1
|
Venanzi AW, McGee LD, Hackam AS. Evaluating the Evidence for Neuroprotective and Axonal Regenerative Activities of Different Inflammatory Cell Types After Optic Nerve Injury. Mol Neurobiol 2025; 62:6212-6227. [PMID: 39738875 PMCID: PMC11953096 DOI: 10.1007/s12035-024-04679-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 12/19/2024] [Indexed: 01/02/2025]
Abstract
The optic nerve contains retinal ganglion cell (RGC) axons and functions to transmit visual stimuli to the brain. Injury to the optic nerve from ischemia, trauma, or disease leads to retrograde axonal degeneration and subsequent RGC dysfunction and death, causing irreversible vision loss. Inflammatory responses to neurological damage and axonal injuries in the central nervous system (CNS) are typically harmful to neurons and prevent recovery. However, recent evidence indicates that certain inflammatory cell types and signaling pathways are protective after optic nerve injury and promote RGC survival and axonal regeneration. The objective of this review is to examine the evidence for diverse effects of inflammatory cell types on the retina and optic nerve after injury. Additionally, we highlight promising avenues for further research.
Collapse
Affiliation(s)
- Alexander W Venanzi
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, 1638 NW 10Th Ave, Rm 404, Miami, FL, 33136, USA
| | - Laura D McGee
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, 1638 NW 10Th Ave, Rm 404, Miami, FL, 33136, USA
| | - Abigail S Hackam
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, 1638 NW 10Th Ave, Rm 404, Miami, FL, 33136, USA.
| |
Collapse
|
2
|
Bhattacharya S, Deka J, Avallone T, Todd L. The neuroimmune interface in retinal regeneration. Prog Retin Eye Res 2025; 106:101361. [PMID: 40287050 DOI: 10.1016/j.preteyeres.2025.101361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 04/12/2025] [Accepted: 04/23/2025] [Indexed: 04/29/2025]
Abstract
Retinal neurodegeneration leads to irreversible blindness due to the mammalian nervous system's inability to regenerate lost neurons. Efforts to regenerate retina involve two main strategies: stimulating endogenous cells to reprogram into neurons or transplanting stem-cell derived neurons into the degenerated retina. However, both approaches must overcome a major barrier in getting new neurons to grow back down the optic nerve and connect to appropriate visual targets in environments that differ significantly from developmental conditions. While immune privilege has historically been associated with the central nervous system, an emerging literature highlights the active role of immune cells in shaping neurodegeneration and regeneration. This review explores the neuroimmune interface in retinal repair, dissecting how immune interactions influence glial reprogramming, transplantation outcomes, and axonal regeneration. By integrating insights from regenerative species with mammalian models, we highlight novel immunomodulatory strategies to optimize retinal regeneration.
Collapse
Affiliation(s)
- Sucheta Bhattacharya
- Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, Syracuse, NY, 13210, USA; Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
| | - Jugasmita Deka
- Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, Syracuse, NY, 13210, USA; Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
| | - Thomas Avallone
- Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
| | - Levi Todd
- Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, Syracuse, NY, 13210, USA; Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA.
| |
Collapse
|
3
|
Yin Z, Kang J, Cheng X, Gao H, Huo S, Xu H. Investigating Müller glia reprogramming in mice: a retrospective of the last decade, and a look to the future. Neural Regen Res 2025; 20:946-959. [PMID: 38989930 PMCID: PMC11438324 DOI: 10.4103/nrr.nrr-d-23-01612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 02/05/2024] [Indexed: 07/12/2024] Open
Abstract
Müller glia, as prominent glial cells within the retina, plays a significant role in maintaining retinal homeostasis in both healthy and diseased states. In lower vertebrates like zebrafish, these cells assume responsibility for spontaneous retinal regeneration, wherein endogenous Müller glia undergo proliferation, transform into Müller glia-derived progenitor cells, and subsequently regenerate the entire retina with restored functionality. Conversely, Müller glia in the mouse and human retina exhibit limited neural reprogramming. Müller glia reprogramming is thus a promising strategy for treating neurodegenerative ocular disorders. Müller glia reprogramming in mice has been accomplished with remarkable success, through various technologies. Advancements in molecular, genetic, epigenetic, morphological, and physiological evaluations have made it easier to document and investigate the Müller glia programming process in mice. Nevertheless, there remain issues that hinder improving reprogramming efficiency and maturity. Thus, understanding the reprogramming mechanism is crucial toward exploring factors that will improve Müller glia reprogramming efficiency, and for developing novel Müller glia reprogramming strategies. This review describes recent progress in relatively successful Müller glia reprogramming strategies. It also provides a basis for developing new Müller glia reprogramming strategies in mice, including epigenetic remodeling, metabolic modulation, immune regulation, chemical small-molecules regulation, extracellular matrix remodeling, and cell-cell fusion, to achieve Müller glia reprogramming in mice.
Collapse
Affiliation(s)
- Zhiyuan Yin
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | | | | | | | | | | |
Collapse
|
4
|
Yang T, Ronning KE, Augustin S, Blond F, Nous C, Argyriou F, Touhami S, Delarasse C, Guillonneau X, Sennlaub F. Chronic intermittent hypoxia disrupts protective microgliosis in ischemic proliferative retinopathy. J Neuroinflammation 2025; 22:82. [PMID: 40087728 PMCID: PMC11909870 DOI: 10.1186/s12974-025-03392-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Accepted: 02/20/2025] [Indexed: 03/17/2025] Open
Abstract
Sleep apnea that leads to chronic intermittent hypoxia (CIH) is an independent risk factor for advanced, debilitating ischemic proliferative retinopathies, such as diabetic retinopathy (DR) and retinopathy of prematurity (ROP). The underlying mechanisms are unknown. Here we investigated the consequences of CIH on the ischemic retina of the oxygen-induced retinopathy model. We show that experimental CIH inhibited colony stimulating factor 1 (CSF1) expression, blunting the reactive microgliosis during the ischemic phase of OIR. CIH severely delayed beneficial revascularization of the ischemic retina and increased pathological neovascularization. CIH also induced photoreceptor segment thinning and accentuated OIR-induced inner and outer retinal functional deficits. Mechanistically we demonstrated that local CSF1R inhibition during ischemic retinopathy reduced the number of microglial cells, inhibited revascularization, and exacerbated pathological neovascularization, recapitulating the effects of CIH. Our findings provide a novel mechanism by which sleep apnea and CIH aggravate ischemic retinopathies, underscoring the importance of treating apnea in DR and ROP to help prevent sight threatening severe disease.
Collapse
Affiliation(s)
- Tianxiang Yang
- Sorbonne University, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, Paris, F-75012, France
- Aier Eye Institute, Changsha, Hunan Province, 410015, China
| | - Kaitryn E Ronning
- Sorbonne University, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, Paris, F-75012, France
| | - Sébastien Augustin
- Sorbonne University, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, Paris, F-75012, France
| | - Frédéric Blond
- Sorbonne University, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, Paris, F-75012, France
| | - Caroline Nous
- Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, INSERM-DHOS CIC 503, Paris, France
| | - Foteini Argyriou
- Sorbonne University, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, Paris, F-75012, France
| | - Sara Touhami
- Pitié Salpêtrière University Hospital, Sorbonne Université, 47-83 Boulevard de l'Hôpital, Paris, 75013, France
| | - Cécile Delarasse
- Sorbonne University, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, Paris, F-75012, France
| | - Xavier Guillonneau
- Sorbonne University, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, Paris, F-75012, France
| | - Florian Sennlaub
- Sorbonne University, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, Paris, F-75012, France.
- Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, INSERM-DHOS CIC 503, Paris, France.
| |
Collapse
|
5
|
Hu C, Cui T, Xu Z, Yang K, Wu Y, Cai W, Yu J, Qiu Y. Inhibiting HMGB1/AGER/NF-κB pathway prevents pro-inflammatory microglia polarization and protect photoreceptors in retinitis pigmentosa. Int Immunopharmacol 2025; 149:114192. [PMID: 39904032 DOI: 10.1016/j.intimp.2025.114192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 01/23/2025] [Accepted: 01/27/2025] [Indexed: 02/06/2025]
Abstract
PURPOSE Retinitis pigmentosa (RP) is an inherited retinal neurodegenerative disease which is a significant contributor to blindness. Microglia-mediated inflammation plays a crucial role in retinitis pigmentosa. However, the activation mechanisms of microglia and the role of polarized microglia in RP remain unclear. High-mobility group box 1 (HMGB1) is a key contributor to aseptic inflammation, and glycyrrhizin exerts anti-inflammatory effects by targeting HMGB1. This study aimed to investigate the role of HMGB1 and microglia in RP and explore the protective effects of glycyrrhizin on photoreceptors. METHODS Male C57BL/6 mice and age-matched rd1 mice were used for in vivo models, while zaprinast-treated 661w cells and HMGB1-treated BV-2 cells were used for in vitro models. In this study, the expression of HMGB1 was analyzed using QPCR and western blot (WB). Immunofluorescence staining and ELISA were performed to assess HMGB1 translocation and secretion. Glycyrrhizin was used to inhibit HMGB1, while FPS-ZM1 served as an inhibitor of the receptor for advanced glycation end products (AGER). Microglial polarization was evaluated by QPCR, and the HMGB1/ AGER/ NF-κB signaling pathway was analyzed through WB. Photoreceptor degeneration and visual function were assessed through H&E staining, electroretinography, and TUNEL staining. RESULTS We observed elevated levels of HMGB1 in the retina of rd1 mice and demonstrated in vitro that photoreceptors may serve as a significant source of HMGB1 in the retina. Additionally, HMGB1 was observed to cause microglial polarization via the HMGB1/AGER/ NF-κB pathway and the polarized microglia secrete inflammatory factors including TNF-α and IL-1β which accelerates the degeneration of photoreceptors. Glycyrrhizin reversed the degeneration of photoreceptors and loss of visual function in rd1 mice through the HMGB1/AGER/ NF-κB pathway. CONCLUSION Our findings showed that HMGB1 secreted by photoreceptors activated the microglia through the HMGB1/AGER/NF-κB pathway and the polarized microglia accelerates the degeneration of photoreceptors. Glycyrrhizin reversed the polarization caused by HMGB1 in vitro and delayed the progression of RP in vivo, presenting a potential novel approach for treating retinitis pigmentosa.
Collapse
Affiliation(s)
- Chengyu Hu
- Department of Ophthalmology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072 China
| | - Tao Cui
- Tianjin Medical Device Evaluation and Inspection Center, Tianjin, China
| | - Zihang Xu
- Department of Ophthalmology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072 China
| | - Kun Yang
- Tianjin Medical Device Evaluation and Inspection Center, Tianjin, China
| | - Yan Wu
- Department of Ophthalmology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072 China
| | - Wenting Cai
- Department of Ophthalmology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072 China
| | - Jing Yu
- Department of Ophthalmology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072 China; Department of Ophthalmology, The Third People's Hospital of Bengbu, Bengbu, China.
| | - Yaoyan Qiu
- Department of Ophthalmology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072 China.
| |
Collapse
|
6
|
You T, Yang Y, A L, Cheng X, Lin X, Liang Q, Ge L, Xie J, Chen S, Liu N, He J, Xu H, Ma X. IFNγ preconditioning improves neuroprotection of MSC-derived vesicles on injured retinal ganglion cells by suppressing microglia activation via miRNA-dependent ribosome activity. EXTRACELLULAR VESICLES AND CIRCULATING NUCLEIC ACIDS 2025; 6:87-111. [PMID: 40206798 PMCID: PMC11977360 DOI: 10.20517/evcna.2024.66] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 01/19/2025] [Accepted: 01/25/2025] [Indexed: 04/11/2025]
Abstract
Aim: Microglial activation plays a pivotal role in the pathogenesis of retinal ganglion cell (RGC) degeneration resulting from optic nerve crush (ONC). Small extracellular vesicles (sEVs) secreted by mesenchymal stem cells (MSCs) have the potential to prevent retinal degeneration by modulating microglial activation. In this study, we elucidated the specific effects of sEVs derived from IFN-γ-primed MSCs on the phenotypic transition of microglia and the associated pathways in ONC mice. Methods: The ONC mice model was established and administered intravitreal injection with the sEVs derived from native MSCs (native sEVs) and the sEVs derived from MSCs primed with IFN-γ (IFNγ-sEVs). Their respective effects on the survival of the retinal ganglion cells (RGCs) and the transition of microglia phenotypes were determined through visual function testing and immunohistochemical staining. Combined with mRNA seq and microRNA seq techniques, we elucidated the mechanism of modulation of microglia phenotypic transformation by sEVs derived from MSCs primed by IFNγ. Results: It demonstrated that IFNγ-sEVs exhibited superior protective effects against RGC loss and reduced inflammatory responses in the ONC retina compared to native sEVs. Both types of sEVs promoted microglia activation to disease-associated microglia (DAM) phenotype, while IFNγ-sEVs especially suppressed interferon-responsive microglia (IRM) activation during RGCs degeneration. Subsequent miRNA sequencing suggested that miR-423-5p, which exhibited the most significant differential expression between the two sEVs types and elevated expression in IFNγ-sEVs, inhibited the expression of IRM and ribosomal genes. Conclusion: These findings suggest that IFN-γ-preconditioned MSCs may enhance sEVs of neuroprotection on RGCs by suppressing IRM activation through the secretion of sEVs containing specific microRNAs in ONC mice.
Collapse
Affiliation(s)
- Tianjing You
- Department of Ophthalmology, The First Affiliated Hospital of Dalian Medical University, Dalian 116014, Liaoning, China
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Amy Medical University), Chongqing 400038, China
- Key Lab of Visual Damage and Regeneration and Restoration of Chongqing, Southwest Eye Hospital, Southwest Hospital, Chongqing 400038, China
- Authors contributed equally
| | - Yuanxing Yang
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Amy Medical University), Chongqing 400038, China
- Key Lab of Visual Damage and Regeneration and Restoration of Chongqing, Southwest Eye Hospital, Southwest Hospital, Chongqing 400038, China
- Authors contributed equally
| | - Luodan A
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Amy Medical University), Chongqing 400038, China
- Key Lab of Visual Damage and Regeneration and Restoration of Chongqing, Southwest Eye Hospital, Southwest Hospital, Chongqing 400038, China
- Authors contributed equally
| | - Xuan Cheng
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Amy Medical University), Chongqing 400038, China
- Key Lab of Visual Damage and Regeneration and Restoration of Chongqing, Southwest Eye Hospital, Southwest Hospital, Chongqing 400038, China
| | - Xi Lin
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Amy Medical University), Chongqing 400038, China
- Key Lab of Visual Damage and Regeneration and Restoration of Chongqing, Southwest Eye Hospital, Southwest Hospital, Chongqing 400038, China
| | - Qingle Liang
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Amy Medical University), Chongqing 400038, China
- Key Lab of Visual Damage and Regeneration and Restoration of Chongqing, Southwest Eye Hospital, Southwest Hospital, Chongqing 400038, China
| | - Lingling Ge
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Amy Medical University), Chongqing 400038, China
- Key Lab of Visual Damage and Regeneration and Restoration of Chongqing, Southwest Eye Hospital, Southwest Hospital, Chongqing 400038, China
| | - Jing Xie
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Amy Medical University), Chongqing 400038, China
- Key Lab of Visual Damage and Regeneration and Restoration of Chongqing, Southwest Eye Hospital, Southwest Hospital, Chongqing 400038, China
| | - Siyu Chen
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Amy Medical University), Chongqing 400038, China
- Key Lab of Visual Damage and Regeneration and Restoration of Chongqing, Southwest Eye Hospital, Southwest Hospital, Chongqing 400038, China
| | - Na Liu
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Amy Medical University), Chongqing 400038, China
- Key Lab of Visual Damage and Regeneration and Restoration of Chongqing, Southwest Eye Hospital, Southwest Hospital, Chongqing 400038, China
| | - Juncai He
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Amy Medical University), Chongqing 400038, China
- Key Lab of Visual Damage and Regeneration and Restoration of Chongqing, Southwest Eye Hospital, Southwest Hospital, Chongqing 400038, China
- Department of Ophthalmology, The 920 Hospital of PLA Joint Logistics Support Force, Kunming 650032, Yunnan, China
| | - Haiwei Xu
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Amy Medical University), Chongqing 400038, China
- Key Lab of Visual Damage and Regeneration and Restoration of Chongqing, Southwest Eye Hospital, Southwest Hospital, Chongqing 400038, China
| | - Xiang Ma
- Department of Ophthalmology, The First Affiliated Hospital of Dalian Medical University, Dalian 116014, Liaoning, China
| |
Collapse
|
7
|
Taylor O, Kelly L, El-Hodiri H, Fischer AJ. Sphingosine-1-phosphate signaling through Müller glia regulates neuroprotection and the accumulation of immune cells in the rodent retina. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.03.636254. [PMID: 39975061 PMCID: PMC11838470 DOI: 10.1101/2025.02.03.636254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
The purpose of this study was to investigate how Sphingosine-1-phosphate (S1P) signaling regulates glial phenotype, neuroprotection, and reprogramming of Müller glia (MG) into neurogenic MG-derived progenitor cells (MGPCs) in the adult mouse retina. We found that S1P-related genes were dynamically regulated following retinal damage. S1pr1 (encoding S1P receptor 1) and Sphk1 (encoding sphingosine kinase 1) are expressed at low levels by resting MG and are rapidly upregulated following acute damage. Overexpression of the neurogenic bHLH transcription factor Ascl1 in MG downregulates S1pr1, and inhibition of Sphk1 and S1pr1/3 enhances Ascl1-driven differentiation of bipolar-like cells and suppresses glial differentiation. Treatments that activate S1pr1 or increase retinal levels of S1P initiate pro-inflammatory NFκB-signaling in MG, whereas treatments that inhibit S1pr1 or decreased levels of S1P suppress NFκB-signaling in MG in damaged retinas. Conditional knock-out of NFκB-signaling in MG increases glial expression of S1pr1 but decreases levels of S1pr3 and Sphk1. Conditional knock-out (cKO) of S1pr1 in MG, but not Sphk1, enhances the accumulation of immune cells in acutely damaged retinas. cKO of S1pr1 is neuroprotective to ganglion cells, whereas cKO of Sphk1 is neuroprotective to amacrine cells in NMDA-damaged retinas. Consistent with these findings, pharmacological treatments that inhibit S1P receptors or inhibit Sphk1 had protective effects upon inner retinal neurons. We conclude that the S1P-signaling pathway is activated in MG after damage and this pathway acts secondarily to restrict the accumulation of immune cells, impairs neuron survival and suppresses the reprogramming of MG into neurogenic progenitors in the adult mouse retina.
