1
|
Dempsey MP, Conrady CD. The Host-Pathogen Interplay: A Tale of Two Stories within the Cornea and Posterior Segment. Microorganisms 2023; 11:2074. [PMID: 37630634 PMCID: PMC10460047 DOI: 10.3390/microorganisms11082074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 08/10/2023] [Accepted: 08/10/2023] [Indexed: 08/27/2023] Open
Abstract
Ocular infectious diseases are an important cause of potentially preventable vision loss and blindness. In the following manuscript, we will review ocular immunology and the pathogenesis of herpesviruses and Pseudomonas aeruginosa infections of the cornea and posterior segment. We will highlight areas of future research and what is currently known to promote bench-to-bedside discoveries to improve clinical outcomes of these debilitating ocular diseases.
Collapse
Affiliation(s)
- Michael P. Dempsey
- Department of Ophthalmology and Visual Sciences, Truhlsen Eye Center, University of Nebraska Medical Center, Omaha, NE 68105, USA
| | - Christopher D. Conrady
- Department of Ophthalmology and Visual Sciences, Truhlsen Eye Center, University of Nebraska Medical Center, Omaha, NE 68105, USA
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
2
|
Du Y, Yan B. Ocular immune privilege and retinal pigment epithelial cells. J Leukoc Biol 2023; 113:288-304. [PMID: 36805720 DOI: 10.1093/jleuko/qiac016] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Indexed: 02/04/2023] Open
Abstract
The ocular tissue microenvironment is immune-privileged and uses multiple immunosuppressive mechanisms to prevent the induction of inflammation. The retinal pigment epithelium plays an essential role in ocular immune privilege. In addition to serving as a blood barrier separating the fenestrated choriocapillaris from the retina, the retinal pigment epithelium is a source of immunosuppressive cytokines and membrane-bound negative regulators that modulate the activity of immune cells within the retina. This article reviews the current understanding of how retinal pigment epithelium cells mediate immune regulation, focusing on the changes under pathologic conditions.
Collapse
Affiliation(s)
- Yuxiang Du
- Institute of Precision Medicine, Jining Medical University, No. 133, Hehua Road, Taibaihu New District, Jining, Shandong 272067, People's Republic of China
| | - Bo Yan
- Institute of Precision Medicine, Jining Medical University, No. 133, Hehua Road, Taibaihu New District, Jining, Shandong 272067, People's Republic of China
| |
Collapse
|
3
|
Huang Z, Li W, Su W. Tregs in Autoimmune Uveitis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1278:205-227. [PMID: 33523450 DOI: 10.1007/978-981-15-6407-9_11] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Uveitis is a chronic disease with relapsing and remitting ocular attack, which requires corticosteroids and systemic immunosuppression to prevent severe vision loss. Classically, uveitis is referred to an autoimmune disease, mediated by pro-inflammatory Th17 cells and immunosuppressive CD4+CD25+FoxP3+ T-regulatory cells (Tregs). More and more evidence indicates that Tregs are involved in development, resolution, and remission of uveitis. Clinically, many researchers have conducted quantitative and functional analyses of peripheral blood from patients with different subtypes of uveitis, in an attempt to find the changing rules of Tregs. Consistently, using the experimental autoimmune uveitis (EAU) model, researchers have explored the development and resolution mechanism of uveitis in many aspects. In addition, many drug and Tregs therapy investigations have yielded encouraging results. In this chapter, we introduced the current understanding of Tregs, summarized the clinical changes in the number and function of patients with uveitis and the immune mechanism of Tregs involved in EAU model, as well as discussed the progress and shortcomings of Tregs-related drug therapy and Tregs therapy. Although the exact mechanism of Tregs-mediated uveitis protection remains to be elucidated, the strategy of Tregs regulation may provide a specific and meaningful way for the prevention and treatment of uveitis.
Collapse
Affiliation(s)
- Zhaohao Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Wenli Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Wenru Su
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
4
|
Song J, Han D, Lee H, Kim DJ, Cho JY, Park JH, Seok SH. A Comprehensive Proteomic and Phosphoproteomic Analysis of Retinal Pigment Epithelium Reveals Multiple Pathway Alterations in Response to the Inflammatory Stimuli. Int J Mol Sci 2020; 21:ijms21093037. [PMID: 32344885 PMCID: PMC7246457 DOI: 10.3390/ijms21093037] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 04/21/2020] [Accepted: 04/23/2020] [Indexed: 12/31/2022] Open
Abstract
Overwhelming and persistent inflammation of retinal pigment epithelium (RPE) induces destructive changes in the retinal environment. However, the precise mechanisms remain unclear. In this study, we aimed to investigate RPE-specific biological and metabolic responses against intense inflammation and identify the molecular characteristics determining pathological progression. We performed quantitative analyses of the proteome and phosphoproteome of the human-derived RPE cell line ARPE-19 after treatment with lipopolysaccharide (LPS) for 45 min or 24 h using the latest isobaric tandem-mass tags (TMTs) labeling approach. This approach led to the identification of 8984 proteins, of which 261 showed a 1.5-fold change in abundance after 24 h of treatment with LPS. A parallel phosphoproteome analysis identified 20,632 unique phosphopeptides from 3207 phosphoproteins with 3103 phosphorylation sites. Integrated proteomic and phosphoproteomic analyses showed significant downregulation of proteins related to mitochondrial respiration and cell cycle checkpoint, while proteins related to lipid metabolism, amino acid metabolism, cell-matrix adhesion, and endoplasmic reticulum (ER) stress were upregulated after LPS stimulation. Further, phosphorylation events in multiple pathways, including MAPKK and Wnt/β-catenin signalings, were identified as involved in LPS-triggered pathobiology. In essence, our findings reveal multiple integrated signals exerted by RPE under inflammation and are expected to give insight into the development of therapeutic interventions for RPE disorders.
