is confirmed as a multifactorial disease. Studies demonstrated that genetic factors play an essential role in its pathogenesis. Our study aimed to make a relatively comprehensive study about biological functions of AMD related genes and crosstalk of their enriched pathways. 1691 AMD genetic studies were reviewed, GO enrichment and pathway crosstalk analyses were conducted to elucidate the biological features of these genes and to demonstrate the pathways that these genes participate. Moreover, we identified novel AMD-specific genes using shortest path algorithm in the context of human interactome. We retrieved 176 significantly AMD-related genes. GO results showed that the most significant term in each of these three GO categories was: signaling receptor binding (PBH = 4.835 × 10−7), response to oxygen-containing compound (PBH = 2.764 × 10−21), and extracellular space (PBH = 2.081 × 10−19). The pathway enrichment analysis showed that complement pathway is the most enriched. The pathway crosstalk study showed that the pathways could be divided into two main modules. These two modules were connected by cytokine-cytokine receptor interaction pathway. 42 unique genes potentially participating AMD development were obtained. The aberrant expression of the mRNA of FASN and LRP1 were validated in AMD cell and mouse models. Collectively, our study carried out a comprehensive analysis based on genetic association study of AMD and put forward several evidence-based genes for future study of AMD.
Collapse
|
44
|
Molins B, Mesquida M, Adan A. Bioengineering approaches for modelling retinal pathologies of the outer blood-retinal barrier. Prog Retin Eye Res 2022:101097. [PMID: 35840488 DOI: 10.1016/j.preteyeres.2022.101097] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 05/31/2022] [Accepted: 06/29/2022] [Indexed: 11/18/2022]
Abstract
Alterations of the junctional complex of the outer blood-retinal barrier (oBRB), which is integrated by the close interaction of the retinal pigment epithelium, the Bruch's membrane, and the choriocapillaris, contribute to the loss of neuronal signalling and subsequent vision impairment in several retinal inflammatory disorders such as age-related macular degeneration and diabetic retinopathy. Reductionist approaches into the mechanisms that underlie such diseases have been hindered by the absence of adequate in vitro models using human cells to provide the 3D dynamic architecture that enables expression of the in vivo phenotype of the oBRB. Conventional in vitro cell models are based on 2D monolayer cellular cultures, unable to properly recapitulate the complexity of living systems. The main drawbacks of conventional oBRB models also emerge from the cell sourcing, the lack of an appropriate Bruch's membrane analogue, and the lack of choroidal microvasculature with flow. In the last years, the advent of organ-on-a-chip, bioengineering, and stem cell technologies is providing more advanced 3D models with flow, multicellularity, and external control over microenvironmental properties. By incorporating additional biological complexity, organ-on-a-chip devices can mirror physiologically relevant properties of the native tissue while offering additional set ups to model and study disease. In this review we first examine the current understanding of oBRB biology as a functional unit, highlighting the coordinated contribution of the different components to barrier function in health and disease. Then we describe recent advances in the use of pluripotent stem cells-derived retinal cells, Bruch's membrane analogues, and co-culture techniques to recapitulate the oBRB. We finally discuss current advances and challenges of oBRB-on-a-chip technologies for disease modelling.
Collapse
Affiliation(s)
- Blanca Molins
- Group of Ocular Inflammation: Clinical and Experimental Studies, Institut d'Investigacions Biomèdiques Agustí Pi I Sunyer (IDIBAPS), C/ Sabino de Arana 1, 08028, Barcelona, Spain.
