1
|
Bao H, Tian Y, Wang H, Ye T, Wang S, Zhao J, Qiu Y, Li J, Pan C, Ma G, Wei W, Tao Y. Exosome-loaded degradable polymeric microcapsules for the treatment of vitreoretinal diseases. Nat Biomed Eng 2024; 8:1436-1452. [PMID: 37872369 DOI: 10.1038/s41551-023-01112-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 09/26/2023] [Indexed: 10/25/2023]
Abstract
The therapeutic benefits of many cell types involve paracrine mechanisms. Inspired by the paracrine functions of exosomes and the sustained degradation properties of microcapsules, here we report the therapeutic benefits of exosome-loaded degradable poly(lactic-co-glycolic acid) microcapsules with micrometric pores for the treatment of vitreoretinal diseases. On intravitreal injection in a mouse model of retinal ischaemia-reperfusion injury, microcapsules encapsulating mouse mesenchymal-stem-cell-derived exosomes settled in the inferior vitreous cavity, released exosomes for over one month as they underwent degradation and led to the restoration of retinal thickness to nearly that of the healthy retina. In mice and non-human primates with primed mycobacterial uveitis, intravitreally injected microcapsules loaded with exosomes from monkey regulatory T cells resulted in a substantial reduction in the levels of inflammatory cells. The exosome-encapsulating microcapsules, which can be lyophilised, may offer alternative treatment options for vitreoretinal diseases.
Collapse
Affiliation(s)
- Han Bao
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, P. R. China
- Department of Ophthalmology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, P. R. China
| | - Ying Tian
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, P. R. China
- Department of Ophthalmology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, P. R. China
| | - Haixin Wang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, P. R. China
| | - Tong Ye
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, P. R. China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Shuang Wang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, P. R. China
| | - Jiawei Zhao
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, P. R. China
| | - Yefeng Qiu
- Laboratory Animal Center of the Academy of Military Medical Sciences, Beijing, P. R. China
| | - Jian Li
- Department of Ophthalmology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, P. R. China
| | - Chao Pan
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, P. R. China
| | - Guanghui Ma
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, P. R. China.
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, P. R. China.
| | - Wei Wei
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, P. R. China.
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, P. R. China.
| | - Yong Tao
- Department of Ophthalmology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, P. R. China.
| |
Collapse
|
2
|
Sin TN, Tng N, Dragoli J, Ramesh Kumar S, Villafuerte-Trisolini C, Chung SH, Tu L, Le SM, Shim JH, Pepple KL, Ravindran R, Khan IH, Moshiri A, Thomasy SM, Yiu G. Safety and efficacy of CRISPR-mediated genome ablation of VEGFA as a treatment for choroidal neovascularization in nonhuman primate eyes. Mol Ther 2024:S1525-0016(24)00651-8. [PMID: 39342431 DOI: 10.1016/j.ymthe.2024.09.027] [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: 04/05/2024] [Revised: 08/30/2024] [Accepted: 09/25/2024] [Indexed: 10/01/2024] Open
Abstract
CRISPR-based genome editing enables permanent suppression of angiogenic factors such as vascular endothelial growth factor (VEGF) as a potential treatment for choroidal neovascularization (CNV)-a major cause of blindness in age-related macular degeneration. We previously designed adeno-associated viral (AAV) vectors with S. pyogenes Cas 9 (SpCas9) and guide RNAs (gRNAs) to target conserved sequences in VEGFA across mouse, rhesus macaque, and human, with successful suppression of VEGF and laser-induced CNV in mice. Here, we advanced the platform to nonhuman primates and found that subretinal AAV8-SpCas9 with gRNAs targeting VEGFA may reduce VEGF and CNV severity as compared with SpCas9 without gRNAs. However, all eyes that received AAV8-SpCas9 regardless of gRNA presence developed subfoveal deposits, concentric macular rings, and outer retinal disruption that worsened at higher dose. Immunohistochemistry showed subfoveal accumulation of retinal pigment epithelial cells, collagen, and vimentin, disrupted photoreceptor structure, and retinal glial and microglial activation. Subretinal AAV8-SpCas9 triggered aqueous elevations in CCL2, but minimal systemic humoral or cellular responses against AAV8, SpCas9, or GFP reporter. Our findings suggest that CRISPR-mediated VEGFA ablation in nonhuman primate eyes may suppress VEGF and CNV, but can also lead to unexpected subretinal fibrosis, photoreceptor damage, and retinal inflammation despite minimal systemic immune responses.