Collapse
Affiliation(s)
- Juha Song
- Department of Microbiology and Immunology, and Institute of Endemic Disease, Seoul National University College of Medicine, Chongno-gu, Seoul 03080, Korea;
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Seoul National University, Gwanak-gu, Seoul 08826, Korea
| | - Dohyun Han
- Proteomics Core Facility, Biomedical Research Institute, Seoul National University Hospital, Seoul 03080, Korea; (D.H.); (H.L.)
| | - Heonyi Lee
- Proteomics Core Facility, Biomedical Research Institute, Seoul National University Hospital, Seoul 03080, Korea; (D.H.); (H.L.)
| | - Da Jung Kim
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul 03080, Korea; (D.J.K.); (J.-Y.C.)
- Department of Biomedical Sciences, Seoul National University College of Medicine, Chongno-gu, Seoul 03080, Korea
| | - Joo-Youn Cho
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul 03080, Korea; (D.J.K.); (J.-Y.C.)
- Department of Biomedical Sciences, Seoul National University College of Medicine, Chongno-gu, Seoul 03080, Korea
| | - Jae-Hak Park
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Seoul National University, Gwanak-gu, Seoul 08826, Korea
- Correspondence: (J.-H.P.); (S.H.S.); Tel.: +82-2-880-1256 (J.-H.P.); +82-2-740-8302 (S.H.S.); Fax: +82-2-763-5206 (S.H.S.)
| | - Seung Hyeok Seok
- Department of Microbiology and Immunology, and Institute of Endemic Disease, Seoul National University College of Medicine, Chongno-gu, Seoul 03080, Korea;
- Department of Biomedical Sciences, Seoul National University College of Medicine, Chongno-gu, Seoul 03080, Korea
- Correspondence: (J.-H.P.); (S.H.S.); Tel.: +82-2-880-1256 (J.-H.P.); +82-2-740-8302 (S.H.S.); Fax: +82-2-763-5206 (S.H.S.)
| |
Collapse
|
5
|
Subhi Y, Krogh Nielsen M, Molbech CR, Oishi A, Singh A, Nissen MH, Sørensen TL. Polypoidal Choroidal Vasculopathy Associate With Diminished Regulatory T Cells That Are Polarized Into a T Helper 2-Like Phenotype. Invest Ophthalmol Vis Sci 2019; 60:2583-2590. [PMID: 31219532 DOI: 10.1167/iovs.19-26882] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose To investigate possible roles of T helper (Th) cells, regulatory T cells (Tregs), and the recently mapped Th-like Tregs in patients with polypoidal choroidal vasculopathy (PCV). Methods In this prospective case-control study, we obtained fresh venous blood from patients with PCV (n = 24), age-matched healthy controls (n = 32), and patients with neovascular AMD (n = 45). All participants underwent a comprehensive ocular examination including fluorescein and indocyanine green angiography for where retinal disease was suspected. Using flow cytometry, we identified Th subsets, Tregs, and Th-like Tregs. Plasma samples were stored at -80°C to investigate plasma cytokines of interest. Results Compared to healthy controls, patients with PCV had lower percentages of Tregs (8.7% ± 2.8% vs. 7.3% ± 1.7%, P = 0.027), which were significantly more Th2-like polarized (42.6% ± 13.3% vs. 50.5% ± 13.0%, P = 0.029). These changes differed from that observed in neovascular AMD, which compared to healthy controls had fewer Th1/Th17 cells (3.6% ± 2.7% vs. 2.4% ± 2.5%, P = 0.049), comparable Treg levels, and no distinct polarization of Th-like Tregs. Because of these findings, we measured plasma IL-4 and IL-33 levels. Plasma IL-33 in patients with PCV (median 0.30 pg/mL) was twice as high compared to healthy controls (median 0.16 pg/mL; P = 0.037). Conclusions PCV associate with diminished Tregs that are polarized more into a Th2-like phenotype. This is correlated to IL-33 levels, which we also find increased in patients with PCV. Our findings suggest a possible role for Th2-like Tregs and IL-33 in PCV.