Collapse
Affiliation(s)
- Olivia Taylor
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Neuroscience Graduate Program, The Ohio State University, Columbus, OH 43210, USA
| | - Lisa Kelly
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Heithem El-Hodiri
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Andy J. Fischer
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
8
|
Moekotte L, de Boer JH, Hiddingh S, de Ligt A, Nguyen XTA, Hoyng CB, Inglehearn CF, McKibbin M, Lamey TM, Thompson JA, Chen FK, McLaren TL, AlTalbishi A, Panneman DM, Boonen EGM, Banfi S, Bocquet B, Meunier I, De Baere E, Koenekoop R, Ołdak M, Rivolta C, Roberts L, Ramesar R, Strupaitė-Šileikienė R, Kohl S, Farrar GJ, van Vugt M, van Setten J, Roosing S, van den Born LI, Boon CJF, van Genderen MM, Kuiper JJW. Elevated Plasma Complement Factors in CRB1-Associated Inherited Retinal Dystrophies. Invest Ophthalmol Vis Sci 2025; 66:55. [PMID: 39982393 PMCID: PMC11855139 DOI: 10.1167/iovs.66.2.55] [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: 03/21/2024] [Accepted: 09/03/2024] [Indexed: 02/22/2025] Open
Abstract
Purpose To determine the profile of inflammation-related proteins and complement system factors in the plasma of CRB1-associated inherited retinal dystrophies (CRB1-IRDs). Methods We used the Olink Explore 384 Inflammation II panel for targeted proteomics in 30 cases and 29 controls (cohort I) to identify immune pathways involved in CRB1-IRDs. Genotyping was performed in cohort I and a second cohort of 123 patients from 14 countries and 1292 controls (cohort II). Results A significant shift in complement cascade factors was observed in plasma proteomes of CRB1-IRD patients (enrichment for complement cascade, Padj = 3.03 × 10-15). We detected higher plasma levels of complement factor I and complement factor H [CFH] (q = 0.008 and q = 0.046, respectively, adjusted for age and sex), inhibitors of complement component 3 (C3), which correlated significantly (Pearson's coefficient >0.6) with elevated levels of C3 (q = 0.064). The CRB1 missense variants frequently found in patients showed a strong linkage disequilibrium with the common CFH variant rs7535263 (D' = 0.97 for p.(Cys948Tyr); D' = 1.0 for p.(Arg764Cys)), known to be linked with altered plasma CFH-related protein levels. Correction for the CFH genotype revealed significantly elevated plasma levels of CFH-related 2 (CFHR2) in CRB1-IRD patients (q = 0.041). Conclusions CRB1-IRDs are characterized by changes in plasma levels of complement factors and proteins of the innate immune system, and linkage between CRB1 and CFH genes implicates functional variants of the CFH-CFHR locus with specific pathogenic variants of CRB1.
Collapse
Affiliation(s)
- Lude Moekotte
- Department of Ophthalmology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Joke H. de Boer
- Department of Ophthalmology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Sanne Hiddingh
- Department of Ophthalmology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Aafke de Ligt
- Department of Ophthalmology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Xuan-Thanh-An Nguyen
- Department of Ophthalmology, Leiden University Medical Center, Leiden, the Netherlands
| | - Carel B. Hoyng
- Department of Ophthalmology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Chris F. Inglehearn
- Division of Molecular Medicine, Leeds Institute of Medical Research, University of Leeds, Leeds, United Kingdom
| | - Martin McKibbin
- Division of Molecular Medicine, Leeds Institute of Medical Research, University of Leeds, Leeds, United Kingdom
- Department of Ophthalmology, St. James's University Hospital, Leeds, United Kingdom
| | - Tina M. Lamey
- Centre for Ophthalmology and Visual Science, The University of Western Australia, Perth, Western Australia, Australia
| | - Jennifer A. Thompson
- Australian Inherited Retinal Disease Registry & DNA Bank, Department of Medical Technology and Physics, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
| | - Fred K. Chen
- Centre for Ophthalmology and Visual Science, The University of Western Australia, Perth, Western Australia, Australia
- Australian Inherited Retinal Disease Registry & DNA Bank, Department of Medical Technology and Physics, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
| | - Terri L. McLaren
- Centre for Ophthalmology and Visual Science, The University of Western Australia, Perth, Western Australia, Australia
- Australian Inherited Retinal Disease Registry & DNA Bank, Department of Medical Technology and Physics, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
| | - Alaa AlTalbishi
- St. John of Jerusalem Eye Hospital Group, East Jerusalem, Palestine
| | - Daan M. Panneman
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Erica G. M. Boonen
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Sandro Banfi
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Béatrice Bocquet
- Institute for Neurosciences of Montpellier (INM), University of Montpellier, INSERM, Montpellier, France
- National Reference Center for Inherited Sensory Diseases, University of Montpellier, CHU, Montpellier, France
| | - Isabelle Meunier
- Institute for Neurosciences of Montpellier (INM), University of Montpellier, INSERM, Montpellier, France
- National Reference Center for Inherited Sensory Diseases, University of Montpellier, CHU, Montpellier, France
| | - Elfride De Baere
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
| | - Robert Koenekoop
- McGill University Health Center (MUHC) Research Institute, Montreal, QC, Canada
- Departments of Paediatric Surgery, Human Genetics, and Adult Ophthalmology, McGill University Health Center, Montreal, QC, Canada
| | - Monika Ołdak
- Department of Histology and Embryology, Medical University of Warsaw, Warsaw, Poland
| | - Carlo Rivolta
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
- Department of Ophthalmology, University of Basel, Basel, Switzerland
- Department of Genetics and Genome Biology, University of Leicester, Leicester, United Kingdom
| | - Lisa Roberts
- UCT/MRC Precision and Genomic Medicine Research Unit, Division of Human Genetics, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Raj Ramesar
- UCT/MRC Precision and Genomic Medicine Research Unit, Division of Human Genetics, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Rasa Strupaitė-Šileikienė
- Center of Eye Diseases, Clinic of Ear, Nose, Throat, and Eye Diseases, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Susanne Kohl
- Institute for Ophthalmic Research, Centre for Ophthalmology, University of Tübingen, Tübingen, Germany
| | - G. Jane Farrar
- The School of Genetics & Microbiology, The University of Dublin Trinity College, Dublin, Ireland
| | - Marion van Vugt
- Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Jessica van Setten
- Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Susanne Roosing
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
| | | | - Camiel J. F. Boon
- Department of Ophthalmology, Leiden University Medical Center, Leiden, the Netherlands
- Department of Ophthalmology, Amsterdam UMC, Amsterdam, the Netherlands
| | - Maria M. van Genderen
- Department of Ophthalmology, University Medical Center Utrecht, Utrecht, the Netherlands
- Bartiméus, Diagnostic Centre for complex visual disorders, Zeist, the Netherlands
| | - Jonas J. W. Kuiper
- Department of Ophthalmology, University Medical Center Utrecht, Utrecht, the Netherlands
| |
Collapse
|
9
|
Leach LL, Gonzalez RG, Jayawardena SU, Gross JM. Interleukin-34 and debris clearance by mononuclear phagocytes drive retinal pigment epithelium regeneration in zebrafish. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.10.632236. [PMID: 39868193 PMCID: PMC11761032 DOI: 10.1101/2025.01.10.632236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
The retinal pigment epithelium (RPE) surrounds the posterior eye and maintains the health and function of the photoreceptors. Consequently, RPE dysfunction or damage has a devastating impact on vision. Due to complex etiologies, there are currently no cures for patients with RPE degenerative diseases, which remain some of the most prevalent causes of vision loss worldwide. Further, owing to a limited capacity for mammalian tissue repair, we know little about how the RPE regenerates. Here, we utilize zebrafish as a model to uncover novel mechanisms driving intrinsic RPE regeneration. We show that interleukin-34 signaling from damaged RPE is required for precisely timed recruitment of mononuclear phagocytes (MNPs) to the injury site. Additionally, we find that cellular debris clearance by MNPs is indispensable for regeneration, as microglia-deficient zebrafish fail to regenerate RPE and photoreceptor tissues. Together, our results establish specific pro-regenerative functions of MNPs after RPE damage.
Collapse
Affiliation(s)
- Lyndsay L. Leach
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, United States of America
| | - Rebecca G. Gonzalez
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, United States of America
| | - Sayuri U. Jayawardena
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, United States of America
| | - Jeffrey M. Gross
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, United States of America
| |
Collapse
|
10
|
Li J, Li H, Wei C, Chen C, Zheng Z. Astragalus polysaccharide attenuates retinal ischemia reperfusion-induced microglial activation through sortilin-related vacuolar protein sorting 10 domain containing receptor 2/laminin subunit alpha 1 upregulation. Cytojournal 2025; 22:2. [PMID: 39958884 PMCID: PMC11829307 DOI: 10.25259/cytojournal_131_2024] [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: 07/16/2024] [Accepted: 11/29/2024] [Indexed: 02/18/2025] Open
Abstract
Objective Microglial activation is a hallmark of pathogenic retinal conditions such as retinal ischemia-reperfusion (RIR). While sortilin-related vacuolar protein sorting 10 domain containing receptor 2 (Sorcs2) and laminin subunit alpha 1 (Lama1) have been implicated in neuroinflammatory processes, their roles in regulating microglial activation in RIR are not reported. The current work studied the potential of Sorcs2 and Lama1 as negative regulators of microglial activation in RIR and assessed the therapeutic potential of Astragalus polysaccharide (AP). Material and Methods Transcriptome profiling was conducted in retinal specimens of RIR group 72 h after RIR induction. Oxygen-glucose deprivation/reperfusion (OGD/R) in rat microglial cells was employed as the cellular induction model of RIR. The functional role of Sorcs2 and Lama1 in dictating microglial activation was investigated in vitro and in vivo using lentivirus-based gene expression. Further, the potential effect of AP on RIR-mediated microglial activation was investigated. Results Sorcs2 and Lama1 were identified as two downregulated genes in retinal samples following RIR. OGD/R induction triggered pro-inflammatory microglial activation and induced the downregulation of Sorcs2 and Lama1. Sorcs2 or Lama1 overexpression hindered OGD/R-induced microglial activation in vitro and attenuated inflammatory expansion of microglia cells in RIR-induced rat retinal samples. AP treatment was able to neutralize the oxidative stress, promote the expression of Sorcs2 and Lama1, and suppress microglial activation. Conclusion Our findings pinpoint Sorcs2 and Lama1 as negative regulators of microglial activation in RIR. AP could be employed as an antioxidant to attenuate microglial activation and ameliorate the inflammatory damages in RIR.
Collapse
Affiliation(s)
- Juanjuan Li
- Department of Ophthalmologic, The Affiliated Hospital of Yunnan University (Second People’s Hospital of Yunnan Province, Yunnan Eye Hospital), Kunming, Yunnan, China
| | - Hua Li
- Department of Ophthalmologic, The Affiliated Hospital of Yunnan University (Second People’s Hospital of Yunnan Province, Yunnan Eye Hospital), Kunming, Yunnan, China
| | - Chunling Wei
- Department of Ophthalmologic, The Affiliated Hospital of Yunnan University (Second People’s Hospital of Yunnan Province, Yunnan Eye Hospital), Kunming, Yunnan, China
| | - Chen Chen
- Department of Ophthalmologic, The Affiliated Hospital of Yunnan University (Second People’s Hospital of Yunnan Province, Yunnan Eye Hospital), Kunming, Yunnan, China
| | - Zhikun Zheng
- Department of Ophthalmologic, The Affiliated Hospital of Yunnan University (Second People’s Hospital of Yunnan Province, Yunnan Eye Hospital), Kunming, Yunnan, China
| |
Collapse
|
11
|
Taylor OB, El‐Hodiri HM, Palazzo I, Todd L, Fischer AJ. Regulating the formation of Müller glia-derived progenitor cells in the retina. Glia 2025; 73:4-24. [PMID: 39448874 PMCID: PMC11660542 DOI: 10.1002/glia.24635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/18/2024] [Accepted: 09/28/2024] [Indexed: 10/26/2024]
Abstract
We summarize recent findings in different animal models regarding the different cell-signaling pathways and gene networks that influence the reprogramming of Müller glia into proliferating, neurogenic progenitor cells in the retina. Not surprisingly, most of the cell-signaling pathways that guide the proliferation and differentiation of embryonic retinal progenitors also influence the ability of Müller glia to become proliferating Müller glia-derived progenitor cells (MGPCs). Further, the neuronal differentiation of MGPC progeny is potently inhibited by networks of neurogenesis-suppressing genes in chick and mouse models but occurs freely in zebrafish. There are important differences between the model systems, particularly pro-inflammatory signals that are active in mature Müller glia in damaged rodent and chick retinas, but less so in fish retinas. These pro-inflammatory signals are required to initiate the process of reprogramming, but if sustained suppress the potential of Müller glia to become neurogenic MGPCs. Further, there are important differences in how activated Müller glia up- or downregulate pro-glial transcription factors in the different model systems. We review recent findings regarding regulatory cell signaling and gene networks that influence the activation of Müller glia and the transition of these glia into proliferating progenitor cells with neurogenic potential in fish, chick, and mouse model systems.
Collapse
Affiliation(s)
- Olivia B. Taylor
- Department of NeuroscienceCollege of Medicine, The Ohio State UniversityColumbusOhioUSA
- Neuroscience Graduate ProgramThe Ohio State UniversityColumbusOhioUSA
| | - Heithem M. El‐Hodiri
- Department of NeuroscienceCollege of Medicine, The Ohio State UniversityColumbusOhioUSA
| | - Isabella Palazzo
- The Solomon H. Snyder Department of NeuroscienceJohns Hopkins University School of MedicineBaltimoreMassachusettsUSA
| | - Levi Todd
- Department of Ophthalmology and Visual SciencesSUNY Upstate Medical UniversitySyracuseNew YorkUSA
| | - Andy J. Fischer
- Department of NeuroscienceCollege of Medicine, The Ohio State UniversityColumbusOhioUSA
| |
Collapse
|
12
|
Ronning KE, Burns ME, Sennlaub F. Monocytes in Retinal Degeneration: Little Cells with a Big Impact. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1468:133-137. [PMID: 39930185 DOI: 10.1007/978-3-031-76550-6_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
Inflammation has been strongly implicated in retinal degenerative disorders, including inherited retinal degenerations (IRDs) and age-related macular degeneration (AMD). Microglia are the only immune cells in the retina during normal function, but during damage and disease, monocytes are able to invade the retina. Despite similarities to microglia, monocyte-derived cells (MdCs) may play a distinct and often pathogenic role in disease. Recent technological advances are rapidly improving our ability to investigate monocytic cells, yet many questions remain. Still, it is clear monocytes play an important role during retinal degenerative disorders and they are an exciting target for the development of therapeutic interventions.