Collapse
Affiliation(s)
- Tzu-Ni Sin
- Department of Ophthalmology & Vision Sciences, University of California Davis, Davis, CA 95616, USA
| | - Nicole Tng
- Department of Ophthalmology & Vision Sciences, University of California Davis, Davis, CA 95616, USA
| | - Jack Dragoli
- Department of Ophthalmology & Vision Sciences, University of California Davis, Davis, CA 95616, USA
| | - Sruthi Ramesh Kumar
- Department of Ophthalmology & Vision Sciences, University of California Davis, Davis, CA 95616, USA
| | | | - Sook Hyun Chung
- Department of Ophthalmology & Vision Sciences, University of California Davis, Davis, CA 95616, USA
| | - Lien Tu
- Department of Ophthalmology & Vision Sciences, University of California Davis, Davis, CA 95616, USA
| | - Sophie M Le
- Department of Ophthalmology & Vision Sciences, University of California Davis, Davis, CA 95616, USA
| | - Jae Ho Shim
- Department of Surgical & Radiological Sciences, University of California Davis, Davis, CA 95616, USA
| | - Kathryn L Pepple
- Department of Ophthalmology, University of Washington, Seattle, WA 98104, USA
| | - Resmi Ravindran
- Department of Pathology, University of California Davis Medical Center, Sacramento, CA 95817, USA
| | - Imran H Khan
- Department of Pathology, University of California Davis Medical Center, Sacramento, CA 95817, USA
| | - Ala Moshiri
- Department of Ophthalmology & Vision Sciences, University of California Davis, Davis, CA 95616, USA
| | - Sara M Thomasy
- Department of Ophthalmology & Vision Sciences, University of California Davis, Davis, CA 95616, USA; Department of Surgical & Radiological Sciences, University of California Davis, Davis, CA 95616, USA
| | - Glenn Yiu
- Department of Ophthalmology & Vision Sciences, University of California Davis, Davis, CA 95616, USA.
| |
Collapse
|
3
|
Degroote RL, Schmalen A, Hauck SM, Deeg CA. Unveiling Differential Responses of Granulocytes to Distinct Immunostimulants with Implications in Autoimmune Uveitis. Biomedicines 2023; 12:19. [PMID: 38275380 PMCID: PMC10812922 DOI: 10.3390/biomedicines12010019] [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: 11/23/2023] [Revised: 12/13/2023] [Accepted: 12/15/2023] [Indexed: 01/27/2024] Open
Abstract
The perception of circulating granulocytes as cells with a predetermined immune response mainly triggered by pathogens is evolving, recognizing their functional heterogeneity and adaptability, particularly within the neutrophil subset. The involvement of these cells in the pathophysiology of autoimmune uveitis has become increasingly clear, yet their exact role remains elusive. We used an equine model for autoimmune-mediated recurrent pan-uveitis to investigate early responses of granulocytes in different inflammatory environments. For this purpose, we performed differential proteomics on granulocytes from healthy and diseased horses stimulated with IL8, LPS, or PMA. Compared to healthy horses, granulocytes from the recurrent uveitis model significantly changed the cellular abundance of 384 proteins, with a considerable number of specific changes for each stimulant. To gain more insight into the functional impact of these stimulant-specific proteome changes in ERU pathogenesis, we used Ingenuity Pathway Analysis for pathway enrichment. This resulted in specific reaction patterns for each stimulant, with IL8 predominantly promoting Class I MHC-mediated antigen processing and presentation, LPS enhancing processes in phospholipid biosynthesis, and PMA, clearly inducing neutrophil degranulation. These findings shed light on the remarkably differentiated responses of neutrophils, offering valuable insights into their functional heterogeneity in a T-cell-driven disease. Raw data are available via ProteomeXchange with identifier PXD013648.