Collapse
Affiliation(s)
- Yousif Subhi
- Clinical Eye Research Division, Department of Ophthalmology, Zealand University Hospital, Roskilde, Denmark.,Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark
| | - Marie Krogh Nielsen
- Clinical Eye Research Division, Department of Ophthalmology, Zealand University Hospital, Roskilde, Denmark.,Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark
| | - Christopher Rue Molbech
- Clinical Eye Research Division, Department of Ophthalmology, Zealand University Hospital, Roskilde, Denmark.,Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark
| | - Akio Oishi
- Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Amardeep Singh
- Clinical Eye Research Division, Department of Ophthalmology, Zealand University Hospital, Roskilde, Denmark.,Department of Ophthalmology, Skåne University Hospital Malmö-Lund, Lund, Sweden
| | - Mogens Holst Nissen
- Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark.,Eye Research Unit, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Torben Lykke Sørensen
- Clinical Eye Research Division, Department of Ophthalmology, Zealand University Hospital, Roskilde, Denmark.,Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
6
|
Ben M’Barek K, Habeler W, Regent F, Monville C. Developing Cell-Based Therapies for RPE-Associated Degenerative Eye Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1186:55-97. [DOI: 10.1007/978-3-030-28471-8_3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
7
|
Chen M, Luo C, Zhao J, Devarajan G, Xu H. Immune regulation in the aging retina. Prog Retin Eye Res 2018; 69:159-172. [PMID: 30352305 DOI: 10.1016/j.preteyeres.2018.10.003] [Citation(s) in RCA: 157] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 10/13/2018] [Accepted: 10/19/2018] [Indexed: 12/24/2022]
Abstract
The retina is an immune privileged tissue, which is protected from external and internal insults by its blood-retina barriers and immune suppressive microenvironment. Apart from the avoidance and tolerance strategies, the retina is also protected by its own defense system, i.e., microglia and the complement system. The immune privilege and defense mechanisms work together to maintain retinal homeostasis. During aging, the retina is at an increased risk of developing various degenerative diseases such as age-related macular degeneration, diabetic retinopathy, and glaucomatous retinopathy. Previously, we have shown that aging induces a para-inflammatory response in the retina. In this review, we explore the impact of aging on retinal immune regulation and the connection between homeostatic control of retinal immune privilege and para-inflammation under aging conditions and present a view that may explain why aging puts the retina at risk of developing degenerative diseases.
Collapse
Affiliation(s)
- Mei Chen
- Centre for Experimental Medicine, School of Medicine, Dentistry & Biomedical Sciences, Queen's University Belfast, UK
| | - Chang Luo
- Centre for Experimental Medicine, School of Medicine, Dentistry & Biomedical Sciences, Queen's University Belfast, UK; Aier Eye Institute, Aier School of Ophthalmology, Central South University, China
| | - Jiawu Zhao
- Centre for Experimental Medicine, School of Medicine, Dentistry & Biomedical Sciences, Queen's University Belfast, UK
| | | | - Heping Xu
- Centre for Experimental Medicine, School of Medicine, Dentistry & Biomedical Sciences, Queen's University Belfast, UK; Aier Eye Institute, Aier School of Ophthalmology, Central South University, China.
| |
Collapse
|
8
|
Ramirez MU, Stirling ER, Emenaker NJ, Roberts DD, Soto-Pantoja DR. Thrombospondin-1 interactions regulate eicosanoid metabolism and signaling in cancer-related inflammation. Cancer Metastasis Rev 2018; 37:469-476. [PMID: 29909440 PMCID: PMC6295347 DOI: 10.1007/s10555-018-9737-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The metabolism of arachidonic acid and other polyunsaturated fatty acids produces eicosanoids, a family of biologically active lipids that are implicated in homeostasis and in several pathologies that involve inflammation. Inflammatory processes mediated by eicosanoids promote carcinogenesis by exerting direct effects on cancer cells and by affecting the tumor microenvironment. Therefore, understanding how eicosanoids mediate cancer progression may lead to better approaches and chemopreventive strategies for the treatment of cancer. The matricellular protein thrombospondin-1 is involved in processes that profoundly regulate inflammatory pathways that contribute to carcinogenesis and metastatic spread. This review focuses on interactions of thrombospondin-1 and eicosanoids in the microenvironment that promote carcinogenesis and how the microenvironment can be targeted for cancer prevention to increase curative responses of cancer patients.
Collapse
Affiliation(s)
- Manuel U Ramirez
- Department of Surgery, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
- Department of Physiology & Pharmacology, Wake Forest University Health Sciences, Bethesda, MD, 20892, USA
| | - Elizabeth R Stirling
- Department of Surgery, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
- Department of Cancer Biology, Wake Forest University Health Sciences, Bethesda, MD, 20892, USA
| | - Nancy J Emenaker
- Nutritional Science Research Group, Division of Cancer Prevention, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - David D Roberts
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - David R Soto-Pantoja
- Department of Surgery, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA.
- Department of Physiology & Pharmacology, Wake Forest University Health Sciences, Bethesda, MD, 20892, USA.
- Department of Cancer Biology, Wake Forest University Health Sciences, Bethesda, MD, 20892, USA.
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA.
- Comprehensive Cancer Center Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA.
| |
Collapse
|
9
|
Luo C, Zhao J, Chen M, Xu H. The expression of C1 inhibitor (C1INH) in macrophages is upregulated by retinal pigment epithelial cells - implication in subretinal immune privilege in the aging eye. Aging (Albany NY) 2018; 10:1380-1389. [PMID: 29905533 PMCID: PMC6046230 DOI: 10.18632/aging.101474] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Accepted: 06/07/2018] [Indexed: 04/18/2023]
Abstract
Age-related para-inflammation in the retina-choroidal interface is featured by low-levels of complement activation and subretinal macrophage accumulation. This study aimed to understand how complement expression in macrophages is regulated by retinal pigment epithelium (RPE). Bone marrow-derived macrophages (BMDMs) and RPE cells were cultured from 8-10 weeks old C57BL/6J mice. The BMDMs were co-cultured with normal RPE, or oxidized photoreceptor outer segment (oxPOS) or TNF-α pre-treated RPE, or apoptotic RPE, or RPE-choroid eyecups. Macrophages were then isolated and processed for real-time RT-PCR. The expression of complement inhibitor C1INH in BMDMs was significantly upregulated by RPE and RPE-choroid eyecups. The eyecups also upregulated CFH, CD59a, and Crry in BMDMs. oxPOS pre-treated RPE upregulated C1qb but down-regulated C3 expression in BMDMs. TNF-α pre-treated RPE enhanced C1INH and CFB expression. When BMDMs were treated with apoptotic RPE, the expression of C1qb, CFH, and CD59a was reduced, whereas the expression of C3, CFB and C1INH was increased. Our results suggest that RPE can modulate macrophages complement expression at the retina-choroidal interface even under aging or oxidative conditions. However, during inflammation, they may promote the alternative pathway of complement activation through down-regulating CFH and CD59a and upregulating CFB and C3.