| | - Marina Mesquida
- Group of Ocular Inflammation: Clinical and Experimental Studies, Institut d'Investigacions Biomèdiques Agustí Pi I Sunyer (IDIBAPS), C/ Sabino de Arana 1, 08028, Barcelona, Spain; Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Alfredo Adan
- Group of Ocular Inflammation: Clinical and Experimental Studies, Institut d'Investigacions Biomèdiques Agustí Pi I Sunyer (IDIBAPS), C/ Sabino de Arana 1, 08028, Barcelona, Spain; Instituto Clínic de Oftalmología, Hospital Clínic Barcelona, C/ Sabino de Arana 1, 08028, Barcelona, Spain
| |
Collapse
|
45
|
Paques M, Norberg N, Chaumette C, Sennlaub F, Rossi E, Borella Y, Grieve K. Long Term Time-Lapse Imaging of Geographic Atrophy: A Pilot Study. Front Med (Lausanne) 2022; 9:868163. [PMID: 35814763 PMCID: PMC9257004 DOI: 10.3389/fmed.2022.868163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 05/26/2022] [Indexed: 11/21/2022] Open
Abstract
Geographic atrophy (GA), the late stage of age-related macular degeneration, is a major cause of visual disability whose pathophysiology remains largely unknown. Modern fundus imaging and histology revealed the complexity of the cellular changes that accompanies atrophy. Documenting the activity of the disease in the margins of atrophy, where the transition from health to disease occurs, would contribute to a better understanding of the progression of GA. Time-lapse imaging facilitates the identification of structural continuities in changing environments. In this retrospective pilot study, we documented the long-term changes in atrophy margins by time-lapse imaging of infrared scanning laser ophthalmoscopy (SLO) and optical coherence tomography (OCT) images in 6 cases of GA covering a mean period of 32.8 months (range, 18–72). The mean interval between imaging sessions was 2.4 months (range, 1.4–3.8). By viewing time-lapse sequences we observed extensive changes in the pattern of marginal hyperreflective spots, which associated fragmentation, increase and/or disappearance. Over the entire span of the follow-up, the most striking changes were those affecting hyperreflective spots closest to margins of atrophy, on the non-atrophic side of the retina; a continuum between the successive positions of some of the hyperreflective spots was detected, both by SLO and OCT. This continuum in their successive positions resulted in a subjective impression of a centrifugal motion of hyperreflective spots ahead of atrophy progression. Such mobilization of hyperreflective spots was detected up to several hundred microns away from atrophic borders. Such process is likely to reflect the inflammatory and degenerative process underlying GA progression and hence deserves further investigations. These results highlight the interest of multimodal time-lapse imaging to document cell-scale dynamics during progression of GA.
Collapse
Affiliation(s)
- Michel Paques
- Paris Eye Imaging Group, Clinical Investigation Center 1423, Quinze-Vingts Hospital, INSERM-DHOS, Sorbonne Université, INSERM, Paris, France
- Institut de la Vision, Paris, France
| | - Nathaniel Norberg
- Paris Eye Imaging Group, Clinical Investigation Center 1423, Quinze-Vingts Hospital, INSERM-DHOS, Sorbonne Université, INSERM, Paris, France
| | - Céline Chaumette
- Paris Eye Imaging Group, Clinical Investigation Center 1423, Quinze-Vingts Hospital, INSERM-DHOS, Sorbonne Université, INSERM, Paris, France
| | | | - Ethan Rossi
- Department of Ophthalmology, The University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Ysé Borella
- Paris Eye Imaging Group, Clinical Investigation Center 1423, Quinze-Vingts Hospital, INSERM-DHOS, Sorbonne Université, INSERM, Paris, France
- Institut de la Vision, Paris, France
| | - Kate Grieve
- Paris Eye Imaging Group, Clinical Investigation Center 1423, Quinze-Vingts Hospital, INSERM-DHOS, Sorbonne Université, INSERM, Paris, France
- Institut de la Vision, Paris, France
| |
Collapse
|
46
|
Liisborg C, Skov V, Kjær L, Hasselbalch HC, Lykke Sørensen T. Lower CXCR3 expression in both patients with neovascular AMD and advanced stages of chronic myeloproliferative blood cancers. PLoS One 2022; 17:e0269960. [PMID: 35709177 PMCID: PMC9202899 DOI: 10.1371/journal.pone.0269960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 05/31/2022] [Indexed: 11/19/2022] Open
Abstract
Purpose
Peripheral T cell CXCR3 expression has been found uniquely lower in patients having neovascular age-related macular degeneration (nAMD) than in healthy individuals. The CXCR3-axis has been shown to have angiostatic and antifibrotic properties. We have recently investigated systemic markers in patients with myeloproliferative neoplasms (MPNs) because of their higher prevalence of AMD, and we have observed higher systemic chronic low-grade inflammation and immunosenescence signs in MPNs with drusen (MPNd) compared to those with normal retinas (MPNn). The MPNs evolve in a biological continuum from early cancer-stages (essential thrombocytosis, polycythemia vera) to the advanced myelofibrosis stage. Especially myelofibrosis is characterized by bone marrow angiogenesis and fibrosis, similarly to retinal observations in nAMD. We speculate if we can find lower CXCR3 expression in MPNs, particularly myelofibrosis and if differences are seen between MPNd and MPNn. We also wanted to compare expression in nAMD and intermediate (i)AMD.