Collapse
Affiliation(s)
- Roxane L. Degroote
- Chair of Physiology, Department of Veterinary Sciences, LMU Munich, D-82152 Martinsried, Germany; (R.L.D.); (A.S.)
| | - Adrian Schmalen
- Chair of Physiology, Department of Veterinary Sciences, LMU Munich, D-82152 Martinsried, Germany; (R.L.D.); (A.S.)
- Metabolomics and Proteomics Core, Helmholtz Center Munich, German Research Center for Environmental Health, D-80939 Munich, Germany;
| | - Stefanie M. Hauck
- Metabolomics and Proteomics Core, Helmholtz Center Munich, German Research Center for Environmental Health, D-80939 Munich, Germany;
| | - Cornelia A. Deeg
- Chair of Physiology, Department of Veterinary Sciences, LMU Munich, D-82152 Martinsried, Germany; (R.L.D.); (A.S.)
| |
Collapse
|
4
|
Wu X, Tao M, Zhu L, Zhang T, Zhang M. Pathogenesis and current therapies for non-infectious uveitis. Clin Exp Med 2023; 23:1089-1106. [PMID: 36422739 PMCID: PMC10390404 DOI: 10.1007/s10238-022-00954-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 11/16/2022] [Indexed: 11/25/2022]
Abstract
Non-infectious uveitis (NIU) is a disorder with various etiologies and is characterized by eye inflammation, mainly affecting people of working age. An accurate diagnosis of NIU is crucial for appropriate therapy. The aim of therapy is to improve vision, relieve ocular inflammation, prevent relapse, and avoid treatment side effects. At present, corticosteroids are the mainstay of topical or systemic therapy. However, repeated injections are required for the treatment of chronic NIU. Recently, new drug delivery systems that may ensure intraocular delivery of therapeutic drug levels have been highlighted. Furthermore, with the development of immunosuppressants and biologics, specific therapies can be selected based on the needs of each patient. Immunosuppressants used in the treatment of NIU include calcineurin inhibitors and antimetabolites. However, systemic immunosuppressive therapy itself is associated with adverse effects due to the inhibition of immune function. In patients with refractory NIU or those who cannot tolerate corticosteroids and immunosuppressors, biologics have emerged as alternative treatments. Thus, to improve the prognosis of patients with NIU, NIU should be managed with different drugs according to the response to treatment and possible side effects.
Collapse
Affiliation(s)
- Xue Wu
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Save Sight Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2000, Australia
| | - Mengying Tao
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ling Zhu
- Save Sight Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2000, Australia
| | - Ting Zhang
- Save Sight Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2000, Australia
| | - Ming Zhang
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
5
|
Nirbhavane P, Moksha L, Sharma G, Velpandian T, Singh B, Katare OP. Cationic Nano-Lipidic Carrier Mediated Ocular Delivery of Triamcinolone Acetonide: A Preclinical Investigation in the Management of Uveitis. Life (Basel) 2023; 13:life13041057. [PMID: 37109586 PMCID: PMC10143093 DOI: 10.3390/life13041057] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 03/29/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
The current study was undertaken to evaluate the efficacy of a novel nano-lipoidal eye drop formulation of triamcinolone acetonide (TA) for the topical treatment of uveitis. The triamcinolone acetonide-loaded nanostructured lipid carriers (cTA-NLC) were developed by employing 'hot microemulsion method' using biocompatible lipids, which exhibited a sustained release nature and enhanced efficacy when evaluated in vitro. The in vivo efficacy of this developed formulation was tested on Wistar rats, and a single-dose pharmacokinetic study was conducted in rabbits. The eyes of animals were examined for any signs of inflammation using the 'Slit-lamp microscopic' method. The aqueous humor collected from the sacrificed rats was tested for total protein count and cell count. The total protein count was determined using BSA assay method, while the total cell count was determined by Neubaur's hemocytometer method. The results showed that the cTA-NLC formulation had negligible signs of inflammation, with a clinical score of uveitis 0.82 ± 0.166, which is much less than control/untreated (3.80 ± 0.3) and free drug suspension (2.66 ± 0.405). The total cell count was also found to be significantly low for cTA-NLC (8.73 ± 1.79 × 105) as compared to control (52.4 ± 7.71 × 105) and free drug suspension (30.13 ± 3.021 × 105). Conclusively, the animal studies conducted showed that our developed formulation holds the potential for effective management of uveitis.