Collapse
Affiliation(s)
- Chang Luo
- Centre for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen’s University Belfast, Belfast, UK
- AIER Eye Institute, Changsha, China
- AIER School of Ophthalmology, Central South University, Changsha, China
| | - Jiawu Zhao
- Centre for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen’s University Belfast, Belfast, UK
| | - Mei Chen
- Centre for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen’s University Belfast, Belfast, UK
| | - Heping Xu
- Centre for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen’s University Belfast, Belfast, UK
- AIER Eye Institute, Changsha, China
- AIER School of Ophthalmology, Central South University, Changsha, China
| |
Collapse
|
10
|
Immune Privilege and Eye-Derived T-Regulatory Cells. J Immunol Res 2018; 2018:1679197. [PMID: 29888291 PMCID: PMC5985108 DOI: 10.1155/2018/1679197] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 04/18/2018] [Indexed: 02/08/2023] Open
Abstract
Certain cellular components of the eye, such as neural retina, are unable to regenerate and replicate after destructive inflammation. Ocular immune privilege provides the eye with immune protection against intraocular inflammation in order to minimize the risk to vision integrity. The eye and immune system use strategies to maintain the ocular immune privilege by regulating the innate and adaptive immune response, which includes immunological ignorance, peripheral tolerance to eye-derived antigens, and intraocular immunosuppressive microenvironment. In this review, we summarize current knowledge regarding the molecular mechanism responsible for the development and maintenance of ocular immune privilege via regulatory T cells (Tregs), which are generated by the anterior chamber-associated immune deviation (ACAID), and ocular resident cells including corneal endothelial (CE) cells, ocular pigment epithelial (PE) cells, and aqueous humor. Furthermore, we examined the therapeutic potential of Tregs generated by RPE cells that express transforming growth factor beta (TGF-β), cytotoxic T lymphocyte-associated antigen-2 alpha (CTLA-2α), and retinoic acid for autoimmune uveoretinitis and evaluated a new strategy using human RPE-induced Tregs for clinical application in inflammatory ocular disease. We believe that a better understanding of the ocular immune privilege associated with Tregs might offer a new approach with regard to therapeutic interventions for ocular autoimmunity.
Collapse
|
11
|
Diabetic retinopathy and dysregulated innate immunity. Vision Res 2017; 139:39-46. [PMID: 28571700 DOI: 10.1016/j.visres.2017.04.013] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 04/12/2017] [Accepted: 04/13/2017] [Indexed: 02/07/2023]
Abstract
Diabetic retinopathy (DR) is the progressive degeneration of retinal blood vessels and neurons. Inflammation is known to play an important role in the pathogenesis of DR. During diabetes, metabolic disorder leads to the release of damage-associated molecular patterns (DAMPs) both in the retina and elsewhere in the body. The innate immune system provides the first line of defense against the DAMPs. In the early stages of DR when the blood retinal barrier (BRB) is intact, retinal microglia and the complement system are activated at low levels. This low-level of inflammation (para-inflammation) is believed to be essential to maintain homeostasis and restore functionality. However, prolonged stimulation by DAMPs in the diabetic eye leads to maladaptation of the innate immune system and dysregulated para-inflammation may contribute to DR development. In the advanced stages of DR where immune privilege is comprised, circulating immune cells and serum proteins may infiltrate the retina and participate in retinal chronic inflammation and retinal vascular and neuronal damage. This review discusses how the innate immune system is activated in diabetes and DR. The view also discusses why the protective immune response becomes detrimental in DR.
Collapse
|
12
|
Xu H, Chen M. Targeting the complement system for the management of retinal inflammatory and degenerative diseases. Eur J Pharmacol 2016; 787:94-104. [PMID: 26948311 PMCID: PMC5026403 DOI: 10.1016/j.ejphar.2016.03.001] [Citation(s) in RCA: 117] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Revised: 01/12/2016] [Accepted: 03/01/2016] [Indexed: 12/20/2022]
Abstract
The retina, an immune privileged tissue, has specialized immune defense mechanisms against noxious insults that may exist in diseases such as age-related macular degeneration (AMD), diabetic retinopathy (DR), uveoretinitis and glaucoma. The defense system consists of retinal innate immune cells (including microglia, perivascular macrophages, and a small population of dendritic cells) and the complement system. Under normal aging conditions, retinal innate immune cells and the complement system undergo a low-grade activation (parainflammation) which is important for retinal homeostasis. In disease states such as AMD and DR, the parainflammatory response is dysregulated and develops into detrimental chronic inflammation. Complement activation in the retina is an important part of chronic inflammation and may contribute to retinal pathology in these disease states. Here, we review the evidence that supports the role of uncontrolled or dysregulated complement activation in various retinal degenerative and angiogenic conditions. We also discuss current strategies that are used to develop complement-based therapies for retinal diseases such as AMD. The potential benefits of complement inhibition in DR, uveoretinitis and glaucoma are also discussed, as well as the need for further research to better understand the mechanisms of complement-mediated retinal damage in these disease states.