Collapse
Affiliation(s)
- Kaitryn E Ronning
- Sorbonne Université, CNRS, Inserm, Institut de la Vision, Paris, France.
| | - Marie E Burns
- Center for Neuroscience, University of California Davis, Davis, CA, USA
- Department of Cell Biology and Human Anatomy, University of California Davis, Davis, CA, USA
- Department of Ophthalmology & Vision Science, University of California Davis, Davis, CA, USA
| | - Florian Sennlaub
- Sorbonne Université, CNRS, Inserm, Institut de la Vision, Paris, France
| |
Collapse
|
13
|
Hata M, Hata M, Dejda A, Pilon F, Diaz-Marin R, Fournier F, Joyal JS, Cagnone G, Ochi Y, Crespo-Garcia S, Wilson AM, Sapieha P. Corticosteroids reduce pathological angiogenesis yet compromise reparative vascular remodeling in a model of retinopathy. Proc Natl Acad Sci U S A 2024; 121:e2411640121. [PMID: 39693344 DOI: 10.1073/pnas.2411640121] [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: 06/11/2024] [Accepted: 10/11/2024] [Indexed: 12/20/2024] Open
Abstract
Tissue inflammation is often broadly associated with cellular damage, yet sterile inflammation also plays critical roles in beneficial tissue remodeling. In the central nervous system, this is observed through a predominantly innate immune response in retinal vascular diseases such as age-related macular degeneration, diabetic retinopathy, and retinopathy of prematurity. Here, we set out to elucidate the dynamics of the immune response during progression and regression of pathological neovascularization in retinopathy. In a mouse model of oxygen-induced retinopathy, we report that dexamethasone, a broad-spectrum corticosteroid, suppresses initial formation of pathological preretinal neovascularization in early stages of disease, yet blunts reparative inflammation by impairing distinct myeloid cell populations, and hence reduces beneficial vascular remodeling in later stages of disease. Using genetic depletion of distinct components of the innate immune response, we demonstrate that CX3C chemokine receptor 1-expressing microglia contribute to angiogenesis. Conversely, myeloid cells expressing lysozyme 2 are recruited to sites of damaged blood vessels and pathological neovascularization where they partake in a reparative process that ultimately restores circulatory homeostasis to the retina. Hence, the Janus-faced properties of anti-inflammatory drugs should be considered, particularly in stages associated with persistent neovascularization.
Collapse
Affiliation(s)
- Masayuki Hata
- Departments of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, QC H1T 2M4, Canada
- Department of Biochemistry and Molecular Medicine, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, QC H1T 2M4, Canada
| | - Maki Hata
- Departments of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, QC H1T 2M4, Canada
| | - Agnieszka Dejda
- Departments of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, QC H1T 2M4, Canada
| | - Frédérique Pilon
- Departments of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, QC H1T 2M4, Canada
| | - Roberto Diaz-Marin
- Department of Biochemistry and Molecular Medicine, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, QC H1T 2M4, Canada
| | - Frédérik Fournier
- Departments of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, QC H1T 2M4, Canada
| | - Jean-Sebastien Joyal
- Departments of Pediatrics, Ophthalmology, and Pharmacology, Centre Hospitalier, Universitaire Ste-Justine Research Center, Montréal, QC H3T 1C5, Canada
| | - Gael Cagnone
- Departments of Pediatrics, Ophthalmology, and Pharmacology, Centre Hospitalier, Universitaire Ste-Justine Research Center, Montréal, QC H3T 1C5, Canada
| | - Yotaro Ochi
- Department of Pathology and Tumour Biology, Graduate School of Medicine, Kyoto University, Kyoto 606-8315, Japan
| | - Sergio Crespo-Garcia
- Departments of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, QC H1T 2M4, Canada
- Department of Biochemistry and Molecular Medicine, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, QC H1T 2M4, Canada
| | - Ariel M Wilson
- Departments of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, QC H1T 2M4, Canada
| | - Przemyslaw Sapieha
- Departments of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, QC H1T 2M4, Canada
- Department of Biochemistry and Molecular Medicine, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, QC H1T 2M4, Canada
| |
Collapse
|
14
|
Chen J, Lu T, Chen C, Zheng W, Lu L, Li N. Elevation of ANXA1 associated with potential protective mechanism against ferroptosis and immune cell infiltration in age-related macular degeneration. Eur J Med Res 2024; 29:615. [PMID: 39710756 DOI: 10.1186/s40001-024-02163-1] [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: 09/10/2024] [Accepted: 11/19/2024] [Indexed: 12/24/2024] Open
Abstract
BACKGROUND Age-related macular degeneration (AMD), is a neurodegenerative ocular disease. This study investigated the role of ferroptosis-related genes and their interaction with immune cell infiltration in AMD. METHODS We screened differential expression genes (DEGs) of AMD from data sets in Gene Expression Omnibus. We identified ferroptosis-related differentially expressed genes (ferroDEGs) by intersecting DEGs with ferroptosis-related genes. Protein-protein interactions network and Cytoscape were used for screening hub genes. Next, we analyzed immune cell infiltration using CIBERSORT and examined the crosstalk between hub ferroDEGs and immune cell infiltration. Hub genes expression in each cell cluster and the proportions of different cell clusters between AMD and normal samples were examined using single-cell data. The hub ferroDEG expressions were verified in cell and mouse models using RT-qPCR, western blot, and immunofluorescence assay. The roles of ANXA1 in ferroptosis and its crosstalk with microglia were investigated. RESULTS We identified hub ferroDEGs that include six genes (ANXA1, DKK1, CD44, VIM, TGFB2, DUSP1). Functional analysis of those hub ferroDEGs was found to be correlated with leukocyte migration and chemotaxis, macrophage migration, and gliogenesis. The high-risk ferroptosis group exhibited elevated levels of CD8+ T cells, activated NK cells, and M2 macrophages. Single-cell sequencing data revealed a high degree of cell heterogeneity in macular degeneration and the monocytes proportion in the macular area was higher in AMD samples. Moreover, we observed elevated mRNA and protein levels of CD44, ANXA1 (P < 0.01), while ANXA1 knockdown reduced GPX4 expression in the cell model. Finally, we validated increased ANXA1 expression and observed its colocalization with microglia in mouse models using immunofluorescence assays. CONCLUSIONS This study offers insights into the AMD pathogenesis and identifies ANXA1 as a potential target related to protecting from ferroptosis and immune response for future research.
Collapse
Affiliation(s)
- Jing Chen
- Department of Ophthalmology, Zhongshan City People's Hospital, Zhongshan, Guangdong, China
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Tu Lu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Sun Yat-Sen University, Guangzhou, 510060, China
- Department of Ophthalmology, First Hospital of China Medical University, Shenyang, China
| | - Chen Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Wenbin Zheng
- Department of Ophthalmology, Zhongshan City People's Hospital, Zhongshan, Guangdong, China
| | - Lin Lu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Sun Yat-Sen University, Guangzhou, 510060, China.
| | - Naiyang Li
- Department of Ophthalmology, Zhongshan City People's Hospital, Zhongshan, Guangdong, China.
| |
Collapse
|
15
|
Méjécase C, Nair N, Sarkar H, Soro-Barrio P, Toms M, Halliday S, Linkens K, Jaroszynska N, Maurer C, Owen N, Moosajee M. Oxidative Stress, Inflammation and Altered Glucose Metabolism Contribute to the Retinal Phenotype in the Choroideremia Zebrafish. Antioxidants (Basel) 2024; 13:1587. [PMID: 39765914 PMCID: PMC11673030 DOI: 10.3390/antiox13121587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 12/16/2024] [Accepted: 12/19/2024] [Indexed: 01/11/2025] Open
Abstract
Reactive oxygen species (ROS) within the retina play a key role in maintaining function and cell survival. However, excessive ROS can lead to oxidative stress, inducing dysregulation of metabolic and inflammatory pathways. The chmru848 zebrafish models choroideremia (CHM), an X-linked chorioretinal dystrophy, which predominantly affects the photoreceptors, retinal pigment epithelium (RPE), and choroid. In this study, we examined the transcriptomic signature of the chmru848 zebrafish retina to reveal the upregulation of cytokine pathways and glia migration, upregulation of oxidative, ER stress and apoptosis markers, and the dysregulation of glucose metabolism with the downregulation of glycolysis and the upregulation of the oxidative phase of the pentose phosphate pathway. Glucose uptake was impaired in the chmru848 retina using the 2-NBDG glucose uptake assay. Following the overexpression of human PFKM, partial rescue was seen with the preservation of photoreceptors and RPE and increased glucose uptake, but without modifying glycolysis and oxidative stress markers. Therapies targeting glucose metabolism in CHM may represent a potential remedial approach.
Collapse
Affiliation(s)
- Cécile Méjécase
- Development, Ageing and Disease, UCL Institute of Ophthalmology, London EC1V 9EL, UK; (C.M.); (N.N.); (H.S.); (M.T.); (K.L.); (N.J.); (C.M.); (N.O.)
- Ocular Genomics and Therapeutics, The Francis Crick Institute, London NW1 1AT, UK
| | - Neelima Nair
- Development, Ageing and Disease, UCL Institute of Ophthalmology, London EC1V 9EL, UK; (C.M.); (N.N.); (H.S.); (M.T.); (K.L.); (N.J.); (C.M.); (N.O.)
- Ocular Genomics and Therapeutics, The Francis Crick Institute, London NW1 1AT, UK
| | - Hajrah Sarkar
- Development, Ageing and Disease, UCL Institute of Ophthalmology, London EC1V 9EL, UK; (C.M.); (N.N.); (H.S.); (M.T.); (K.L.); (N.J.); (C.M.); (N.O.)
- Ocular Genomics and Therapeutics, The Francis Crick Institute, London NW1 1AT, UK
| | - Pablo Soro-Barrio
- Bioinformatics and Biostatistics Science Technology Platform, The Francis Crick Institute, London NW1 1AT, UK
| | - Maria Toms
- Development, Ageing and Disease, UCL Institute of Ophthalmology, London EC1V 9EL, UK; (C.M.); (N.N.); (H.S.); (M.T.); (K.L.); (N.J.); (C.M.); (N.O.)
- Ocular Genomics and Therapeutics, The Francis Crick Institute, London NW1 1AT, UK
| | - Sophia Halliday
- Development, Ageing and Disease, UCL Institute of Ophthalmology, London EC1V 9EL, UK; (C.M.); (N.N.); (H.S.); (M.T.); (K.L.); (N.J.); (C.M.); (N.O.)
| | - Katy Linkens
- Development, Ageing and Disease, UCL Institute of Ophthalmology, London EC1V 9EL, UK; (C.M.); (N.N.); (H.S.); (M.T.); (K.L.); (N.J.); (C.M.); (N.O.)
- Ocular Genomics and Therapeutics, The Francis Crick Institute, London NW1 1AT, UK
| | - Natalia Jaroszynska
- Development, Ageing and Disease, UCL Institute of Ophthalmology, London EC1V 9EL, UK; (C.M.); (N.N.); (H.S.); (M.T.); (K.L.); (N.J.); (C.M.); (N.O.)
| | - Constance Maurer
- Development, Ageing and Disease, UCL Institute of Ophthalmology, London EC1V 9EL, UK; (C.M.); (N.N.); (H.S.); (M.T.); (K.L.); (N.J.); (C.M.); (N.O.)
| | - Nicholas Owen
- Development, Ageing and Disease, UCL Institute of Ophthalmology, London EC1V 9EL, UK; (C.M.); (N.N.); (H.S.); (M.T.); (K.L.); (N.J.); (C.M.); (N.O.)
| | - Mariya Moosajee
- Development, Ageing and Disease, UCL Institute of Ophthalmology, London EC1V 9EL, UK; (C.M.); (N.N.); (H.S.); (M.T.); (K.L.); (N.J.); (C.M.); (N.O.)
- Ocular Genomics and Therapeutics, The Francis Crick Institute, London NW1 1AT, UK
- Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 9JH, UK
- Department of Genetics, Moorfields Eye Hospital NHS Foundation Trust, London EC1V 2PD, UK
| |
Collapse
|
16
|
Zhao B, Zhao Y, Sun X. Mechanism and therapeutic targets of circulating immune cells in diabetic retinopathy. Pharmacol Res 2024; 210:107505. [PMID: 39547465 DOI: 10.1016/j.phrs.2024.107505] [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: 08/08/2024] [Revised: 11/06/2024] [Accepted: 11/11/2024] [Indexed: 11/17/2024]
Abstract
Diabetic retinopathy (DR) continues to be the leading cause of preventable vision loss among working-aged adults, marked by immune dysregulation within the retinal microenvironment. Typically, the retina is considered as an immune-privileged organ, where circulating immune cells are restricted from entry under normal conditions. However, during the progression of DR, this immune privilege is compromised as circulating immune cells breach the barrier and infiltrate the retina. Increasing evidence suggests that vascular and neuronal degeneration in DR is largely driven by the infiltration of immune cells, particularly neutrophils, monocyte-derived macrophages, and lymphocytes. This review delves into the mechanisms and therapeutic targets associated with these immune cell populations in DR, offering a promising and innovative approach to managing the disease.
Collapse
Affiliation(s)
- Bowen Zhao
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yin Zhao
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Xufang Sun
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
17
|
Ren Q, Lu F, Hao R, Chen Y, Liang C. Subretinal microglia support donor photoreceptor survival in rd1 mice. Stem Cell Res Ther 2024; 15:436. [PMID: 39563450 PMCID: PMC11575076 DOI: 10.1186/s13287-024-04052-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 11/06/2024] [Indexed: 11/21/2024] Open
Abstract
PURPOSE To investigate the potential relationship between subretinal microglia and transplanted donor photoreceptors. METHODS Photoreceptor precursors were transplanted into wild-type mice and rd1 mice by trans-scleral injection. Immunohistochemistry was employed to detect microglia and macrophages. PlX5622 feed was used to achieve microglia depletion and microglia repopulation. RNA-seq and qPCR were utilized to evaluate gene expression. Confocal microscopy was used to observe the interaction between microglia and donor photoreceptors. RESULTS Donor photoreceptors survived in rd1 mice but not in wild-type mice after trans-scleral injection. The microglial cells closely interacted with donor cells. While donor cells failed to survive in rd1 mice after microglia depletion, they could survive following microglia repopulation. The RNA-seq analysis showed a pro-neurodevelopmental effect of sub-retinal microglia/RPE tissue in rd1 mice. CONCLUSIONS Subretinal microglia supported donor photoreceptor survival in rd1 mice.
Collapse
Affiliation(s)
- Qinjia Ren
- Department of Ophthalmology, West China Hospital, Sichuan University, Cheng Du, Sichuan, China
| | - Fang Lu
- Department of Ophthalmology, West China Hospital, Sichuan University, Cheng Du, Sichuan, China
| | - Ruwa Hao
- Department of Ophthalmology, West China Hospital, Sichuan University, Cheng Du, Sichuan, China
| | - Yingying Chen
- Department of Ophthalmology, West China Hospital, Sichuan University, Cheng Du, Sichuan, China
| | - Chen Liang
- Department of Ophthalmology, West China Hospital, Sichuan University, Cheng Du, Sichuan, China.
| |
Collapse
|
18
|
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
|
19
|
Lu C, Mao X, Yuan S. Decoding physiological and pathological roles of innate immune cells in eye diseases: the perspectives from single-cell RNA sequencing. Front Immunol 2024; 15:1490719. [PMID: 39544948 PMCID: PMC11560449 DOI: 10.3389/fimmu.2024.1490719] [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: 09/03/2024] [Accepted: 10/09/2024] [Indexed: 11/17/2024] Open
Abstract
Single-cell RNA sequencing (scRNA-seq) has facilitated a deeper comprehension of the molecular mechanisms behind eye diseases and has prompted the selection of precise therapeutic targets by examining the cellular and molecular intricacies at the single-cell level. This review delineates the pivotal role of scRNA-seq in elucidating the functions of innate immune cells within the context of ocular pathologies. Recent advancements in scRNA-seq have revealed that innate immune cells, both from the periphery and resident in the retina, are actively engaged in various stages of multiple eye diseases. Notably, resident microglia and infiltrating neutrophils exhibit swift responses during the initial phase of injury, while peripheral monocyte-derived macrophages exhibit transcriptomic profiles akin to those of activated microglia, suggesting their potential for long-term residence within the retina. The scRNA-seq analyses have underscored the cellular heterogeneity and gene expression alterations within innate immune cells, which, while sharing commonalities, exhibit disease-specific variations. These insights have not only broadened our understanding of the cellular and molecular mechanisms in eye diseases but also paved the way for the identification of candidate targets for targeted therapeutic interventions. The application of scRNA-seq technology has heralded a new era in the study of ocular pathologies, enabling a more detailed appreciation of the roles that innate immune cells play across a spectrum of eye diseases.