Collapse
Affiliation(s)
- Pradip Nirbhavane
- UGC-Centre of Advanced Study, Division of Pharmaceutics, University Institute of Pharmaceutical Sciences (UIPS), Panjab University, Chandigarh 160014, India
| | - Laxmi Moksha
- Dr. Rajendra Prasad Centre for Ophthalmic Sciences, Ocular Pharmacology and Pharmacy Division, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Gajanand Sharma
- UGC-Centre of Advanced Study, Division of Pharmaceutics, University Institute of Pharmaceutical Sciences (UIPS), Panjab University, Chandigarh 160014, India
| | - Thirumurthy Velpandian
- Dr. Rajendra Prasad Centre for Ophthalmic Sciences, Ocular Pharmacology and Pharmacy Division, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Bhupinder Singh
- UGC-Centre of Advanced Study, Division of Pharmaceutics, University Institute of Pharmaceutical Sciences (UIPS), Panjab University, Chandigarh 160014, India
| | - O P Katare
- UGC-Centre of Advanced Study, Division of Pharmaceutics, University Institute of Pharmaceutical Sciences (UIPS), Panjab University, Chandigarh 160014, India
| |
Collapse
|
6
|
Wilson L, Lewis KE, Evans LS, Dillon SR, Pepple KL. Systemic Administration of Acazicolcept, a Dual CD28 and Inducible T cell Costimulator Inhibitor, Ameliorates Experimental Autoimmune Uveitis. Transl Vis Sci Technol 2023; 12:27. [PMID: 36976157 PMCID: PMC10064916 DOI: 10.1167/tvst.12.3.27] [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: 02/08/2023] [Accepted: 02/21/2023] [Indexed: 03/29/2023] Open
Abstract
Purpose Combined inhibition of CD28 and inducible T cell costimulator (ICOS) pathways with acazicolcept (ALPN-101) represents a potential new treatment for uveitis. Here, we evaluate preclinical efficacy using experimental autoimmune uveitis (EAU) in Lewis rats. Methods Efficacy was tested in 57 Lewis rats treated with either systemic (subcutaneous) or local (intravitreal) administration of acazicolcept and compared to treatment with a matched Fc-only control or corticosteroid. Impact of treatment on uveitis was assessed using clinical scoring, optical coherence tomography (OCT), and histology. Ocular effector T cell populations were determined using flow cytometry, and multiplex ELISA used to measure aqueous cytokine concentrations. Results When compared to Fc control treatment, systemic acazicolcept led to statistically significant decreases in clinical score (P < 0.01), histologic score (P < 0.05), and number of ocular CD45+ cells (P < 0.01). Number of ocular CD4+ and CD8+ T cells expressing IL-17A+ and IFNγ+ were also decreased with statistical significance (P < 0.01). Similar results were achieved with corticosteroids. Intravitreal acazicolcept decreased inflammation scores when compared to untreated fellow eyes and to Fc control treated eyes, although not statistically significant. Systemic toxicity, measured by weight loss, occurred in the corticosteroid-treated, but not in the acazicolcept-treated animals. Conclusions Systemic treatment with acazicolcept statistically significantly suppressed EAU. Acazicolcept was well-tolerated without the weight loss associated with corticosteroids. Acazicolcept may be an effective alternative to corticosteroids for use in treating autoimmune uveitis. Additional studies are needed to clarify the optimal dose and route for use in humans. Translational Relevance We show that T cell costimulatory blockade could be an effective mechanism for treating uveitis.