Collapse
Affiliation(s)
- Heping Xu
- Centre for Experimental Medicine, School of Medicine, Dentistry & Biomedical Sciences, Queen's University Belfast, UK.
| | - Mei Chen
- Centre for Experimental Medicine, School of Medicine, Dentistry & Biomedical Sciences, Queen's University Belfast, UK.
| |
Collapse
|
13
|
Sugita S, Iwasaki Y, Makabe K, Kimura T, Futagami T, Suegami S, Takahashi M. Lack of T Cell Response to iPSC-Derived Retinal Pigment Epithelial Cells from HLA Homozygous Donors. Stem Cell Reports 2016; 7:619-634. [PMID: 27641646 PMCID: PMC5063628 DOI: 10.1016/j.stemcr.2016.08.011] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 07/25/2016] [Accepted: 08/16/2016] [Indexed: 12/11/2022] Open
Abstract
Allografts of retinal pigment epithelial (RPE) cells have been considered for the treatment of ocular diseases. We recently started the transplantation of induced pluripotent stem cell (iPSC)-derived RPE cells for patients with age-related macular degeneration (autogenic grafts). However, there are at least two problems with this approach: (1) high cost, and (2) uselessness for acute patients. To resolve these issues, we established RPE cells from induced iPSCs in HLA homozygote donors. In vitro, human T cells directly recognized allogeneic iPSC-derived RPE cells that expressed HLA class I/II antigens. However, these T cells failed to respond to HLA-A, -B, and -DRB1-matched iPSC-derived RPE cells from HLA homozygous donors. Because of the lack of T cell response to iPSC-derived RPE cells from HLA homozygous donors, we can use these allogeneic iPSC-derived RPE cells in future clinical trials if the recipient and donor are HLA matched.
Collapse
Affiliation(s)
- Sunao Sugita
- Laboratory for Retinal Regeneration, RIKEN Center for Developmental Biology, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Yuko Iwasaki
- Laboratory for Retinal Regeneration, RIKEN Center for Developmental Biology, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan; Department of Ophthalmology and Visual Science, Tokyo Medical and Dental University Graduate School of Medicine and Dental Sciences, Tokyo 113-8519, Japan
| | - Kenichi Makabe
- Laboratory for Retinal Regeneration, RIKEN Center for Developmental Biology, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Takafumi Kimura
- Department of Fundamental Cell Technology, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Takaomi Futagami
- Department of Fundamental Cell Technology, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan; HLA Foundation Laboratory, Kyoto 600-8813, Japan
| | - Shinji Suegami
- Department of Fundamental Cell Technology, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan; HLA Foundation Laboratory, Kyoto 600-8813, Japan
| | - Masayo Takahashi
- Laboratory for Retinal Regeneration, RIKEN Center for Developmental Biology, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan.
| |
Collapse
|
14
|
YAN FENG, HE JIN, TANG LI, KONG YI, SHI YUHUA, CHEN SUIHUA, HUANG ZHENPING. Transforming growth factor-β2 increases the capacity of retinal pigment epithelial cells to induce the generation of regulatory T cells. Mol Med Rep 2015; 13:1367-72. [DOI: 10.3892/mmr.2015.4666] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2015] [Accepted: 10/26/2015] [Indexed: 11/05/2022] Open
|
15
|
Chen M, Xu H. Parainflammation, chronic inflammation, and age-related macular degeneration. J Leukoc Biol 2015; 98:713-25. [PMID: 26292978 PMCID: PMC4733662 DOI: 10.1189/jlb.3ri0615-239r] [Citation(s) in RCA: 255] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 07/28/2015] [Indexed: 12/13/2022] Open
Abstract
Inflammation is an adaptive response of the immune system to noxious insults to maintain homeostasis and restore functionality. The retina is considered an immune-privileged tissue as a result of its unique anatomic and physiologic properties. During aging, the retina suffers from a low-grade chronic oxidative insult, which sustains for decades and increases in level with advancing age. As a result, the retinal innate-immune system, particularly microglia and the complement system, undergoes low levels of activation (parainflammation). In many cases, this parainflammatory response can maintain homeostasis in the healthy aging eye. However, in patients with age-related macular degeneration, this parainflammatory response becomes dysregulated and contributes to macular damage. Factors contributing to the dysregulation of age-related retinal parainflammation include genetic predisposition, environmental risk factors, and old age. Dysregulated parainflammation (chronic inflammation) in age-related macular degeneration damages the blood retina barrier, resulting in the breach of retinal-immune privilege, leading to the development of retinal lesions. This review discusses the basic principles of retinal innate-immune responses to endogenous chronic insults in normal aging and in age-related macular degeneration and explores the difference between beneficial parainflammation and the detrimental chronic inflammation in the context of age-related macular degeneration.