Collapse
Affiliation(s)
- Chen Lu
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiying Mao
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Songtao Yuan
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
20
|
Ortega AJ, Daniel S, Renwick M, Kambhampati P, Thompson KN, Collier GE, Baker EL, Zaki H, Hulleman JD. Genetic removal of Nlrp3 protects against sporadic and R345W Efemp1-induced basal laminar deposit formation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.14.618289. [PMID: 39464028 PMCID: PMC11507711 DOI: 10.1101/2024.10.14.618289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Chronic, unresolved inflammation has long been speculated to serve as an initiating and propagating factor in numerous neurodegenerative diseases, including a leading cause of irreversible blindness in the elderly, age-related macular degeneration (AMD). Intracellular multiprotein complexes called inflammasomes in combination with activated caspases facilitate production of pro-inflammatory cytokines such as interleukin 1 beta. Specifically, the nucleotide-binding oligomerization (NOD)-like receptor protein 3 (NLRP3) has received heightened attention due to the wide range of stimuli to which it can respond and its potential involvement in AMD. In this study, we directly tested the role of Nlrp3 and its downstream effector, caspase 1 (Casp1) in mediating early AMD-like pathology (i.e., basal laminar deposits [BLamDs]) in wild-type (WT) mice and the Malattia Leventinese/Doyne honeycomb retinal dystrophy (ML/DHRD) mouse model (p.R345W mutation in Efemp1). Compared to aged-matched controls, R345W+/+ knockin mice demonstrated increased Muller cell gliosis, subretinal Iba-1+ microglial cells, higher Nlrp3 immunoreactivity in the retina, as well as significant transcriptional upregulation of complement component 3, Nlrp3, pro-Il1b, pro-caspase-1, and tissue inhibitor of matrix metalloproteinase 3 in the retinal pigmented epithelium (RPE)/choroid. These findings were accompanied by an age-related increase in BLamD formation in the R345W+/+ mice. Genetic elimination of either Nlrp3 or Casp1 significantly reduced both the size and coverage of BLamDs in the R345W+/+ background, highlighting an important and underappreciated pathway that could affect ML/DHRD onset and progression. Moreover, Nlrp3 knockout reduced spontaneous, idiopathic BLamDs in WT mice, suggesting translatability of our findings not only to rare inherited retinal dystrophies, but also potentially to AMD itself.
Collapse
Affiliation(s)
- Antonio J. Ortega
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, 2001 6 St. SE, Minneapolis, Minnesota, 55455, United States
| | - Steffi Daniel
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, 2001 6 St. SE, Minneapolis, Minnesota, 55455, United States
| | - Marian Renwick
- Department of Ophthalmology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas, 75390, United States
| | - Pravallika Kambhampati
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, 2001 6 St. SE, Minneapolis, Minnesota, 55455, United States
| | - Krista N. Thompson
- Department of Ophthalmology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas, 75390, United States
| | - Gracen E. Collier
- Department of Ophthalmology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas, 75390, United States
| | - Emily L. Baker
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, 2001 6 St. SE, Minneapolis, Minnesota, 55455, United States
| | - Hasan Zaki
- Department of Pathology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas, 75390, United States
| | - John D. Hulleman
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, 2001 6 St. SE, Minneapolis, Minnesota, 55455, United States
| |
Collapse
|
21
|
Karmoker JR, Bounds SE, Cai J. Aryl hydrocarbon receptor (AhR)-mediated immune responses to degeneration of the retinal pigment epithelium. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167351. [PMID: 39004382 PMCID: PMC11330344 DOI: 10.1016/j.bbadis.2024.167351] [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: 03/07/2024] [Revised: 07/05/2024] [Accepted: 07/08/2024] [Indexed: 07/16/2024]
Abstract
Injuries to the retinal pigment epithelium (RPE) trigger immune responses, orchestrating interactions within the innate and adaptive immune systems in the outer retina and choroid. We previously reported that interleukin 17 (IL-17) is a pivotal signaling molecule originating from choroidal γδ T cells, exerting protective effects by mediating functional connections between the RPE and subretinal microglia. In this current study, we generated mice with aryl hydrocarbon receptor (AhR) knockout specifically in IL-17-producing cells. These animals had deficiency in IL-17 production from γδ T cells, and exhibited increased sensitivity to both acute and chronic insults targeting the RPE. These findings imply that IL-17 plays a crucial role as a signaling cytokine in preserving the homeostasis of the outer retina and choroid.
Collapse
Affiliation(s)
- James Regun Karmoker
- Department of Biochemistry & Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| | - Sarah E Bounds
- Department of Biochemistry & Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| | - Jiyang Cai
- Department of Biochemistry & Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
22
|
Liu J, Lei F, Yan B, Cui N, Sharma J, Correa V, Roach L, Nicolaou S, Pitts K, Chodosh J, Maidana DE, Vavvas D, Margeta MA, Zhang H, Weitz D, Mostoslavsky R, Paschalis EI. Epigenetic adaptation drives monocyte differentiation into microglia-like cells upon engraftment into the retina. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.09.612126. [PMID: 39314467 PMCID: PMC11419019 DOI: 10.1101/2024.09.09.612126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
The identification of specific markers for microglia has been a long-standing challenge. Recently, markers such as P2ry12, TMEM119, and Fcrls have been proposed as microglia-specific and widely used to explore microglial functions within various central nervous system (CNS) contexts. The specificity of these markers was based on the assumption that circulating monocytes retain their distinct signatures even after infiltrating the CNS. However, recent findings reveal that infiltrating monocytes can adopt microglia-like characteristics while maintaining a pro-inflammatory profile upon permanent engraftment in the CNS.In this study, we utilize bone marrow chimeras, single-cell RNA sequencing, ATAC-seq, flow cytometry, and immunohistochemistry to demonstrate that engrafted monocytes acquire expression of established microglia markers-P2ry12, TMEM119, Fcrls-and the pan-myeloid marker Iba1, which has been commonly mischaracterized as microglia-specific. These changes are accompanied by alterations in chromatin accessibility and shifts in chromatin binding motifs that are indicative of microglial identity. Moreover, we show that engrafted monocytes dynamically regulate the expression of CX3CR1, CCR2, Ly6C, and transcription factors PU.1, CTCF, RUNX, AP-1, CEBP, and IRF2, all of which are crucial for shaping microglial identity. This study is the first to illustrate that engrafted monocytes in the retina undergo both epigenetic and transcriptional changes, enabling them to express microglia-like signatures. These findings highlight the need for future research to account for these changes when assessing the roles of monocytes and microglia in CNS pathology.
Collapse
Affiliation(s)
- Jie Liu
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Fengyang Lei
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Bin Yan
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, 139 Middle Renmin Road, Changsha, Hunan, 410011, China
| | - Naiwen Cui
- School of Engineering and Applied Sciences (SEAS), Harvard University, Cambridge, MA, USA
| | - Jyoti Sharma
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Victor Correa
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Lara Roach
- Center for Regenerative Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Savvas Nicolaou
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Kristen Pitts
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - James Chodosh
- Department of Ophthalmology and Visual Sciences, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Daniel E. Maidana
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, IL
| | - Demetrios Vavvas
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Milica A Margeta
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Huidan Zhang
- School of Engineering and Applied Sciences (SEAS), Harvard University, Cambridge, MA, USA
| | - David Weitz
- School of Engineering and Applied Sciences (SEAS), Harvard University, Cambridge, MA, USA
| | - Raul Mostoslavsky
- Center for Regenerative Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Eleftherios I. Paschalis
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
23
|
Napoli D, Orsini N, Salamone G, Calvello MA, Capsoni S, Cattaneo A, Strettoi E. Human NGF "Painless" Ocular Delivery for Retinitis Pigmentosa: An In Vivo Study. eNeuro 2024; 11:ENEURO.0096-24.2024. [PMID: 39293937 PMCID: PMC11412101 DOI: 10.1523/eneuro.0096-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 05/29/2024] [Accepted: 06/03/2024] [Indexed: 09/20/2024] Open
Abstract
Retinitis pigmentosa (RP) is a family of genetically heterogeneous diseases still without a cure. Despite the causative genetic mutation typically not expressed in cone photoreceptors, these cells inevitably degenerate following the primary death of rods, causing blindness. The reasons for the "bystander" degeneration of cones are presently unknown but decrement of survival factors, oxidative stress, and inflammation all play a role. Targeting these generalized biological processes represents a strategy to develop mutation-agnostic therapies for saving vision in large populations of RP individuals. A classical method to support neuronal survival is by employing neurotrophic factors, such as NGF. This study uses painless human NGF (hNGFp), a TrkA receptor-biased variant of the native molecule with lower affinity for nociceptors and limited activity as a pain inducer; the molecule has identical neurotrophic power of the native form but a reduced affinity for the p75NTR receptors, known to trigger apoptosis. hNGFp has a recognized activity on brain microglial cells, which are induced to a phenotype switch from a highly activated to a more homeostatic configuration. hNGFp was administered to RP-like mice in vivo with the aim of decreasing retinal inflammation and also providing retinal neuroprotection. However, the ability of this treatment to counteract the bystander degeneration of cones remained limited.
Collapse
Affiliation(s)
- Debora Napoli
- CNR Neuroscience Institute, Pisa 56124, Italy
- Regional Doctorate School in Neuroscience, University of Florence, Italy
| | - Noemi Orsini
- CNR Neuroscience Institute, Pisa 56124, Italy
- Regional Doctorate School in Neuroscience, University of Florence, Italy
| | | | | | - Simona Capsoni
- Section of Human Physiology, Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara 44121, Italy
| | - Antonino Cattaneo
- Bio@SNS Laboratory of Biology, Scuola Normale Superiore, Pisa, Italy
- Rita Levi-Montalcini European Brain Research Institute (EBRI), Roma 00161, Italy
| | | |
Collapse
|
24
|
Martín-Loro F, Cano-Cano F, Ortega MJ, Cuevas B, Gómez-Jaramillo L, González-Montelongo MDC, Freisenhausen JC, Lara-Barea A, Campos-Caro A, Zubía E, Aguilar-Diosdado M, Arroba AI. Arylphthalide Delays Diabetic Retinopathy via Immunomodulating the Early Inflammatory Response in an Animal Model of Type 1 Diabetes Mellitus. Int J Mol Sci 2024; 25:8440. [PMID: 39126007 PMCID: PMC11313200 DOI: 10.3390/ijms25158440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 07/30/2024] [Accepted: 07/30/2024] [Indexed: 08/12/2024] Open
Abstract
Diabetic retinopathy (DR) is one of the most prevalent secondary complications associated with diabetes. Specifically, Type 1 Diabetes Mellitus (T1D) has an immune component that may determine the evolution of DR by compromising the immune response of the retina, which is mediated by microglia. In the early stages of DR, the permeabilization of the blood-retinal barrier allows immune cells from the peripheral system to interact with the retinal immune system. The use of new bioactive molecules, such as 3-(2,4-dihydroxyphenyl)phthalide (M9), with powerful anti-inflammatory activity, might represent an advance in the treatment of diseases like DR by targeting the immune systems responsible for its onset and progression. Our research aimed to investigate the molecular mechanisms involved in the interaction of specific cells of the innate immune system during the progression of DR and the reduction in inflammatory processes contributing to the pathology. In vitro studies were conducted exposing Bv.2 microglial and Raw264.7 macrophage cells to proinflammatory stimuli for 24 h, in the presence or absence of M9. Ex vivo and in vivo approaches were performed in BB rats, an animal model for T1D. Retinal explants from BB rats were cultured with M9. Retinas from BB rats treated for 15 days with M9 via intraperitoneal injection were analyzed to determine survival, cellular signaling, and inflammatory markers using qPCR, Western blot, or immunofluorescence approaches. Retinal structure images were acquired via Spectral-Domain-Optical Coherence Tomography (SD-OCT). Our results show that the treatment with M9 significantly reduces inflammatory processes in in vitro, ex vivo, and in vivo models of DR. M9 works by inhibiting the proinflammatory responses during DR progression mainly affecting immune cell responses. It also induces an anti-inflammatory response, primarily mediated by microglial cells, leading to the synthesis of Arginase-1 and Hemeoxygenase-1(HO-1). Ultimately, in vivo administration of M9 preserves the retinal integrity from the degeneration associated with DR progression. Our findings demonstrate a specific interaction between both retinal and systemic immune cells in the progression of DR, with a differential response to treatment, mainly driven by microglia in the anti-inflammatory action. In vivo treatment with M9 induces a switch in immune cell phenotypes and functions that contributes to delaying the DR progression, positioning microglial cells as a new and specific therapeutic target in DR.
Collapse
Affiliation(s)
- Francisco Martín-Loro
- Diabetes Mellitus Laboratory, Instituto de Investigación e Innovación en Ciencias Biomédicas de la Provincia de Cádiz (INiBICA), Hospital Universitario Puerta del Mar, 11009 Cádiz, Spain; (F.M.-L.); (F.C.-C.); (B.C.); (L.G.-J.); (M.d.C.G.-M.); (M.A.-D.)
| | - Fátima Cano-Cano
- Diabetes Mellitus Laboratory, Instituto de Investigación e Innovación en Ciencias Biomédicas de la Provincia de Cádiz (INiBICA), Hospital Universitario Puerta del Mar, 11009 Cádiz, Spain; (F.M.-L.); (F.C.-C.); (B.C.); (L.G.-J.); (M.d.C.G.-M.); (M.A.-D.)
| | - María J. Ortega
- Departamento de Química Orgánica, Facultad de Ciencias del Mar y Ambientales, Universidad de Cádiz, 11510 Puerto Real, Spain; (M.J.O.); (E.Z.)
| | - Belén Cuevas
- Diabetes Mellitus Laboratory, Instituto de Investigación e Innovación en Ciencias Biomédicas de la Provincia de Cádiz (INiBICA), Hospital Universitario Puerta del Mar, 11009 Cádiz, Spain; (F.M.-L.); (F.C.-C.); (B.C.); (L.G.-J.); (M.d.C.G.-M.); (M.A.-D.)
- Departamento de Química Orgánica, Facultad de Ciencias del Mar y Ambientales, Universidad de Cádiz, 11510 Puerto Real, Spain; (M.J.O.); (E.Z.)
| | - Laura Gómez-Jaramillo
- Diabetes Mellitus Laboratory, Instituto de Investigación e Innovación en Ciencias Biomédicas de la Provincia de Cádiz (INiBICA), Hospital Universitario Puerta del Mar, 11009 Cádiz, Spain; (F.M.-L.); (F.C.-C.); (B.C.); (L.G.-J.); (M.d.C.G.-M.); (M.A.-D.)
| | - María del Carmen González-Montelongo
- Diabetes Mellitus Laboratory, Instituto de Investigación e Innovación en Ciencias Biomédicas de la Provincia de Cádiz (INiBICA), Hospital Universitario Puerta del Mar, 11009 Cádiz, Spain; (F.M.-L.); (F.C.-C.); (B.C.); (L.G.-J.); (M.d.C.G.-M.); (M.A.-D.)
| | - Jan Cedric Freisenhausen
- Dermatology and Venereology Division, Department of Medicine, Karolinska Institute, SE-171 77 Solna, Sweden;
- Center for Molecular Medicine, Karolinska University Hospital, SE-171 76 Solna, Sweden
| | - Almudena Lara-Barea
- Department of Endocrinology and Metabolism, University Hospital Puerta del Mar, 11009 Cádiz, Spain;
| | - Antonio Campos-Caro
- Área Genética, Departamento Biomedicina Biotecnología y Salud Pública, Universidad de Cádiz, 11510 Puerto Real, Spain;
| | - Eva Zubía
- Departamento de Química Orgánica, Facultad de Ciencias del Mar y Ambientales, Universidad de Cádiz, 11510 Puerto Real, Spain; (M.J.O.); (E.Z.)
| | - Manuel Aguilar-Diosdado
- Diabetes Mellitus Laboratory, Instituto de Investigación e Innovación en Ciencias Biomédicas de la Provincia de Cádiz (INiBICA), Hospital Universitario Puerta del Mar, 11009 Cádiz, Spain; (F.M.-L.); (F.C.-C.); (B.C.); (L.G.-J.); (M.d.C.G.-M.); (M.A.-D.)
- Department of Endocrinology and Metabolism, University Hospital Puerta del Mar, 11009 Cádiz, Spain;
| | - Ana I. Arroba
- Diabetes Mellitus Laboratory, Instituto de Investigación e Innovación en Ciencias Biomédicas de la Provincia de Cádiz (INiBICA), Hospital Universitario Puerta del Mar, 11009 Cádiz, Spain; (F.M.-L.); (F.C.-C.); (B.C.); (L.G.-J.); (M.d.C.G.-M.); (M.A.-D.)