Methods
Patients in this cross-sectional study were 29 nAMD, 28 iAMD, 35 MPNd, and 27 MPNn. We performed flowcytometry on blood to measure CXCR3 expression.
Results
CD8+CXCR3 expression in nAMD was 6,1%, significantly lower than in iAMD 16%, MPNd 11%, MPNn 12% (p-values<0.05). Similar results were seen for CD4+CXCR3 expression. We also found CXCR3 expression decreasing over the MPN-continuum. For instance, in myelofibrosis, intermediate monocytes expression was 6.2%, significantly lower than 18% in ET and 18% in PV (p-values<0.05).
Conclusions
We find CXCR3 downregulation on T-cells and some monocyte subset in nAMD compared to iAMD, MPNd, and MPNn, in line with previous nAMD studies. We also find CXCR3 downregulation in most monocyte subsets over the MPN continuum. Systemic leukocyte CXCR3 expression could both be involved in changes seen in the retina and the bone marrow. Further understanding the CXCR3-axis in AMD and MPNs may elucidate underlying pathogenic mechanisms and reveal new targets for treatment.
Collapse
Affiliation(s)
- Charlotte Liisborg
- Department of Ophthalmology, Zealand University Hospital, Roskilde, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- * E-mail:
| | - Vibe Skov
- Department of Hematology, Zealand University Hospital, Roskilde, Denmark
| | - Lasse Kjær
- Department of Hematology, Zealand University Hospital, Roskilde, Denmark
| | - Hans Carl Hasselbalch
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Hematology, Zealand University Hospital, Roskilde, Denmark
| | - Torben Lykke Sørensen
- Department of Ophthalmology, Zealand University Hospital, Roskilde, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
47
|
Fan W, Huang W, Chen J, Li N, Mao L, Hou S. Retinal microglia: Functions and diseases. Immunology 2022; 166:268-286. [PMID: 35403700 DOI: 10.1111/imm.13479] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 01/26/2022] [Accepted: 02/22/2022] [Indexed: 11/29/2022] Open
Affiliation(s)
- Wei Fan
- The First Affiliated Hospital of Chongqing Medical University Chongqing China
- Chongqing Key Laboratory of Ophthalmology Chongqing China
- Chongqing Eye Institute Chongqing China
- Chongqing Branch of National Clinical Research Center for Ocular Diseases Chongqing China
| | - Weidi Huang
- The First Affiliated Hospital of Chongqing Medical University Chongqing China
- Department of Ophthalmology, Second Xiangya Hospital Central South University Changsha Hunan China
| | - Jiayi Chen
- The First Affiliated Hospital of Chongqing Medical University Chongqing China
| | - Na Li
- College of Basic Medicine Chongqing Medical University Chongqing China
| | - Liming Mao
- Department of Immunology School of Medicine, Nantong University, 19 Qixiu Road Nantong Jiangsu China
| | - Shengping Hou
- The First Affiliated Hospital of Chongqing Medical University Chongqing China
- Chongqing Key Laboratory of Ophthalmology Chongqing China
- Chongqing Eye Institute Chongqing China
- Chongqing Branch of National Clinical Research Center for Ocular Diseases Chongqing China
| |
Collapse
|
48
|
Liu W, Jiang X, Li X, Sun K, Yang Y, Yang M, Li S, Zhu X. LMBR1L regulates proliferation and migration of endothelial cells through Norrin/β-catenin signaling. J Cell Sci 2022; 135:274701. [PMID: 35146515 DOI: 10.1242/jcs.259468] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 02/07/2022] [Indexed: 11/20/2022] Open
Abstract
Precise Norrin/β-catenin signaling is critical for proper angiogenesis. Dysregulation of the signaling leads various diseases, of which retinal exudative vitreoretinopathy is the most prevalent. Here, we used global knockout mouse model to show that endothelial cells-derived limb region 1 like (LMBR1L), a transmembrane protein of unknown function in angiogenesis, is essential for retinal vascular development. In vitro experiments revealed that LMBR1L depletion resulted in aberrant activation of Norrin/β-catenin signaling pathway via decreased ubiquitination of FZD4, increased Norrin co-receptor LRP5 and p-GSK3β-Ser9 expression level, which caused accumulation of β-catenin. Moreover, inhibition of LMBR1L in human retinal microvascular endothelial cells (HRECs) caused increased proliferation ability and defective cell migration, which might due to upregulated expression levels of the AJ components. Treatment of p-GSK3β-Ser9 inhibitor AR-A014418 restored the phenotypes in LMBR1L-null HRECs, which further demonstrated the important regulatory role of LMBR1L in Norrin/β-catenin signaling pathway. Taken together, our data unravels an essential role of LMBR1L in angiogenesis.