Collapse
Affiliation(s)
- Mei Chen
- Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, United Kingdom
| | - Heping Xu
- Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, United Kingdom
| |
Collapse
|
16
|
Stem cell based therapies for age-related macular degeneration: The promises and the challenges. Prog Retin Eye Res 2015; 48:1-39. [DOI: 10.1016/j.preteyeres.2015.06.004] [Citation(s) in RCA: 133] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 06/05/2015] [Accepted: 06/11/2015] [Indexed: 12/21/2022]
|
17
|
Madelung CF, Falk MK, Sørensen TL. The association between neovascular age-related macular degeneration and regulatory T cells in peripheral blood. Clin Ophthalmol 2015; 9:1147-54. [PMID: 26170606 PMCID: PMC4485850 DOI: 10.2147/opth.s82116] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
PURPOSE To investigate regulatory T cells (Tregs) and subsets of the Treg population in patients with neovascular age-related macular degeneration (AMD). PATIENTS AND METHODS Twenty-one neovascular AMD cases and 12 age-matched controls without retinal pathology were selected. Patients were recruited from our outpatient retinal clinic. Control individuals were typically spouses. The diagnosis of neovascular AMD was confirmed using fluorescein and indocyaningreen angiography. Fresh venous blood was analyzed by flow cytometry using fluorochrome-conjugated antibodies to the Treg surface antigens CD4, CD25, CD127, CD45RA, and CD31. Main outcome measures were the percentage of CD25(high)CD127(low) Tregs, the percentage of CD45RA(+) naïve Tregs, and the percentage of CD31(+) recent thymic emigrant Tregs. RESULTS Comparing patients with neovascular AMD to controls, no significant differences were found in the percentages of CD4(+) lymphocytes, CD25(high)CD127(low) Tregs, CD45RA(+) naïve Tregs, or CD31(+) recent thymic emigrant Tregs. CONCLUSION Our data does not indicate an altered state of systemic Treg cells in neovascular AMD.
Collapse
Affiliation(s)
- Christopher Fugl Madelung
- Clinical Eye Research Unit, Department of Ophthalmology, Copenhagen University Hospital, Roskilde, Denmark ; University of Copenhagen, Copenhagen, Denmark
| | - Mads Krüger Falk
- Clinical Eye Research Unit, Department of Ophthalmology, Copenhagen University Hospital, Roskilde, Denmark ; University of Copenhagen, Copenhagen, Denmark
| | - Torben Lykke Sørensen
- Clinical Eye Research Unit, Department of Ophthalmology, Copenhagen University Hospital, Roskilde, Denmark ; University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
18
|
Horie S, Robbie SJ, Liu J, Wu WK, Ali RR, Bainbridge JW, Nicholson LB, Mochizuki M, Dick AD, Copland DA. CD200R signaling inhibits pro-angiogenic gene expression by macrophages and suppresses choroidal neovascularization. Sci Rep 2013; 3:3072. [PMID: 24170042 PMCID: PMC3812658 DOI: 10.1038/srep03072] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Accepted: 10/11/2013] [Indexed: 12/23/2022] Open
Abstract
Macrophages are rapidly conditioned by cognate and soluble signals to acquire phenotypes that deliver specific functions during inflammation, wound healing and angiogenesis. Whether inhibitory CD200R signaling regulates pro-angiogenic macrophage phenotypes with the potential to suppress ocular neovascularization is unknown. CD200R-deficient bone marrow derived macrophages (BMMΦ) were used to demonstrate that macrophages lacking this inhibitory receptor exhibit enhanced levels of Vegfa, Arg-1 and Il-1β when stimulated with PGE2 or RPE-conditioned (PGE2-enriched) media. Endothelial tube formation in HUVECs was increased when co-cultured with PGE2-conditioned CD200R−/− BMMΦ, and laser-induced choroidal neovascularization was enhanced in CD200R-deficient mice. In corroboration, signaling through CD200R results in the down-regulation of BMMΦ angiogenic and pro-inflammatory phenotypes. Translational potential of this pathway was investigated in the laser-induced model of choroidal neovascularization. Local delivery of a CD200R agonist mAb to target myeloid infiltrate alters macrophage phenotype and inhibits pro-angiogenic gene expression, which suppresses pathological angiogenesis and CNV development.
Collapse
Affiliation(s)
- Shintaro Horie
- 1] Academic Unit of Ophthalmology, School of Clinical Sciences, University of Bristol, Bristol, UK [2] Department of Ophthalmology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan [3]
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
The local immune response to intraocular Toxoplasma re-challenge: less pathology and better parasite control through Treg/Th1/Th2 induction. Int J Parasitol 2013; 43:721-8. [PMID: 23702129 DOI: 10.1016/j.ijpara.2013.04.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Revised: 04/09/2013] [Accepted: 04/10/2013] [Indexed: 01/10/2023]
Abstract
Ocular toxoplasmosis is a major cause of blindness world-wide. Ocular involvement is frequently seen following congenital infection. Many of these infections are quiescent but pose a life-time risk of reactivation. However, the physiopathology of ocular toxoplasmosis reactivation is largely unexplored. We previously developed a Swiss-Webster outbred mouse model for congenital toxoplasmosis by neonatal injection of Toxoplasma gondii cysts. We also used a mouse model of direct intraocular infection to show a deleterious local T helper 17 type response upon primary infection. In the present study, our two models were combined to study intravitreal re-challenge of neonatally infected mice, as an approximate model of reactivation, in comparison with a primary ocular infection. Using BioPlex proteomic assays in aqueous humour and reverse transcription-PCR for T helper cell transcription factors, we observed diminished T helper 17 type reaction in reinfection, compared with primary infection. In contrast, T helper 2 and T regulatory responses were enhanced. Interestingly, this was also true for T helper 1 markers such as IFN-γ, which was paralleled by better parasite control. Secretion of IL-27, a central cytokine for shifting the immune response from T helper 17 to T helper 1, was also greatly enhanced. We observed a similar protective immune reaction pattern in the eye upon reinfection with the virulent RH strain, with the notable exception of IFN-γ. In summary, our results show that the balance is shifted from T helper 17 to a less pathogenic but more effective anti-parasite Treg/T helper 1/T helper 2 pattern in a reactivation setting.