- Department of Endocrinology and Metabolism, University Hospital Puerta del Mar, 11009 Cádiz, Spain;
| |
Collapse
|
25
|
Zeng Y, Wen F, Zhuang X, He G, Zhang X. EPIRETINAL MACROPHAGE-LIKE CELLS ON OPTICAL COHERENCE TOMOGRAPHY: Potential Inflammatory Imaging Biomarker of Severity in Diabetic Retinopathy. Retina 2024; 44:1314-1322. [PMID: 38478763 DOI: 10.1097/iae.0000000000004100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/26/2024]
Abstract
PURPOSE To investigate the clinical value of epiretinal macrophage-like cells (eMLCs) in different stages of diabetic retinopathy (DR), including diabetic macular edema (DME). METHODS One hundred and seventy-six eyes of 176 patients, including 31 diabetes eyes without retinopathy, 24 with mild nonproliferative DR (NPDR), 37 with moderate NPDR, 32 with severe NPDR, and 52 with proliferative DR, were compared with 30 healthy controls. The optic nerve head and macular region were examined using en face optical coherence tomography and optical coherence tomography angiography. A previously described technique was employed to measure the eMLCs on the inner limiting membrane. RESULTS The optic nerve head and macular eMLC densities increased with worsening DR stage and were higher in moderate, severe NPDR, and proliferative DR, especially in eyes with DME (all adjusted P < 0.01). The optic nerve head and macular eMLC densities in the DME groups were higher than those in their corresponding non-DME groups at the same DR stage (all P < 0.05). The average macular thickness was correlated with macular eMLC density (standardized β = 0.661, P < 0.001) in patients with diabetes. The proportion of eyes with larger and plumper eMLCs increased with worsening DR and was higher in moderate, severe NPDR, proliferative DR, and eyes with DME ( P = 0.018, P < 0.001, P < 0.001, and P < 0.001, respectively). CONCLUSION Increased density along with morphological changes of eMLCs is observed beginning with moderate NPDR and correlates with the progression of DR, including DME.
Collapse
Affiliation(s)
- Yunkao Zeng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China ; and
- Department of Ophthalmology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Feng Wen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China ; and
| | - Xuenan Zhuang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China ; and
| | - Guiqin He
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China ; and
| | - Xiongze Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China ; and
| |
Collapse
|
26
|
Hu A, Schmidt MHH, Heinig N. Microglia in retinal angiogenesis and diabetic retinopathy. Angiogenesis 2024; 27:311-331. [PMID: 38564108 PMCID: PMC11303477 DOI: 10.1007/s10456-024-09911-1] [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: 02/18/2024] [Indexed: 04/04/2024]
Abstract
Diabetic retinopathy has a high probability of causing visual impairment or blindness throughout the disease progression and is characterized by the growth of new blood vessels in the retina at an advanced, proliferative stage. Microglia are a resident immune population in the central nervous system, known to play a crucial role in regulating retinal angiogenesis in both physiological and pathological conditions, including diabetic retinopathy. Physiologically, they are located close to blood vessels and are essential for forming new blood vessels (neovascularization). In diabetic retinopathy, microglia become widely activated, showing a distinct polarization phenotype that leads to their accumulation around neovascular tufts. These activated microglia induce pathogenic angiogenesis through the secretion of various angiogenic factors and by regulating the status of endothelial cells. Interestingly, some subtypes of microglia simultaneously promote the regression of neovascularization tufts and normal angiogenesis in neovascularization lesions. Modulating the state of microglial activation to ameliorate neovascularization thus appears as a promising potential therapeutic approach for managing diabetic retinopathy.
Collapse
Affiliation(s)
- Aiyan Hu
- Institute of Anatomy, Medical Faculty Carl Gustav Carus, Technische Universität Dresden School of Medicine, Fetscherstr 74, 01307, Dresden, Germany
| | - Mirko H H Schmidt
- Institute of Anatomy, Medical Faculty Carl Gustav Carus, Technische Universität Dresden School of Medicine, Fetscherstr 74, 01307, Dresden, Germany.
| | - Nora Heinig
- Institute of Anatomy, Medical Faculty Carl Gustav Carus, Technische Universität Dresden School of Medicine, Fetscherstr 74, 01307, Dresden, Germany.
| |
Collapse
|
27
|
Lebon C, Malaise D, Rimbert N, Billet M, Ramasamy G, Villaret J, Pouzoulet F, Matet A, Behar-Cohen F. Role of inflammation in a rat model of radiation retinopathy. J Neuroinflammation 2024; 21:162. [PMID: 38915029 PMCID: PMC11197380 DOI: 10.1186/s12974-024-03151-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 06/11/2024] [Indexed: 06/26/2024] Open
Abstract
Radiation retinopathy (RR) is a major side effect of ocular tumor treatment by plaque brachytherapy or proton beam therapy. RR manifests as delayed and progressive microvasculopathy, ischemia and macular edema, ultimately leading to vision loss, neovascular glaucoma, and, in extreme cases, secondary enucleation. Intravitreal anti-VEGF agents, steroids and laser photocoagulation have limited effects on RR. The role of retinal inflammation and its contribution to the microvascular damage occurring in RR remain incompletely understood. To explore cellular and vascular events after irradiation, we analyzed their time course at 1 week, 1 month and 6 months after rat eyes received 45 Gy X-beam photons. Müller glial cells, astrocytes and microglia were rapidly activated, and these markers of retinal inflammation persisted for 6 months after irradiation. This was accompanied by early cell death in the outer retina, which persisted at later time points, leading to retinal thinning. A delayed loss of small retinal capillaries and retinal hypoxia were observed after 6 months, indicating inner blood‒retinal barrier (BRB) alteration but without cell death in the inner retina. Moreover, activated microglial cells invaded the entire retina and surrounded retinal vessels, suggesting the role of inflammation in vascular alteration and in retinal cell death. Radiation also triggered early and persistent invasion of the retinal pigment epithelium by microglia and macrophages, contributing to outer BRB disruption. This study highlights the role of progressive and long-lasting inflammatory mechanisms in RR development and demonstrates the relevance of this rat model to investigate human pathology.
Collapse
Affiliation(s)
- Cécile Lebon
- Centre de Recherche des Cordeliers, INSERM UMRS 1138, Sorbonne Université, Physiopathology of Ocular Diseases: Therapeutic Innovations, 15 rue de l'école de Médecine, Paris, 75006, France
| | - Denis Malaise
- Centre de Recherche des Cordeliers, INSERM UMRS 1138, Sorbonne Université, Physiopathology of Ocular Diseases: Therapeutic Innovations, 15 rue de l'école de Médecine, Paris, 75006, France
- Ocular Oncology Department, Institut Curie, 26 rue d'Ulm, Paris, 75005, France
| | - Nicolas Rimbert
- Centre de Recherche des Cordeliers, INSERM UMRS 1138, Sorbonne Université, Physiopathology of Ocular Diseases: Therapeutic Innovations, 15 rue de l'école de Médecine, Paris, 75006, France
| | - Manon Billet
- Translational Research Department, Experimental Radiotherapy Platform, Institut Curie, Université Paris Saclay, 15 Rue Georges Clemenceau, Orsay, 91400, France
| | - Gabriel Ramasamy
- Translational Research Department, Experimental Radiotherapy Platform, Institut Curie, Université Paris Saclay, 15 Rue Georges Clemenceau, Orsay, 91400, France
| | - Jérémie Villaret
- Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, Paris, 75012, France
- Centre Rétine Gallien, Bordeaux, 33000, France
| | - Frédéric Pouzoulet
- Translational Research Department, Experimental Radiotherapy Platform, Institut Curie, Université Paris Saclay, 15 Rue Georges Clemenceau, Orsay, 91400, France
| | - Alexandre Matet
- Centre de Recherche des Cordeliers, INSERM UMRS 1138, Sorbonne Université, Physiopathology of Ocular Diseases: Therapeutic Innovations, 15 rue de l'école de Médecine, Paris, 75006, France.
- Ocular Oncology Department, Institut Curie, 26 rue d'Ulm, Paris, 75005, France.
- Université Paris Cité, 15 rue de l'école de Médecine, Paris, 75006, France.
| | - Francine Behar-Cohen
- Centre de Recherche des Cordeliers, INSERM UMRS 1138, Sorbonne Université, Physiopathology of Ocular Diseases: Therapeutic Innovations, 15 rue de l'école de Médecine, Paris, 75006, France.
- Assistance Publique, Hôpitaux de Paris, Hôpital Cochin, 27 rue du Faubourg Saint-Jacques, Ophtalmopole, Paris, 75014, France.
| |
Collapse
|
28
|
Rutan Woods CT, Makia MS, Lewis TR, Crane R, Zeibak S, Yu P, Kakakhel M, Castillo CM, Arshavsky VY, Naash MI, Al-Ubaidi MR. Downregulation of rhodopsin is an effective therapeutic strategy in ameliorating peripherin-2-associated inherited retinal disorders. Nat Commun 2024; 15:4756. [PMID: 38834544 PMCID: PMC11150396 DOI: 10.1038/s41467-024-48846-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 05/15/2024] [Indexed: 06/06/2024] Open
Abstract
Given the absence of approved treatments for pathogenic variants in Peripherin-2 (PRPH2), it is imperative to identify a universally effective therapeutic target for PRPH2 pathogenic variants. To test the hypothesis that formation of the elongated discs in presence of PRPH2 pathogenic variants is due to the presence of the full complement of rhodopsin in absence of the required amounts of functional PRPH2. Here we demonstrate the therapeutic potential of reducing rhodopsin levels in ameliorating disease phenotype in knockin models for p.Lys154del (c.458-460del) and p.Tyr141Cys (c.422 A > G) in PRPH2. Reducing rhodopsin levels improves physiological function, mitigates the severity of disc abnormalities, and decreases retinal gliosis. Additionally, intravitreal injections of a rhodopsin-specific antisense oligonucleotide successfully enhance the physiological function of photoreceptors and improves the ultrastructure of discs in mutant mice. Presented findings shows that reducing rhodopsin levels is an effective therapeutic strategy for the treatment of inherited retinal degeneration associated with PRPH2 pathogenic variants.
Collapse
Affiliation(s)
| | - Mustafa S Makia
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Tylor R Lewis
- Department of Ophthalmology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Ryan Crane
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Stephanie Zeibak
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Paul Yu
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Mashal Kakakhel
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Carson M Castillo
- Department of Ophthalmology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Vadim Y Arshavsky
- Department of Ophthalmology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Muna I Naash
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA.
| | - Muayyad R Al-Ubaidi
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA.
| |
Collapse
|
29
|
Du SW, Komirisetty R, Lewandowski D, Choi EH, Panas D, Suh S, Tabaka M, Radu RA, Palczewski K. Conditional deletion of miR-204 and miR-211 in murine retinal pigment epithelium results in retinal degeneration. J Biol Chem 2024; 300:107344. [PMID: 38705389 PMCID: PMC11140208 DOI: 10.1016/j.jbc.2024.107344] [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: 03/15/2024] [Revised: 04/18/2024] [Accepted: 04/27/2024] [Indexed: 05/07/2024] Open
Abstract
MicroRNAs (miRs) are short, evolutionarily conserved noncoding RNAs that canonically downregulate expression of target genes. The miR family composed of miR-204 and miR-211 is among the most highly expressed miRs in the retinal pigment epithelium (RPE) in both mouse and human and also retains high sequence identity. To assess the role of this miR family in the developed mouse eye, we generated two floxed conditional KO mouse lines crossed to the RPE65-ERT2-Cre driver mouse line to perform an RPE-specific conditional KO of this miR family in adult mice. After Cre-mediated deletion, we observed retinal structural changes by optical coherence tomography; dysfunction and loss of photoreceptors by retinal imaging; and retinal inflammation marked by subretinal infiltration of immune cells by imaging and immunostaining. Single-cell RNA sequencing of diseased RPE and retinas showed potential miR-regulated target genes, as well as changes in noncoding RNAs in the RPE, rod photoreceptors, and Müller glia. This work thus highlights the role of miR-204 and miR-211 in maintaining RPE function and how the loss of miRs in the RPE exerts effects on the neural retina, leading to inflammation and retinal degeneration.
Collapse
Affiliation(s)
- Samuel W Du
- Gavin Herbert Eye Institute-Center for Translational Vision Research, Department of Ophthalmology, University of California, Irvine, California, USA; Department of Physiology and Biophysics, University of California, Irvine, California, USA.
| | - Ravikiran Komirisetty
- Department of Ophthalmology and UCLA Stein Eye Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - Dominik Lewandowski
- Gavin Herbert Eye Institute-Center for Translational Vision Research, Department of Ophthalmology, University of California, Irvine, California, USA
| | - Elliot H Choi
- Gavin Herbert Eye Institute-Center for Translational Vision Research, Department of Ophthalmology, University of California, Irvine, California, USA
| | - Damian Panas
- International Centre for Translational Eye Research, Warsaw, Poland; Institute of Physical Chemistry, Polish Academy of Sciences, Warsaw, Poland
| | - Susie Suh
- Gavin Herbert Eye Institute-Center for Translational Vision Research, Department of Ophthalmology, University of California, Irvine, California, USA
| | - Marcin Tabaka
- International Centre for Translational Eye Research, Warsaw, Poland; Institute of Physical Chemistry, Polish Academy of Sciences, Warsaw, Poland
| | - Roxana A Radu
- Department of Ophthalmology and UCLA Stein Eye Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - Krzysztof Palczewski
- Gavin Herbert Eye Institute-Center for Translational Vision Research, Department of Ophthalmology, University of California, Irvine, California, USA; Department of Physiology and Biophysics, University of California, Irvine, California, USA; Department of Chemistry, University of California, Irvine, Irvine, California, USA; Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, California, USA.
| |
Collapse
|
30
|
Tan LX, Oertel FC, Cheng A, Cobigo Y, Keihani A, Bennett DJ, Abdelhak A, Montes SC, Chapman M, Chen RY, Cordano C, Ward ME, Casaletto K, Kramer JH, Rosen HJ, Boxer A, Miller BL, Green AJ, Elahi FM, Lakkaraju A. Targeting complement C3a receptor resolves mitochondrial hyperfusion and subretinal microglial activation in progranulin-deficient frontotemporal dementia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.29.595206. [PMID: 38854134 PMCID: PMC11160746 DOI: 10.1101/2024.05.29.595206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Mutations in progranulin ( GRN ) cause frontotemporal dementia ( GRN -FTD) due to deficiency of the pleiotropic protein progranulin. GRN -FTD exhibits diverse pathologies including lysosome dysfunction, lipofuscinosis, microgliosis, and neuroinflammation. Yet, how progranulin loss causes disease remains unresolved. Here, we report that non-invasive retinal imaging of GRN -FTD patients revealed deficits in photoreceptors and the retinal pigment epithelium (RPE) that correlate with cognitive decline. Likewise, Grn -/- mice exhibit early RPE dysfunction, microglial activation, and subsequent photoreceptor loss. Super-resolution live imaging and transcriptomic analyses identified RPE mitochondria as an early driver of retinal dysfunction. Loss of mitochondrial fission protein 1 (MTFP1) in Grn -/- RPE causes mitochondrial hyperfusion and bioenergetic defects, leading to NF-kB-mediated activation of complement C3a-C3a receptor signaling, which drives further mitochondrial hyperfusion and retinal inflammation. C3aR antagonism restores RPE mitochondrial integrity and limits subretinal microglial activation. Our study identifies a previously unrecognized mechanism by which progranulin modulates mitochondrial integrity and complement-mediated neuroinflammation.
Collapse
|
31
|
Albalawi FE, Alsharif I, Moawadh MS, Alkhoshaiban A, Falah Alshehri F, Albalawi AE, Althobaiti NA, Alharbi ZM, Almohaimeed HM. Immunomodulatory effects of Kaempferol on microglial and Macrophage cells during the progression of diabetic retinopathy. Int Immunopharmacol 2024; 133:112021. [PMID: 38626549 DOI: 10.1016/j.intimp.2024.112021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 04/01/2024] [Accepted: 04/03/2024] [Indexed: 04/18/2024]
Abstract
BACKGROUND Diabetic retinopathy (DR) stands as a prevalent secondary complication of diabetes, notably Type 1 Diabetes Mellitus (T1D), characterized by immune system involvement potentially impacting the retinal immune response mediated by microglia. Early stages of DR witness blood-retinal barrier permeabilization, facilitating peripheral immune cell interaction with the retinal immune system. Kaempferol (Kae), known for its potent anti-inflammatory activity, presents a promising avenue in DR treatment by targeting the immune mechanisms underlying its onset and progression. Our investigation delves into the molecular intricacies of innate immune cell interaction during DR progression and the attenuation of inflammatory processes pivotal to its pathology. METHODS Employing in vitro studies, we exposed HAPI microglial and J774.A1 macrophage cells to pro-inflammatory stimuli in the presence or absence of Kae. Ex vivo and in vivo experiments utilized BB rats, a T1D animal model. Retinal explants from BB rats were cultured with Kae, while intraperitoneal Kae injections were administered to BB rats for 15 days. Quantitative PCR, Western blotting, immunofluorescence, and Spectral Domain - Optical Coherence Tomography (SD-OCT) facilitated survival assessment, cellular signaling analysis, and inflammatory marker determination. RESULTS Results demonstrate Kae significantly mitigates inflammatory processes across in vitro, ex vivo, and in vivo DR models, primarily targeting immune cell responses. Kae administration notably inhibits proinflammatory responses during DR progression while promoting an anti-inflammatory milieu, chiefly through microglia-mediated synthesis of Arginase-1 and Hemeoxygenase-1(HO-1). In vivo, Kae administration effectively preserves retinal integrity amid DR progression. CONCLUSIONS Our findings elucidate the interplay between retinal and systemic immune cells in DR progression, underscoring a differential treatment response predominantly orchestrated by microglia's anti-inflammatory action. Kae treatment induces a phenotypic and functional shift in immune cells, delaying DR progression, thereby spotlighting microglial cells as a promising therapeutic target in DR management.