Collapse
Affiliation(s)
- Wenjing Liu
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Department of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, China.,Key Laboratory of Tibetan Medicine Research, Chinese Academy of Sciences and Qinghai Provincial Key Laboratory of Tibetan Medicine Research, Northwest Institute of Plateau Biology, Xining, Qinghai 810008, China.,Research Unit for Blindness Prevention of the Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, 610072, China
| | - Xiaoyan Jiang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Department of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, China
| | - Xiao Li
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Department of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, China
| | - Kuanxiang Sun
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Department of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, China
| | - Yeming Yang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Department of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, China
| | - Mu Yang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Department of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, China.,Research Unit for Blindness Prevention of the Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, 610072, China
| | - Shujin Li
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Department of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, China.,Research Unit for Blindness Prevention of the Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, 610072, China
| | - Xianjun Zhu
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Department of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, China.,Key Laboratory of Tibetan Medicine Research, Chinese Academy of Sciences and Qinghai Provincial Key Laboratory of Tibetan Medicine Research, Northwest Institute of Plateau Biology, Xining, Qinghai 810008, China.,Research Unit for Blindness Prevention of the Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, 610072, China.,Departement of Ophthalmology, First People's Hospital of Shangqiu, Shangqiu, Henan, 476000, China
| |
Collapse
|
49
|
Clare AJ, Liu J, Copland DA, Theodoropoulou S, Dick AD. Unravelling the therapeutic potential of IL-33 for atrophic AMD. Eye (Lond) 2022; 36:266-272. [PMID: 34531552 PMCID: PMC8807696 DOI: 10.1038/s41433-021-01725-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/08/2021] [Accepted: 07/27/2021] [Indexed: 02/08/2023] Open
Abstract
Age-related macular degeneration (AMD), a degenerative disease affecting the retinal pigment epithelium (RPE) and photoreceptors in the macula, is the leading cause of central blindness in the elderly. AMD progresses to advanced stages of the disease, atrophic AMD (aAMD), or in 15% of cases "wet" or neovascular AMD (nAMD), associated with substantial vision loss. Whilst there has been advancement in therapies treating nAMD, to date, there are no licenced effective treatments for the 85% affected by aAMD, with disease managed by changes to diet, vitamin supplements, and regular monitoring. AMD has a complex pathogenesis, involving highly integrated and common age-related disease pathways, including dysregulated complement/inflammation, impaired autophagy, and oxidative stress. The intricacy of AMD pathogenesis makes therapeutic development challenging and identifying a target that combats the converging disease pathways is essential to provide a globally effective treatment. Interleukin-33 is a cytokine, classically known for the proinflammatory role it plays in allergic disease. Recent evidence across degenerative and inflammatory disease conditions reveals a diverse immune-modulatory role for IL-33, with promising therapeutic potential. Here, we will review IL-33 function in disease and discuss the future potential for this homeostatic cytokine in treating AMD.