Collapse
|
20
|
Forrester JV, Steptoe RJ, Klaska IP, Martin-Granados C, Dua HS, Degli-Esposti MA, Wikstrom ME. Cell-based therapies for ocular inflammation. Prog Retin Eye Res 2013; 35:82-101. [PMID: 23542232 DOI: 10.1016/j.preteyeres.2013.02.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Revised: 01/31/2013] [Accepted: 02/01/2013] [Indexed: 12/13/2022]
Abstract
Since the plasticity and the potential for re-programming cells has become widely accepted, there has been great interest in cell-based therapies. These are being applied to a range of diseases, not least ocular diseases, where it is assumed that there is a reduced risk of immune rejection although this may be more perceived than real. There are two broad classes of cell-based therapies: those aimed at restoring structure and function of specific tissues and cells; and those directed towards restoring immunological homeostasis by controlling the damaging effects of inflammatory disease. Stem cells of all types represent the first group and prototypically have been used with the aim of regenerating failing cells. In contrast, immune cells have been suggested as potential modulators of inflammation. However, there is functional overlap in these two applications, with some types of stem cells, such as mesenchymal stem cells, demonstrating a potent immunomodulatory effect. This review summarises recent information on cell based therapies for ocular disease, with special emphasis on ocular inflammatory disease, and explores current uses, potential and limitations.
Collapse
Affiliation(s)
- John V Forrester
- Immunology Program, Centre for Ophthalmology and Visual Science, The University of Western Australia, Perth, Western Australia, Australia.
| | | | | | | | | | | | | |
Collapse
|
21
|
Mochizuki M, Sugita S, Kamoi K. Immunological homeostasis of the eye. Prog Retin Eye Res 2012; 33:10-27. [PMID: 23108335 DOI: 10.1016/j.preteyeres.2012.10.002] [Citation(s) in RCA: 111] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Revised: 10/05/2012] [Accepted: 10/05/2012] [Indexed: 12/22/2022]
Abstract
Uveitis is a sight-threatening disease caused by autoimmune or infection-related immune responses. Studies in experimental autoimmune uveitis and in human diseases imply that activated CD4(+) T cells, Th1 and Th17 cells, play an effector role in ocular inflammation. The eye has a unique regional immune system to protect vision-related cells and tissues from these effector T cells. The immunological balance between the pathogenic CD4(+) T cells and regional immune system in the eye contributes to the maintenance of ocular homeostasis and good vision. Current studies have demonstrated that ocular parenchymal cells at the inner surface of the blood-ocular barrier, i.e. corneal endothelial (CE) cells, iris pigment epithelial (PE) cells, ciliary body PE cells, and retinal PE cells, contribute to the regional immune system of the eye. Murine ocular resident cells directly suppress activation of bystander T cells and production of inflammatory cytokines. The ocular resident cells possess distinct properties of immunoregulation that are related to disparate anatomical location. CE cells and iris PE cells, which are located at the anterior segment of the eye and face the aqueous humor, suppress activation of T cells via cell-to-cell contact mechanisms, whereas retinal PE cells suppress the activation of T cells via soluble factors. In addition to direct immune suppression, the ocular resident cells have another unique immunosuppressive property, the induction of CD25(+)Foxp3(+) Treg cells that also suppress the activation of bystander T cells. Iris PE cells convert CD8(+) T cells into Treg cells, while retinal PE cells convert CD4(+) T cells greatly and CD8(+) T cells moderately into Treg cells. CE cells also convert both CD4(+) T cells and CD8(+) T cells into Treg cells. The immunomodulation by ocular resident cells is mediated by various soluble or membrane-bound molecules that include TGF-β TSP-1, B7-2 (CD86), CTLA-2α, PD-L1 (B7-H1), galectin 1, pigment epithelial-derived factor PEDF), GIRTL, and retinoic acid. Human retinal PE cells also possess similar immune properties to induce Treg cells. Although there are many issues to be answered, human Treg cells induced by ocular resident cells such as retinal PE cells and related immunosuppressive molecules can be applied as immune therapy for refractive autoimmune uveitis in humans in the future.
Collapse
Affiliation(s)
- Manabu Mochizuki
- Department of Ophthalmology & Visual Science, Tokyo Medical and Dental University, 1-5-45 Yushima, Tokyo 113-8519, Japan.
| | | | | |
Collapse
|
22
|
Sauer A, Pfaff AW, Villard O, Creuzot-Garcher C, Dalle F, Chiquet C, Pelloux H, Speeg-Schatz C, Gaucher D, Prevost G, Bourcier T, Candolfi E. Interleukin 17A as an Effective Target for Anti-inflammatory and Antiparasitic Treatment of Toxoplasmic Uveitis. J Infect Dis 2012; 206:1319-29. [DOI: 10.1093/infdis/jis486] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
|
23
|
Abstract
PURPOSE OF REVIEW Current scientific evidence suggests that the systemic immune response is affected by exposure to light. During the past century man has been exposed for the first time in evolution to light at night, as well as increasing ultraviolet radiation through depletion of the ozone layer in our atmosphere. These ecological changes have enhanced the impact of light on our systemic immune response. We will review the effect of light on the systemic immune response with particular emphasis on ocular immunity. RECENT FINDINGS Visible light is now recognized to be important in the maintenance of immune privilege within the eye; however, little is known about the mechanism through which this effect occurs. Recent studies suggest that the generation of regulatory T cells involved in immune privilege within the eye is dependent on retinoic acid formation by retinal pigment epithelial cells. Light is also important in modulation of multiple pathways including adjustment of circadian rhythm and production of vitamin D. SUMMARY Light regulates our biologic systems in many different ways. Its effect on the systemic immune response suggests that it is important in maintaining health, as well as in the induction of disease. A better understanding of the interaction of light with our biologic systems may allow new preventive measures to avoid disease and novel forms of treatment.