Collapse
Affiliation(s)
- Fahad Eid Albalawi
- Regional laboratory, blood bank and poisons centre, Sakaka 72346, Saudi Arabia; Medical College, Fahad Bin Sultan University, Tabuk 47721, Saudi Arabia.
| | - Ifat Alsharif
- Department of Biology, Jamoum University College, Umm Al-Qura University, 21955, Makkah, Saudi Arabia
| | - Mamdoh S Moawadh
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk 71491, Saudi Arabia
| | | | - Faez Falah Alshehri
- Department of Medical Laboratories, College of Applied Medical Sciences, Ad Dawadimi-17464, Shaqra University, Saudi Arabia
| | - Aishah E Albalawi
- Faculty of science, Department of Biology, University of Tabuk, Tabuk 47913, Saudi Arabia
| | - Norah A Althobaiti
- Biology Department, College of Science and Humanities, Al Quwaiiyah, Shaqra University, Al Quwaiiyah 19257, Saudi Arabia
| | - Zeyad M Alharbi
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Hailah M Almohaimeed
- Department of Basic Science, College of Medicine, Princess Nourah bint Abdulrahman University, P.O.Box 84428, Riyadh 11671, Saudi Arabia
| |
Collapse
|
32
|
Zhao N, Hao XN, Huang JM, Song ZM, Tao Y. Crosstalk Between Microglia and Müller Glia in the Age-Related Macular Degeneration: Role and Therapeutic Value of Neuroinflammation. Aging Dis 2024; 15:1132-1154. [PMID: 37728589 PMCID: PMC11081163 DOI: 10.14336/ad.2023.0823-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 08/23/2023] [Indexed: 09/21/2023] Open
Abstract
Age-related macular degeneration (AMD) is a progressive neurodegeneration disease that causes photoreceptor demise and vision impairments. In AMD pathogenesis, the primary death of retinal neurons always leads to the activation of resident microglia. The migration of activated microglia to the ongoing retinal lesion and their morphological transformation from branching to ameboid-like are recognized as hallmarks of AMD pathogenesis. Activated microglia send signals to Müller cells and promote them to react correspondingly to damaging stimulus. Müller cells are a type of neuroglia cells that maintain the normal function of retinal neurons, modulating innate inflammatory responses, and stabilize retinal structure. Activated Müller cells can accelerate the progression of AMD by damaging neurons and blood vessels. Therefore, the crosstalk between microglia and Müller cells plays a homeostatic role in maintaining the retinal environment, and this interaction is complicatedly modulated. In particular, the mechanism of mutual regulation between the two glia populations is complex under pathological conditions. This paper reviews recent findings on the crosstalk between microglia and Müller glia during AMD pathology process, with special emphasis on its therapeutic potentials.
Collapse
Affiliation(s)
- Na Zhao
- Henan Eye Institute, Henan Eye Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, China.
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Xiao-Na Hao
- Henan Eye Institute, Henan Eye Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, China.
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Jie-Min Huang
- Henan Eye Institute, Henan Eye Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, China.
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Zong-Ming Song
- Henan Eye Institute, Henan Eye Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, China.
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Ye Tao
- Henan Eye Institute, Henan Eye Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, China.
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
33
|
Pichi F, Neri P, Aljneibi S, Hay S, Chaudhry H, Carreño E. Vitreoretinal Interface Cells Correlate In Vivo With Uveitis Activity and Decrease With Anti-Inflammatory Treatment. Transl Vis Sci Technol 2024; 13:15. [PMID: 38767904 PMCID: PMC11114619 DOI: 10.1167/tvst.13.5.15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 04/22/2024] [Indexed: 05/22/2024] Open
Abstract
Purpose To highlight the utility of en face swept-source optical coherence tomography angiography (SS-OCTA) in assessing vitreoretinal interface cells (VRICs) of patients with active uveitis and their dynamics. Methods In this prospective, single-center study, 20 eyes from patients with active uveitis were analyzed using six 6 × 6-mm macular scans at three time points: active inflammation (baseline), clinically improving (T1), and resolved inflammation (T2). VRICs were visualized using 3-µm en face OCT slabs on the inner limiting membrane. The variation of VRIC number, density, and size over time was assessed, and VRIC measurements were compared with clinical grading. Results At baseline, the VRIC count was significantly higher (552.5 VRICs) than that of the healthy controls (478.2 VRICs), with a density of 15.3 cells/mm2. VRIC number decreased significantly to 394.8 (P = 0.007) at T1, with a density of 10.9 cells/mm2 (P = 0.007). VRIC size reduced from 6.8 µm to 6.3 µm at T1 (P = 0.009) and remained stable at T2 (P = 0.3). Correlation coefficients between inflammatory parameters (anterior chamber cells and National Eye Institute vitreous haze), and VRIC count indicated a positive correlation at baseline (r = 0.53), weakening at T1 (r = 0.36), and becoming negative at T2 (r = -0.24). Conclusions En face SS-OCTA revealed increased VRIC number and size in active uveitis, likely due to monocyte recruitment. Post-inflammation control, VRIC number, size, and density significantly decreased, returning to normal despite residual anterior chamber cells or vitreous haze. Translational Relevance Visualization of VRICs by in vivo OCT opens up new opportunities for therapeutic targets.
Collapse
Affiliation(s)
- Francesco Pichi
- Eye Institute, Cleveland Clinic Abu Dhabi, Abu Dhabi, United Arab Emirates
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Piergiorgio Neri
- Eye Institute, Cleveland Clinic Abu Dhabi, Abu Dhabi, United Arab Emirates
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Shaikha Aljneibi
- Eye Institute, Cleveland Clinic Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Steven Hay
- Eye Institute, Cleveland Clinic Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Hannah Chaudhry
- Eye Institute, Cleveland Clinic Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Ester Carreño
- University Hospital Fundación Jiménez Díaz, Madrid, Spain
- University Hospital Rey Juan Carlos, Madrid, Spain
- Instituto de Investigación Sanitaria Fundación Jiménez Díaz, Madrid, Spain
| |
Collapse
|
34
|
Lv L, Zhou LX, Jiang FF. Study on the mechanism of 20-hydroxyeicosatetraenoic acid in retinal ischemia-reperfusion injury. Indian J Ophthalmol 2024; 72:S441-S447. [PMID: 38389249 PMCID: PMC467026 DOI: 10.4103/ijo.ijo_1466_23] [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: 06/05/2023] [Revised: 10/24/2023] [Accepted: 11/01/2023] [Indexed: 02/24/2024] Open
Abstract
PURPOSE To explore the effect of 20-hydroxyeicosatetraenoic acid (20-HETE) on retinal ischemia-reperfusion injury (RIRI) and the protective effect of N-hydroxy-N'-(4-n-butyl-2-methylphenyl)formamidine (HET0016) on RIRI. METHODS Male Sprague-Dawley rats were randomly divided into the normal control group, experimental model group (RIRI group), experimental solvent group (RIRI + solvent group), and experimental treatment group (RIRI + HET0016 group). RESULTS The levels of 20-HETE, tumor necrosis factor-α (TNF-α), and interleukin-1β (IL-1β) in the retina of rats at 24 h after reperfusion were measured by enzyme-linked immunosorbent assay. Hematoxylin-eosin staining was used to observe the retinal morphological and thickness changes at 24 h, 48 h, and 7 days after reperfusion. The number and localized expression of matrix metalloproteinase-9-positive cells in the retina of the rats at 24 h after reperfusion and the activation and localized expression of retinal microglia at 48 h after reperfusion were measured using an immunohistochemical method. The nuclear metastasis of nuclear factor kappa-B (NF-κB, p65) cells at 24 h after reperfusion was observed using an immunofluorescence method. CONCLUSION Overall, 20-HETE might activate microglia to aggravate RIRI by the NF-κB pathway, but HET0016 has significant protective effects for the retina.
Collapse
Affiliation(s)
- Liang Lv
- Department of Ophthalmology, The Fifth Clinical College of Zhengzhou University, Zhengzhou, China
| | - Li-Xiao Zhou
- Department of Ophthalmology, The Fifth Clinical College of Zhengzhou University, Zhengzhou, China
| | - Fei-Fei Jiang
- Department of Ophthalmology, The Fifth Clinical College of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
35
|
Li X, Sedlacek M, Nath A, Szatko KP, Grimes WN, Diamond JS. A metabotropic glutamate receptor agonist enhances visual signal fidelity in a mouse model of retinitis pigmentosa. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.30.591881. [PMID: 38746092 PMCID: PMC11092665 DOI: 10.1101/2024.04.30.591881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Many inherited retinal diseases target photoreceptors, which transduce light into a neural signal that is processed by the downstream visual system. As photoreceptors degenerate, physiological and morphological changes to retinal synapses and circuitry reduce sensitivity and increase noise, degrading visual signal fidelity. Here, we pharmacologically targeted the first synapse in the retina in an effort to reduce circuit noise without sacrificing visual sensitivity. We tested a strategy to partially replace the neurotransmitter lost when photoreceptors die with an agonist of receptors that ON bipolars cells use to detect glutamate released from photoreceptors. In rd10 mice, which express a photoreceptor mutation that causes retinitis pigmentosa (RP), we found that a low dose of the mGluR6 agonist l-2-amino-4-phosphonobutyric acid (L-AP4) reduced pathological noise induced by photoreceptor degeneration. After making in vivo electroretinogram recordings in rd10 mice to characterize the developmental time course of visual signal degeneration, we examined effects of L-AP4 on sensitivity and circuit noise by recording in vitro light-evoked responses from individual retinal ganglion cells (RGCs). L-AP4 decreased circuit noise evident in RGC recordings without significantly reducing response amplitudes, an effect that persisted over the entire time course of rod photoreceptor degeneration. Subsequent in vitro recordings from rod bipolar cells (RBCs) showed that RBCs are more depolarized in rd10 retinas, likely contributing to downstream circuit noise and reduced synaptic gain, both of which appear to be ameliorated by hyperpolarizing RBCs with L-AP4. These beneficial effects may reduce pathological circuit remodeling and preserve the efficacy of therapies designed to restore vision.
Collapse
Affiliation(s)
- Xiaoyi Li
- Synaptic Physiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA 20892
- Department of Biology, Johns Hopkins University, Baltimore, MD, USA 21218
| | - Miloslav Sedlacek
- Synaptic Physiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA 20892
| | - Amurta Nath
- Synaptic Physiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA 20892
| | - Klaudia P. Szatko
- Synaptic Physiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA 20892
| | - William N. Grimes
- Synaptic Physiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA 20892
| | - Jeffrey S. Diamond
- Synaptic Physiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA 20892
| |
Collapse
|
36
|
Wang D, Pu Y, Tan S, Wang X, Zeng L, Lei J, Gao X, Li H. Identification of immune-related biomarkers for glaucoma using gene expression profiling. Front Genet 2024; 15:1366453. [PMID: 38694874 PMCID: PMC11062407 DOI: 10.3389/fgene.2024.1366453] [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: 01/06/2024] [Accepted: 04/03/2024] [Indexed: 05/04/2024] Open
Abstract
Introduction: Glaucoma, a principal cause of irreversible vision loss, is characterized by intricate optic neuropathy involving significant immune mechanisms. This study seeks to elucidate the molecular and immune complexities of glaucoma, aiming to improve our understanding of its pathogenesis. Methods: Gene expression profiles from glaucoma patients were analyzed to identify immune-related differentially expressed genes (DEGs). Techniques used were weighted gene co-expression network analysis (WGCNA) for network building, machine learning algorithms for biomarker identification, establishment of subclusters related to immune reactions, and single-sample gene set enrichment analysis (ssGSEA) to explore hub genes' relationships with immune cell infiltration and immune pathway activation. Validation was performed using an NMDA-induced excitotoxicity model and RT-qPCR for hub gene expression measurement. Results: The study identified 409 DEGs differentiating healthy individuals from glaucoma patients, highlighting the immune response's significance in disease progression. Immune cell infiltration analysis revealed elevated levels of activated dendritic cells, natural killer cells, monocytes, and immature dendritic cells in glaucoma samples. Three hub genes, CD40LG, TEK, and MDK, were validated as potential diagnostic biomarkers for high-risk glaucoma patients, showing increased expression in the NMDA-induced excitotoxicity model. Discussion: The findings propose the three identified immune-related genes (IRGs) as novel diagnostic markers for glaucoma, offering new insights into the disease's pathogenesis and potential therapeutic targets. The strong correlation between these IRGs and immune responses underscores the intricate role of immunity in glaucoma, suggesting a shift in the approach to its diagnosis and treatment.
Collapse
Affiliation(s)
- Dangdang Wang
- Department of Ophthalmology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for the Prevention and Treatment of Major Blinding Eye Diseases, Chongqing Eye Institute, Chongqing Branch of National Clinical Research Center for Ocular Diseases, Chongqing, China
| | - Yanyu Pu
- Department of Ophthalmology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for the Prevention and Treatment of Major Blinding Eye Diseases, Chongqing Eye Institute, Chongqing Branch of National Clinical Research Center for Ocular Diseases, Chongqing, China
| | - Sisi Tan
- Department of Ophthalmology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for the Prevention and Treatment of Major Blinding Eye Diseases, Chongqing Eye Institute, Chongqing Branch of National Clinical Research Center for Ocular Diseases, Chongqing, China
| | - Xiaochen Wang
- Department of Ophthalmology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for the Prevention and Treatment of Major Blinding Eye Diseases, Chongqing Eye Institute, Chongqing Branch of National Clinical Research Center for Ocular Diseases, Chongqing, China
| | - Lihong Zeng
- Department of Ophthalmology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for the Prevention and Treatment of Major Blinding Eye Diseases, Chongqing Eye Institute, Chongqing Branch of National Clinical Research Center for Ocular Diseases, Chongqing, China
| | - Junqin Lei
- Department of Ophthalmology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for the Prevention and Treatment of Major Blinding Eye Diseases, Chongqing Eye Institute, Chongqing Branch of National Clinical Research Center for Ocular Diseases, Chongqing, China
| | - Xi Gao
- Department of Ophthalmology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for the Prevention and Treatment of Major Blinding Eye Diseases, Chongqing Eye Institute, Chongqing Branch of National Clinical Research Center for Ocular Diseases, Chongqing, China
| | - Hong Li
- Department of Ophthalmology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for the Prevention and Treatment of Major Blinding Eye Diseases, Chongqing Eye Institute, Chongqing Branch of National Clinical Research Center for Ocular Diseases, Chongqing, China
| |
Collapse
|
37
|
Sun Y, Liang JJ, Xu J, Zhou K, Fu C, Chen SL, Yang R, Ng TK, Liu Q, Zhang M. Oxidized low-density lipoprotein changes the inflammatory status and metabolomics profiles in human and mouse macrophages and microglia. Heliyon 2024; 10:e28806. [PMID: 38617955 PMCID: PMC11015420 DOI: 10.1016/j.heliyon.2024.e28806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 03/22/2024] [Accepted: 03/25/2024] [Indexed: 04/16/2024] Open
Abstract
The conjunctiva of primary open angle glaucoma patients showed high level of oxidized low-density lipoprotein (ox-LDL), which is associated with the inflammatory response. Microglia and macrophages are the immune cells involved in retinal ganglion cell survival regulation; yet, their roles of the ox-LDL-induced inflammation in glaucoma remain elusive. Here we aimed to investigate the lipid uptake, inflammatory cytokine expression, and metabolomics profiles of human and murine-derived microglial and macrophage cell lines treated with ox-LDL. Under the same ox-LDL concentration, macrophages exhibited higher lipid uptake and expression of pro-inflammatory cytokines as compared to microglia. The ox-LDL increased the levels of fatty acid metabolites in macrophages and sphingomyelin metabolites in microglia. In summary, this study revealed the heterogeneity in the inflammatory capacity and metabolic profiles of macrophages and microglia under the stimulation of ox-LDL.