Collapse
Affiliation(s)
- Alison J. Clare
- grid.5337.20000 0004 1936 7603Academic Unit of Ophthalmology, Translational Health Sciences, University of Bristol, Bristol, UK
| | - Jian Liu
- grid.5337.20000 0004 1936 7603Academic Unit of Ophthalmology, Translational Health Sciences, University of Bristol, Bristol, UK
| | - David A. Copland
- grid.5337.20000 0004 1936 7603Academic Unit of Ophthalmology, Translational Health Sciences, University of Bristol, Bristol, UK
| | - Sofia Theodoropoulou
- grid.5337.20000 0004 1936 7603Academic Unit of Ophthalmology, Translational Health Sciences, University of Bristol, Bristol, UK
| | - Andrew D. Dick
- grid.5337.20000 0004 1936 7603Academic Unit of Ophthalmology, Translational Health Sciences, University of Bristol, Bristol, UK ,grid.5337.20000 0004 1936 7603School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK ,grid.439257.e0000 0000 8726 5837NIHR Biomedical Research Centre of Ophthalmology, Moorfields Eye Hospital, London, UK ,grid.83440.3b0000000121901201UCL Institute of Ophthalmology, London, UK
| |
Collapse
|
50
|
Cho HM, Jo YD, Choung SY. Protective Effects of Spirulina maxima against Blue Light-Induced Retinal Damages in A2E-Laden ARPE-19 Cells and Balb/c Mice. Nutrients 2022; 14:nu14030401. [PMID: 35276761 PMCID: PMC8840079 DOI: 10.3390/nu14030401] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/13/2022] [Accepted: 01/14/2022] [Indexed: 12/18/2022] Open
Abstract
Age-related macular degeneration (AMD) is a significant visual impairment in older people, and there is no treatment for dry AMD. Spirulina maxima (S. maxima), a cyanobacterium, has inhibitory effects against oxidative stress. However, the protective effects of S. maxima and its underlying mechanisms on blue light (BL)-caused macular degeneration are unknown. We aimed to investigate the protective effects of S. maxima on blue light-caused retinal damage and demonstrate its underlying mechanisms in human retinal pigment epithelial (ARPE-19) cells and Balb/c retinas. Additionally, the active component of S. maxima was examined in the RPE cells. In vitro, S. maxima decreased BL-induced RPE cell death by inhibiting reactive oxygen species (ROS) production. S. maxima inhibited BL-induced inflammation via regulating the NF-κB pathway, inflammatory-related gene expression, and the apoptosis pathway in RPE cells. In vivo, administration of S. maxima inhibited BL-induced retinal degeneration by restoring the thicknesses of whole retina, ONL (outer nuclear layer), INL (inner nuclear layer), and PL (photoreceptor layer) by BL exposure. Phycocyanin exerted protective effects in the pre-and post-treatment system. Therefore, S. maxima could be a potential nutraceutical approach to intercept the patho-physiological processes leading to dry AMD and advancement to wet AMD. Moreover, phycocyanin was a major active compound of S. maxima. These findings need to be investigated in human studies, particularly through a clinical trial.
Collapse
Affiliation(s)
- Hye-Mi Cho
- Department of Biomedical and Pharmaceutical Sciences, Graduate School, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea;
| | - Ye-Dam Jo
- Department of Life and Nanopharmaceutical Sciences, Graduate School, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea;
| | - Se-Young Choung
- Department of Biomedical and Pharmaceutical Sciences, Graduate School, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea;
- Department of Preventive Pharmacy and Toxicology, College of Pharmacy, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea
- Correspondence: ; Tel.: +82-2-961-9198; Fax: +82-2-961-0372
| |
Collapse
|