Collapse
|
24
|
Piquet AL, Venkiteswaran K, Marupudi NI, Berk M, Subramanian T. The immunological challenges of cell transplantation for the treatment of Parkinson's disease. Brain Res Bull 2012; 88:320-31. [PMID: 22521427 DOI: 10.1016/j.brainresbull.2012.03.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2012] [Revised: 03/07/2012] [Accepted: 03/09/2012] [Indexed: 01/24/2023]
Abstract
Dopaminergic cell transplantation is an experimental therapy for Parkinson's disease (PD). It has many potential theoretical advantages over current treatment strategies such as providing continuous local dopaminergic replenishment, eliminating motor fluctuations and medication-induced dyskinesias, slowing down disease progression or even reversing disease pathology in the host. Recent studies also show that dopaminergic cell transplants provide long-term neuromodulation in the basal ganglia that simulates the combined effects of oral dopaminergic therapy and surgical therapies like deep brain stimulation, the contemporary therapeutic approach to advanced PD. However, dopaminergic cell transplantation in PD as not been optimized and current experimental techniques have many drawbacks. In published experiments to date of attempted dopaminergic grafting in PD, the major challenges are unacceptable graft-induced dyskinesias or failure of such grafts to exceed the benefits afforded by sham surgery. A deleterious host immune response to the transplant has been implicated as a major putative cause for these adverse outcomes. This article focuses on recent advances in understanding the immunology of the transplantation in PD and possible methods to overcome adverse events such that we could translate cell replacement strategies into viable clinical treatments in the future.
Collapse
Affiliation(s)
- Amanda L Piquet
- Department of Neurology, The Pennsylvania State University College of Medicine, Hershey, United States
| | | | | | | | | |
Collapse
|
25
|
Bharti K, Miller SS, Arnheiter H. The new paradigm: retinal pigment epithelium cells generated from embryonic or induced pluripotent stem cells. Pigment Cell Melanoma Res 2010; 24:21-34. [PMID: 20846177 DOI: 10.1111/j.1755-148x.2010.00772.x] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Compared with neural crest-derived melanocytes, retinal pigment epithelium (RPE) cells in the back of the eye are pigment cells of a different kind. They are a part of the brain, form an epithelial monolayer, respond to distinct extracellular signals, and provide functions that far exceed those of a light-absorbing screen. For instance, they control nutrient and metabolite flow to and from the retina, replenish 11-cis-retinal by re-isomerizing all-trans-retinal generated during photoconversion, phagocytose daily a portion of the photoreceptors' outer segments, and secrete cytokines that locally control the innate and adaptive immune systems. Not surprisingly, RPE cell damage is a major cause of human blindness worldwide, with age-related macular degeneration a prevalent example. RPE replacement therapies using RPE cells generated from embryonic or induced pluripotent stem cells provide a novel approach to a rational treatment of such forms of blindness. In fact, RPE-like cells can be obtained relatively easily when stem cells are subjected to a two-step induction protocol, a first step that leads to a neuroectodermal fate and a second to RPE differentiation. Here, we discuss the characteristics of such cells, propose criteria they should fulfill in order to be considered authentic RPE cells, and point out the challenges one faces when using such cells in attempts to restore vision.
Collapse
Affiliation(s)
- Kapil Bharti
- Mammalian Development Section, National Institutes of Neurological Disorders and Stroke, Bethesda, MD, USA.
| | | | | |
Collapse
|
26
|
Ke Y, Sun D, Jiang G, Kaplan HJ, Shao H. PD-L1(hi) retinal pigment epithelium (RPE) cells elicited by inflammatory cytokines induce regulatory activity in uveitogenic T cells. J Leukoc Biol 2010; 88:1241-9. [PMID: 20739617 DOI: 10.1189/jlb.0610332] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
We previously reported that after exposure to inflammatory cytokines, such as IL-17 and IFN-γ, RPE cells express increased amounts of suppressor of cytokine signaling, leading to general suppression of the inflammatory response. Here, we demonstrate that RPE cells expressed increased levels of PD-L1 in response to IL-17, IFN-γ, or Poly I:C. These PD-L1(hi) RPE cells inhibited the pathogenic activities of IRBP-specific T cells, which usually induced uveitis when injected into naïve mice (EAU). The suppressed pathogenicity of these uveitogenic T cells after exposure to PD-L1(hi) RPE cells could be partially reversed by anti-PD-L1 antibodies. Nevertheless, IRBP-specific T cells pre-exposed to PD-L1(hi) RPE cells displayed substantial suppressor activity, which strongly inhibited the activation of fresh IRBP-Teffs in response to subsequent antigenic challenge and when transferred into naïve mice, inhibited the induction of EAU by IRBP-Teff transfer. These findings suggest that inflammatory cytokine-triggered up-regulation of PD-L1 on RPE constitutes a critical factor for inducing infiltrated uveitogenic T cells with regulatory activities, which may accelerate the natural resolution of T cell-mediated intraocular inflammation.
Collapse
Affiliation(s)
- Yan Ke
- Department of Ophthalmology and Vision Sciences, University of Louisville, 301 E. Muhammad Ali Blvd., Louisville, KY 40202, USA.
| | | | | | | | | |
Collapse
|