Collapse
Affiliation(s)
- Yaru Sun
- Joint Shantou International Eye Center of Shantou University and the Chinese University of Hong Kong, Shantou, Gaungdong, China
- Shantou University Medical College, Shantou, Guangdong, China
| | - Jia-Jian Liang
- Joint Shantou International Eye Center of Shantou University and the Chinese University of Hong Kong, Shantou, Gaungdong, China
| | - Jianming Xu
- Joint Shantou International Eye Center of Shantou University and the Chinese University of Hong Kong, Shantou, Gaungdong, China
- Shantou University Medical College, Shantou, Guangdong, China
| | - Kewen Zhou
- Joint Shantou International Eye Center of Shantou University and the Chinese University of Hong Kong, Shantou, Gaungdong, China
- Shantou University Medical College, Shantou, Guangdong, China
| | - Changzhen Fu
- Joint Shantou International Eye Center of Shantou University and the Chinese University of Hong Kong, Shantou, Gaungdong, China
| | - Shao-Lang Chen
- Joint Shantou International Eye Center of Shantou University and the Chinese University of Hong Kong, Shantou, Gaungdong, China
| | - Rucui Yang
- Joint Shantou International Eye Center of Shantou University and the Chinese University of Hong Kong, Shantou, Gaungdong, China
- Shantou University Medical College, Shantou, Guangdong, China
| | - Tsz Kin Ng
- Joint Shantou International Eye Center of Shantou University and the Chinese University of Hong Kong, Shantou, Gaungdong, China
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Qingping Liu
- Joint Shantou International Eye Center of Shantou University and the Chinese University of Hong Kong, Shantou, Gaungdong, China
| | - Mingzhi Zhang
- Joint Shantou International Eye Center of Shantou University and the Chinese University of Hong Kong, Shantou, Gaungdong, China
| |
Collapse
|
38
|
Jin Q, Liu T, Ma F, Fu T, Yang L, Mao H, Wang Y, Peng L, Li P, Zhan Y. Roles of Sirt1 and its modulators in diabetic microangiopathy: A review. Int J Biol Macromol 2024; 264:130761. [PMID: 38467213 DOI: 10.1016/j.ijbiomac.2024.130761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 02/03/2024] [Accepted: 03/07/2024] [Indexed: 03/13/2024]
Abstract
Diabetic vascular complications include diabetic macroangiopathy and diabetic microangiopathy. Diabetic microangiopathy is characterised by impaired microvascular endothelial function, basement membrane thickening, and microthrombosis, which may promote renal, ocular, cardiac, and peripheral system damage in diabetic patients. Therefore, new preventive and therapeutic strategies are urgently required. Sirt1, a member of the nicotinamide adenine dinucleotide-dependent histone deacetylase class III family, regulates different organ growth and development, oxidative stress, mitochondrial function, metabolism, inflammation, and aging. Sirt1 is downregulated in vascular injury and microangiopathy. Moreover, its expression and distribution in different organs correlate with age and play critical regulatory roles in oxidative stress and inflammation. This review introduces the background of diabetic microangiopathy and the main functions of Sirt1. Then, the relationship between Sirt1 and different diabetic microangiopathies and the regulatory roles mediated by different cells are described. Finally, we summarize the modulators that target Sirt1 to ameliorate diabetic microangiopathy as an essential preventive and therapeutic measure for diabetic microangiopathy. In conclusion, targeting Sirt1 may be a new therapeutic strategy for diabetic microangiopathy.
Collapse
Affiliation(s)
- Qi Jin
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Tongtong Liu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Fang Ma
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Tongfei Fu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Liping Yang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Huimin Mao
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuyang Wang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Liang Peng
- China-Japan Friendship Hospital, Institute of Clinical Medical Sciences, Beijing, China.
| | - Ping Li
- China-Japan Friendship Hospital, Institute of Clinical Medical Sciences, Beijing, China.
| | - Yongli Zhan
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
39
|
Grubaugh CR, Dhingra A, Prakash B, Montenegro D, Sparrow JR, Daniele LL, Curcio CA, Bell BA, Hussain MM, Boesze-Battaglia K. Microsomal triglyceride transfer protein is necessary to maintain lipid homeostasis and retinal function. FASEB J 2024; 38:e23522. [PMID: 38445789 PMCID: PMC10949407 DOI: 10.1096/fj.202302491r] [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: 12/02/2023] [Revised: 02/07/2024] [Accepted: 02/16/2024] [Indexed: 03/07/2024]
Abstract
Lipid processing by the retinal pigment epithelium (RPE) is necessary to maintain retinal health and function. Dysregulation of retinal lipid homeostasis due to normal aging or age-related disease triggers lipid accumulation within the RPE, on Bruch's membrane (BrM), and in the subretinal space. In its role as a hub for lipid trafficking into and out of the neural retina, the RPE packages a significant amount of lipid into lipid droplets for storage and into apolipoprotein B (APOB)-containing lipoproteins (Blps) for export. Microsomal triglyceride transfer protein (MTP), encoded by the MTTP gene, is essential for Blp assembly. Herein we test the hypothesis that MTP expression in the RPE is essential to maintain lipid balance and retinal function using the newly generated RPEΔMttp mouse model. Using non-invasive ocular imaging, electroretinography, and histochemical and biochemical analyses we show that genetic depletion of Mttp from the RPE results in intracellular lipid accumulation, increased photoreceptor-associated cholesterol deposits, and photoreceptor cell death, and loss of rod but not cone function. RPE-specific reduction in Mttp had no significant effect on plasma lipids and lipoproteins. While APOB was decreased in the RPE, most ocular retinoids remained unchanged, with the exception of the storage form of retinoid, retinyl ester. Thus suggesting that RPE MTP is critical for Blp synthesis and assembly but is not directly involved in plasma lipoprotein metabolism. These studies demonstrate that RPE-specific MTP expression is necessary to establish and maintain retinal lipid homeostasis and visual function.
Collapse
Affiliation(s)
- Catharina R. Grubaugh
- Department of Basic and Translational Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Anuradha Dhingra
- Department of Basic and Translational Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Binu Prakash
- Department of Foundations of Medicine, New York University Grossman Long Island School of Medicine, Mineola, NY, 11501 USA
| | - Diego Montenegro
- Department of Ophthalmology and Department of Pathology and Cell Biology, Columbia University, New York, NY, 10027 USA
| | - Janet R. Sparrow
- Department of Ophthalmology and Department of Pathology and Cell Biology, Columbia University, New York, NY, 10027 USA
| | - Lauren L. Daniele
- Department of Basic and Translational Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Christine A. Curcio
- Department of Ophthalmology and Visual Sciences, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Brent A. Bell
- Department of Ophthalmology, University of Pennsylvania, Philadelphia, PA, 19104 USA
| | - M. Mahmood Hussain
- Department of Foundations of Medicine, New York University Grossman Long Island School of Medicine, Mineola, NY, 11501 USA
| | - Kathleen Boesze-Battaglia
- Department of Basic and Translational Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
40
|
Shahror RA, Morris CA, Mohammed AA, Wild M, Zaman B, Mitchell CD, Phillips PH, Rusch NJ, Shosha E, Fouda AY. Role of myeloid cells in ischemic retinopathies: recent advances and unanswered questions. J Neuroinflammation 2024; 21:65. [PMID: 38454477 PMCID: PMC10918977 DOI: 10.1186/s12974-024-03058-y] [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: 12/05/2023] [Accepted: 02/28/2024] [Indexed: 03/09/2024] Open
Abstract
Myeloid cells including microglia and macrophages play crucial roles in retinal homeostasis by clearing cellular debris and regulating inflammation. These cells are activated in several blinding ischemic retinal diseases including diabetic retinopathy, where they may exert both beneficial and detrimental effects on neurovascular function and angiogenesis. Myeloid cells impact the progression of retinal pathologies and recent studies suggest that targeting myeloid cells is a promising therapeutic strategy to mitigate diabetic retinopathy and other ischemic retinal diseases. This review summarizes the recent advances in our understanding of the role of microglia and macrophages in retinal diseases and focuses on the effects of myeloid cells on neurovascular injury and angiogenesis in ischemic retinopathies. We highlight gaps in knowledge and advocate for a more detailed understanding of the role of myeloid cells in retinal ischemic injury to fully unlock the potential of targeting myeloid cells as a therapeutic strategy for retinal ischemia.
Collapse
Affiliation(s)
- Rami A Shahror
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences (UAMS), 4301 West Markham Street, Slot 611, BIOMED-1, B306, Little Rock, AR, 72205, USA
| | - Carol A Morris
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences (UAMS), 4301 West Markham Street, Slot 611, BIOMED-1, B306, Little Rock, AR, 72205, USA
| | - Aya A Mohammed
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences (UAMS), 4301 West Markham Street, Slot 611, BIOMED-1, B306, Little Rock, AR, 72205, USA
| | - Melissa Wild
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences (UAMS), 4301 West Markham Street, Slot 611, BIOMED-1, B306, Little Rock, AR, 72205, USA
| | - Bushra Zaman
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences (UAMS), 4301 West Markham Street, Slot 611, BIOMED-1, B306, Little Rock, AR, 72205, USA
| | - Christian D Mitchell
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences (UAMS), 4301 West Markham Street, Slot 611, BIOMED-1, B306, Little Rock, AR, 72205, USA
| | - Paul H Phillips
- Department of Ophthalmology, Harvey & Bernice Jones Eye Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Nancy J Rusch
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences (UAMS), 4301 West Markham Street, Slot 611, BIOMED-1, B306, Little Rock, AR, 72205, USA
| | - Esraa Shosha
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences (UAMS), 4301 West Markham Street, Slot 611, BIOMED-1, B306, Little Rock, AR, 72205, USA
- Clinical Pharmacy Department, Cairo University, Cairo, Egypt
| | - Abdelrahman Y Fouda
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences (UAMS), 4301 West Markham Street, Slot 611, BIOMED-1, B306, Little Rock, AR, 72205, USA.
- Clinical Pharmacy Department, Cairo University, Cairo, Egypt.
| |
Collapse
|
41
|
Yu C, Lad EM, Mathew R, Shiraki N, Littleton S, Chen Y, Hou J, Schlepckow K, Degan S, Chew L, Amason J, Kalnitsky J, Bowes Rickman C, Proia AD, Colonna M, Haass C, Saban DR. Microglia at sites of atrophy restrict the progression of retinal degeneration via galectin-3 and Trem2. J Exp Med 2024; 221:e20231011. [PMID: 38289348 PMCID: PMC10826045 DOI: 10.1084/jem.20231011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 12/11/2023] [Accepted: 01/12/2024] [Indexed: 02/01/2024] Open
Abstract
Outer retinal degenerations, including age-related macular degeneration (AMD), are characterized by photoreceptor and retinal pigment epithelium (RPE) atrophy. In these blinding diseases, macrophages accumulate at atrophic sites, but their ontogeny and niche specialization remain poorly understood, especially in humans. We uncovered a unique profile of microglia, marked by galectin-3 upregulation, at atrophic sites in mouse models of retinal degeneration and human AMD. In disease models, conditional deletion of galectin-3 in microglia led to phagocytosis defects and consequent augmented photoreceptor death, RPE damage, and vision loss, indicating protective roles. Mechanistically, Trem2 signaling orchestrated microglial migration to atrophic sites and induced galectin-3 expression. Moreover, pharmacologic Trem2 agonization led to heightened protection but in a galectin-3-dependent manner. In elderly human subjects, we identified this highly conserved microglial population that expressed galectin-3 and Trem2. This population was significantly enriched in the macular RPE-choroid of AMD subjects. Collectively, our findings reveal a neuroprotective population of microglia and a potential therapeutic target for mitigating retinal degeneration.
Collapse
Affiliation(s)
- Chen Yu
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, USA
| | - Eleonora M. Lad
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, USA
| | - Rose Mathew
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, USA
| | - Nobuhiko Shiraki
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, USA
| | - Sejiro Littleton
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, USA
- Department of Immunology, Duke University, Durham, NC, USA
| | - Yun Chen
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jinchao Hou
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Kai Schlepckow
- German Center for Neurodegenerative Diseases Munich, Munich, Germany
| | - Simone Degan
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, USA
| | - Lindsey Chew
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, USA
| | - Joshua Amason
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, USA
| | - Joan Kalnitsky
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, USA
| | - Catherine Bowes Rickman
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, USA
- Department of Cell Biology, Duke University, Durham, NC, USA
| | - Alan D. Proia
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, USA
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA
- Department of Pathology, Campbell University Jerry M. Wallace School of Osteopathic Medicine, Lillington, NC, USA
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Christian Haass
- German Center for Neurodegenerative Diseases Munich, Munich, Germany
- Chair of Metabolic Biochemistry, Faculty of Medicine, Biomedical Center, Ludwig-Maximilians-Universität München, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Daniel R. Saban
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, USA
- Department of Immunology, Duke University, Durham, NC, USA
| |
Collapse
|
42
|
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
|
43
|
Tsioti I, Steiner BL, Escher P, Zinkernagel MS, Benz PM, Kokona D. Systemic Lipopolysaccharide Exposure Exacerbates Choroidal Neovascularization in Mice. Ocul Immunol Inflamm 2024; 32:19-30. [PMID: 36441988 DOI: 10.1080/09273948.2022.2147547] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 11/08/2022] [Indexed: 11/29/2022]
Abstract
This study aims to investigate the effect of a systemic lipopolysaccharide (LPS) stimulus in the course of laser-induced choroidal neovascularization (CNV) in C57BL/6 J mice. A group of CNV-subjected mice received 1 mg/kg LPS via the tail vein immediately after CNV induction. Mouse eyes were monitored in vivo with fluorescein angiography for 2 weeks. In situ hybridization and flow cytometry were performed in the retina at different time points. LPS led to increased fluorescein leakage 3 days after CNV, correlated with a large influx of monocyte-derived macrophages and increase of pro-inflammatory microglia/macrophages in the retina. Additionally, LPS enhanced Vegfα mRNA expression by Glul-expressing cells but not Aif1 positive microglia/macrophages in the laser lesion. These findings suggest that systemic LPS exposure has transient detrimental effects in the course of CNV through activation of microglia/macrophages to a pro-inflammatory phenotype and supports the important role of these cells in the CNV course.
Collapse
Affiliation(s)
- Ioanna Tsioti
- Department of Ophthalmology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Beatrice L Steiner
- Department of Ophthalmology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Pascal Escher
- Department of Ophthalmology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Martin S Zinkernagel
- Department of Ophthalmology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Peter M Benz
- Department of CardioMetabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Despina Kokona
- Department of Ophthalmology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department of BioMedical Research, University of Bern, Bern, Switzerland
| |
Collapse
|
44
|
Grubaugh CR, Dhingra A, Prakash B, Montenegro D, Sparrow JR, Daniele LL, Curcio CA, Bell BA, Hussain MM, Boesze-Battaglia K. Microsomal triglyceride transfer protein is necessary to maintain lipid homeostasis and retinal function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.06.570418. [PMID: 38105975 PMCID: PMC10723417 DOI: 10.1101/2023.12.06.570418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Lipid processing by the retinal pigment epithelium (RPE) is necessary to maintain retinal health and function. Dysregulation of retinal lipid homeostasis due to normal aging or to age-related disease triggers lipid accumulation within the RPE, on Bruch's membrane (BrM), and in the subretinal space. In its role as a hub for lipid trafficking into and out of the neural retina, the RPE packages a significant amount of lipid into lipid droplets for storage and into apolipoprotein B (apoB)-containing lipoproteins (Blps) for export. Microsomal triglyceride transfer protein (MTP), encoded by the MTTP gene, is essential for Blp assembly. Herein we test the hypothesis that MTP expression in the RPE is essential to maintain lipid balance and retinal function using the newly generated RPEΔMttp mouse model. Using non-invasive ocular imaging, electroretinography, and histochemical and biochemical analyses we show that genetic deletion of Mttp from the RPE results in intracellular lipid accumulation, increased photoreceptor -associated cholesterol deposits and photoreceptor cell death, and loss of rod but not cone function. RPE-specific ablation of Mttp had no significant effect on plasma lipids and lipoproteins. While, apoB was decreased in the RPE, ocular retinoid concentrations remained unchanged. Thus suggesting that RPE MTP is critical for Blp synthesis and assembly but not directly involved in ocular retinoid and plasma lipoprotein metabolism. These studies demonstrate that RPE-specific MTP expression is necessary to establish and maintain retinal lipid homeostasis and visual function.
Collapse
Affiliation(s)
- Catharina R. Grubaugh
- Department of Basic and Translational Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Anuradha Dhingra
- Department of Basic and Translational Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Binu Prakash
- Department of Foundations of Medicine, New York University Grossman Long Island School of Medicine, Mineola, NY, 11501 USA
| | - Diego Montenegro
- Department of Ophthalmology and Department of Pathology and Cell Biology, Columbia University, New York, NY,10027 USA
| | - Janet R. Sparrow
- Department of Ophthalmology and Department of Pathology and Cell Biology, Columbia University, New York, NY,10027 USA
| | - Lauren L. Daniele
- Department of Basic and Translational Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Christine A. Curcio
- Department of Ophthalmology and Visual Sciences, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Brent A. Bell
- Department of Ophthalmology, University of Pennsylvania, Philadelphia, PA, 19104 USA
| | - M. Mahmood Hussain
- Department of Foundations of Medicine, New York University Grossman Long Island School of Medicine, Mineola, NY, 11501 USA
| | - Kathleen Boesze-Battaglia
- Department of Basic and Translational Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
45
|
Rajanala K, Dotiwala F, Upadhyay A. Geographic atrophy: pathophysiology and current therapeutic strategies. FRONTIERS IN OPHTHALMOLOGY 2023; 3:1327883. [PMID: 38983017 PMCID: PMC11182118 DOI: 10.3389/fopht.2023.1327883] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 11/22/2023] [Indexed: 07/11/2024]
Abstract
Geographic atrophy (GA) is an advanced stage of age-related macular degeneration (AMD) that leads to gradual and permanent vision loss. GA is characterized by the loss of photoreceptor cells and retinal pigment epithelium (RPE), leading to distinct atrophic patches in the macula, which tends to increase with time. Patients with geographic atrophy often experience a gradual and painless loss of central vision, resulting in difficulty reading, recognizing faces, or performing activities that require detailed vision. The primary risk factor for the development of geographic atrophy is advanced age; however, other risk factors, such as family history, smoking, and certain genetic variations, are also associated with AMD. Diagnosis is usually based on a comprehensive eye examination, including imaging tests such as fundus photography, optical coherence tomography (OCT), and fluorescein angiography. Numerous clinical trials are underway, targeting identified molecular pathways associated with GA that are promising. Recent approvals of Syfovre and Izervay by the FDA for the treatment of GA provide hope to affected patients. Administration of these drugs resulted in slowing the rate of progression of the disease. Though these products provide treatment benefits to the patients, they do not offer a cure for geographic atrophy and are limited in efficacy. Considering these safety concerns and limited treatment benefits, there is still a significant need for therapeutics with improved efficacy, safety profiles, and better patient compliance. This comprehensive review discusses pathophysiology, currently approved products, their limitations, and potential future treatment strategies for GA.
Collapse
Affiliation(s)
| | | | - Arun Upadhyay
- Research and Development, Ocugen Inc., Malvern, PA, United States
| |
Collapse
|
46
|
Karg MM, Moorefield M, Hoffmann E, Philipose H, Krasniqi D, Hoppe C, Shu DY, Shirahama S, Ksander BR, Saint-Geniez M. Microglia preserve visual function loss in the aging retina by supporting retinal pigment epithelial health. Immun Ageing 2023; 20:53. [PMID: 37838654 PMCID: PMC10576380 DOI: 10.1186/s12979-023-00358-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 06/23/2023] [Indexed: 10/16/2023]
Abstract
BACKGROUND Increased age is a risk factor for the development and progression of retinal diseases including age-related macular degeneration (AMD). Understanding the changes that occur in the eye due to aging is important in enhancing our understanding of AMD pathogenesis and the development of novel AMD therapies. Microglia, the resident brain and retinal immune cells are associated with both maintaining homeostasis and protection of neurons and loss of microglia homeostasis could be a significant player in age related neurodegeneration. One important characteristic of retinal aging is the migration of microglia from the inner to outer retina where they reside in the subretinal space (SRS) in contact with the retinal pigment epithelial (RPE) cells. The role of aged subretinal microglia is unknown. Here, we depleted microglia in aged C57/BL6 mice fed for 6 weeks with a chow containing PLX5622, a small molecule inhibitor of colony-stimulating factor-1 receptor (Csf1r) required for microglial survival. RESULTS The subretinal P2RY12 + microglia in aged mice displayed a highly amoeboid and activated morphology and were filled with autofluorescence droplets reminiscent of lipofuscin. TEM indicates that subretinal microglia actively phagocytize shed photoreceptor outer segments, one of the main functions of retinal pigmented epithelial cells. PLX5622 treatment depleted up to 90% of the retinal microglia and was associated with significant loss in visual function. Mice on the microglia depletion diet showed reduced contrast sensitivity and significantly lower electroretinogram for the c-wave, a measurement of RPE functionality, compared to age-matched controls. The loss of c-wave coincided with a loss of RPE cells and increased RPE swelling in the absence of microglia. CONCLUSIONS We conclude that microglia preserve visual function in aged mice and support RPE cell function, by phagocytosing shed photoreceptor outer segments and lipids, therefore compensating for the known age-related decline of RPE phagocytosis.
Collapse
Affiliation(s)
- Margarete M Karg
- Schepens Eye Research Institute of Mass Eye and Ear, 20 Staniford St, Boston, MA, 02114, USA
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - May Moorefield
- Schepens Eye Research Institute of Mass Eye and Ear, 20 Staniford St, Boston, MA, 02114, USA
| | - Emma Hoffmann
- Schepens Eye Research Institute of Mass Eye and Ear, 20 Staniford St, Boston, MA, 02114, USA
| | - Hannah Philipose
- Schepens Eye Research Institute of Mass Eye and Ear, 20 Staniford St, Boston, MA, 02114, USA
| | - Drenushe Krasniqi
- Schepens Eye Research Institute of Mass Eye and Ear, 20 Staniford St, Boston, MA, 02114, USA
| | - Cindy Hoppe
- Schepens Eye Research Institute of Mass Eye and Ear, 20 Staniford St, Boston, MA, 02114, USA
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Daisy Y Shu
- Schepens Eye Research Institute of Mass Eye and Ear, 20 Staniford St, Boston, MA, 02114, USA
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Shintaro Shirahama
- Schepens Eye Research Institute of Mass Eye and Ear, 20 Staniford St, Boston, MA, 02114, USA
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Bruce R Ksander
- Schepens Eye Research Institute of Mass Eye and Ear, 20 Staniford St, Boston, MA, 02114, USA.
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA.
| | - Magali Saint-Geniez
- Schepens Eye Research Institute of Mass Eye and Ear, 20 Staniford St, Boston, MA, 02114, USA
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
47
|
Lieffrig SA, Gyimesi G, Mao Y, Finnemann SC. Clearance phagocytosis by the retinal pigment epithelial during photoreceptor outer segment renewal: Molecular mechanisms and relation to retinal inflammation. Immunol Rev 2023; 319:81-99. [PMID: 37555340 PMCID: PMC10615845 DOI: 10.1111/imr.13264] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 07/18/2023] [Indexed: 08/10/2023]
Abstract
Mammalian photoreceptor outer segment renewal is a highly coordinated process that hinges on timed cell signaling between photoreceptor neurons and the adjacent retinal pigment epithelial (RPE). It is a strictly rhythmic, synchronized process that underlies in part circadian regulation. We highlight findings from recently developed methods that quantify distinct phases of outer segment renewal in retinal tissue. At light onset, outer segments expose the conserved "eat-me" signal phosphatidylserine exclusively at their distal, most aged tip. A coordinated two-receptor efferocytosis process follows, in which ligands bridge outer segment phosphatidylserine with the RPE receptors αvβ5 integrin, inducing cytosolic signaling toward Rac1 and focal adhesion kinase/MERTK, and with MERTK directly, additionally inhibiting RhoA/ROCK and thus enabling F-actin dynamics favoring outer segment fragment engulfment. Photoreceptors and RPE persist for life with each RPE cell in the eye servicing dozens of overlying photoreceptors. Thus, RPE cells phagocytose more often and process more material than any other cell type. Mutant mice with impaired outer segment renewal largely retain functional photoreceptors and retinal integrity. However, when anti-inflammatory signaling in the RPE via MERTK or the related TYRO3 is lacking, catastrophic inflammation leads to immune cell infiltration that swiftly destroys the retina causing blindness.
Collapse
Affiliation(s)
- Stephanie A. Lieffrig
- Center for Cancer, Genetic Diseases and Gene Regulation, Department of Biological Sciences, Fordham University, Bronx, NY
| | - Gavin Gyimesi
- Center for Cancer, Genetic Diseases and Gene Regulation, Department of Biological Sciences, Fordham University, Bronx, NY
| | | | - Silvia C. Finnemann
- Center for Cancer, Genetic Diseases and Gene Regulation, Department of Biological Sciences, Fordham University, Bronx, NY
| |
Collapse
|
48
|
Borchert GA, Shamsnajafabadi H, Hu ML, De Silva SR, Downes SM, MacLaren RE, Xue K, Cehajic-Kapetanovic J. The Role of Inflammation in Age-Related Macular Degeneration-Therapeutic Landscapes in Geographic Atrophy. Cells 2023; 12:2092. [PMID: 37626902 PMCID: PMC10453093 DOI: 10.3390/cells12162092] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 07/28/2023] [Accepted: 08/11/2023] [Indexed: 08/27/2023] Open
Abstract
Age-related macular degeneration (AMD) is the leading cause of vision loss and visual impairment in people over 50 years of age. In the current therapeutic landscape, intravitreal anti-vascular endothelial growth factor (anti-VEGF) therapies have been central to the management of neovascular AMD (also known as wet AMD), whereas treatments for geographic atrophy have lagged behind. Several therapeutic approaches are being developed for geographic atrophy with the goal of either slowing down disease progression or reversing sight loss. Such strategies target the inflammatory pathways, complement cascade, visual cycle or neuroprotective mechanisms to slow down the degeneration. In addition, retinal implants have been tried for vision restoration and stem cell therapies for potentially a dual purpose of slowing down the degeneration and restoring visual function. In particular, therapies focusing on the complement pathway have shown promising results with the FDA approved pegcetacoplan, a complement C3 inhibitor, and avacincaptad pegol, a complement C5 inhibitor. In this review, we discuss the mechanisms of inflammation in AMD and outline the therapeutic landscapes of atrophy AMD. Improved understanding of the various pathway components and their interplay in this complex neuroinflammatory degeneration will guide the development of current and future therapeutic options, such as optogenetic therapy.
Collapse
Affiliation(s)
- Grace A. Borchert
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, Oxford University, Oxford OX3 9DU, UK
| | - Hoda Shamsnajafabadi
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, Oxford University, Oxford OX3 9DU, UK
| | - Monica L. Hu
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, Oxford University, Oxford OX3 9DU, UK
| | - Samantha R. De Silva
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, Oxford University, Oxford OX3 9DU, UK
- Oxford Eye Hospital, Oxford University NHS Foundation Trust, Oxford OX3 9DU, UK
| | - Susan M. Downes
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, Oxford University, Oxford OX3 9DU, UK
- Oxford Eye Hospital, Oxford University NHS Foundation Trust, Oxford OX3 9DU, UK
| | - Robert E. MacLaren
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, Oxford University, Oxford OX3 9DU, UK
- Oxford Eye Hospital, Oxford University NHS Foundation Trust, Oxford OX3 9DU, UK
| | - Kanmin Xue
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, Oxford University, Oxford OX3 9DU, UK
- Oxford Eye Hospital, Oxford University NHS Foundation Trust, Oxford OX3 9DU, UK
| | - Jasmina Cehajic-Kapetanovic
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, Oxford University, Oxford OX3 9DU, UK
- Oxford Eye Hospital, Oxford University NHS Foundation Trust, Oxford OX3 9DU, UK
| |
Collapse
|
49
|
Yu C, Lad EM, Mathew R, Littleton S, Chen Y, Schlepckow K, Degan S, Chew L, Amason J, Kalnitsky J, Rickman CB, Proia AD, Colonna M, Haass C, Saban DR. Microglia at Sites of Atrophy Restrict the Progression of Retinal Degeneration via Galectin-3 and Trem2 Interactions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.19.549403. [PMID: 37502831 PMCID: PMC10370087 DOI: 10.1101/2023.07.19.549403] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Degenerative diseases of the outer retina, including age-related macular degeneration (AMD), are characterized by atrophy of photoreceptors and retinal pigment epithelium (RPE). In these blinding diseases, macrophages are known to accumulate ectopically at sites of atrophy, but their ontogeny and functional specialization within this atrophic niche remain poorly understood, especially in the human context. Here, we uncovered a transcriptionally unique profile of microglia, marked by galectin-3 upregulation, at atrophic sites in mouse models of retinal degeneration and in human AMD. Using disease models, we found that conditional deletion of galectin-3 in microglia led to defects in phagocytosis and consequent augmented photoreceptor death, RPE damage and vision loss, suggestive of a protective role. Mechanistically, Trem2 signaling orchestrated the migration of microglial cells to sites of atrophy, and there, induced galectin-3 expression. Moreover, pharmacologic Trem2 agonization led to heightened protection, but only in a galectin-3-dependent manner, further signifying the functional interdependence of these two molecules. Likewise in elderly human subjects, we identified a highly conserved population of microglia at the transcriptomic, protein and spatial levels, and this population was enriched in the macular region of postmortem AMD subjects. Collectively, our findings reveal an atrophy-associated specialization of microglia that restricts the progression of retinal degeneration in mice and further suggest that these protective microglia are conserved in AMD.
Collapse
Affiliation(s)
- Chen Yu
- Department of Ophthalmology, Duke University School of Medicine; Durham, NC 27710, USA
| | - Eleonora M Lad
- Department of Ophthalmology, Duke University School of Medicine; Durham, NC 27710, USA
| | - Rose Mathew
- Department of Ophthalmology, Duke University School of Medicine; Durham, NC 27710, USA
| | - Sejiro Littleton
- Department of Ophthalmology, Duke University School of Medicine; Durham, NC 27710, USA
- Department of Immunology, Duke University; Durham, NC 27710, USA
| | - Yun Chen
- Department of Pathology and Immunology, Washington University School of Medicine; St. Louis, MO 63110, USA
- Department of Neurology, Washington University School of Medicine; St. Louis, MO 63110, USA
| | - Kai Schlepckow
- German Center for Neurodegenerative Diseases (DZNE) Munich; 81377 Munich, Germany
| | - Simone Degan
- Department of Ophthalmology, Duke University School of Medicine; Durham, NC 27710, USA
| | - Lindsey Chew
- Department of Ophthalmology, Duke University School of Medicine; Durham, NC 27710, USA
| | - Joshua Amason
- Department of Ophthalmology, Duke University School of Medicine; Durham, NC 27710, USA
| | - Joan Kalnitsky
- Department of Ophthalmology, Duke University School of Medicine; Durham, NC 27710, USA
| | - Catherine Bowes Rickman
- Department of Ophthalmology, Duke University School of Medicine; Durham, NC 27710, USA
- Department of Cell Biology, Duke University; Durham, NC 27710, USA
| | - Alan D Proia
- Department of Ophthalmology, Duke University School of Medicine; Durham, NC 27710, USA
- Department of Pathology, Duke University School of Medicine; Durham, NC 27710, USA
- Department of Pathology, Campbell University Jerry M. Wallace School of Osteopathic Medicine, Lillington, NC 27546, USA
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine; St. Louis, MO 63110, USA
| | - Christian Haass
- German Center for Neurodegenerative Diseases (DZNE) Munich; 81377 Munich, Germany
- Chair of Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München; 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy); 81377 Munich, Germany
| | - Daniel R Saban
- Department of Ophthalmology, Duke University School of Medicine; Durham, NC 27710, USA
- Department of Immunology, Duke University; Durham, NC 27710, USA
| |
Collapse
|
50
|
Theune WC, Frost MP, Trakhtenberg EF. Transcriptomic profiling of retinal cells reveals a subpopulation of microglia/macrophages expressing Rbpms marker of retinal ganglion cells (RGCs) that confound identification of RGCs. Brain Res 2023; 1811:148377. [PMID: 37121423 PMCID: PMC10246437 DOI: 10.1016/j.brainres.2023.148377] [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: 03/03/2023] [Revised: 04/09/2023] [Accepted: 04/25/2023] [Indexed: 05/02/2023]
Abstract
Analysis of retinal ganglion cells (RGCs) by scRNA-seq is emerging as a state-of-the-art method for studying RGC biology and subtypes, as well as for studying the mechanisms of neuroprotection and axon regeneration in the central nervous system (CNS). Rbpms has been established as a pan-RGC marker, and Spp1 has been established as an αRGC type and macrophage marker. Here, we analyzed by scRNA-seq retinal microglia and macrophages, and found Rbpms+ subpopulations of retinal microglia/macrophages, which pose a potential pitfall in scRNA-seq studies involving RGCs. We performed comparative analysis of cellular identity of the presumed RGC cells isolated in recent scRNA-seq studies, and found that Rbpms+ microglia/macrophages confounded identification of RGCs. We also showed using immunohistological analysis that, Rbpms protein localizes to stress granules in a subpopulation of retinal microglia after optic nerve injury, which was further supported by bioinformatics analysis identifying stress granule-associated genes enriched in the Rbpms+ microglia/macrophages. Our findings suggest that the identification of Rbpms+ RGCs by immunostaining after optic nerve injury should exclude cells in which Rbpms signal is restricted to a subcellular granule, and include only those cells in which the Rbpms signal is labeling cell soma diffusely. Finally, we provide solutions for circumventing this potential pitfall of Rbpms-expressing microglia/macrophages in scRNA-seq studies, by including in RGC and αRGC selection criteria other pan-RGC and αRGC markers.
Collapse
Affiliation(s)
- William C Theune
- Department of Neuroscience, University of Connecticut School of Medicine, 263 Farmington Ave., Farmington, CT 06030, USA
| | - Matthew P Frost
- Department of Neuroscience, University of Connecticut School of Medicine, 263 Farmington Ave., Farmington, CT 06030, USA
| | - Ephraim F Trakhtenberg
- Department of Neuroscience, University of Connecticut School of Medicine, 263 Farmington Ave., Farmington, CT 06030, USA.
| |
Collapse
|