Collapse
Affiliation(s)
- Leslie Wilson
- Department of Ophthalmology, University of Washington, Seattle, WA, USA
- Roger and Angie Karalis Johnson Retina Center, University of Washington School of Medicine, Seattle, WA, USA
| | | | | | - Stacey R. Dillon
- Translational Medicine, Alpine Immune Sciences, Seattle, WA, USA
| | - Kathryn L. Pepple
- Department of Ophthalmology, University of Washington, Seattle, WA, USA
- Roger and Angie Karalis Johnson Retina Center, University of Washington School of Medicine, Seattle, WA, USA
| |
Collapse
|
7
|
Pepple KL, John S, Wilson L, Wang V, Van Gelder RN. Systemic prime exacerbates the ocular immune response to heat-killed Mycobacterium tuberculosis. Exp Eye Res 2022; 223:109198. [PMID: 35921962 PMCID: PMC10240933 DOI: 10.1016/j.exer.2022.109198] [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: 03/31/2022] [Revised: 06/23/2022] [Accepted: 07/18/2022] [Indexed: 11/16/2022]
Abstract
Post-infectious uveitis describes the condition of chronic immune mediated ocular inflammation associated with pathogens such as Mycobacterium tuberculosis (Mtb). Mtb associated post-infectious uveitis can be modeled in mice by intravitreal injection of heat-killed Mtb (HKMtb). To better understand how prior systemic exposure to the pathogen alters the local immune response to Mtb, we used flow cytometry and multiplex ELISAs to compare ocular responses to intravitreal HKMtb in the presence or absence of a systemic "prime" of HKMtb. Priming resulted in exacerbation of local inflammation with significantly increased clinical and histologic inflammation scores and increased vitreous cytokines concentrations one day after intravitreal injection of HKMtb. Seven days after injection, uveitis in unprimed animals had largely resolved. In contrast in primed animals, clinical signs of chronic inflammation were associated with a significant increase in the number of ocular T cells, NK cells, and Ly6Chi macrophages and increasing vitreous concentrations of IL-17, VEGF, MIG(CXCL9), IP-10(CXCL10), IL-12p40 and MIP-1α(CCL3). In mice lacking mature T and B cells (RAG2 deficient), the impact of priming on the ocular immune response was ameliorated with significantly lower vitreous cytokine concentrations and spontaneous resolution of uveitis. Altogether these results suggest that the ocular response to Mtb is exacerbated by prior systemic Mtb infection and chronic post-infectious uveitis is mediated by local production of cytokines and chemokines that amplify Th17 and Th1 responses. This mouse model of chronic Mtb associated uveitis will help elucidate mechanisms of disease in patients with post-infectious uveitis.
Collapse
Affiliation(s)
- Kathryn L Pepple
- University of Washington, Department of Ophthalmology, Seattle, WA, 98104, USA; University of Washington, Roger and Angie Karalis Johnson Retina Center, Seattle, WA, 98109, USA.
| | - Sarah John
- University of Washington, Department of Ophthalmology, Seattle, WA, 98104, USA
| | - Leslie Wilson
- University of Washington, Department of Ophthalmology, Seattle, WA, 98104, USA
| | - Victoria Wang
- University of Washington, Department of Ophthalmology, Seattle, WA, 98104, USA
| | - Russell N Van Gelder
- University of Washington, Department of Ophthalmology, Seattle, WA, 98104, USA; University of Washington, Department of Biological Structure, Seattle, WA, 98195, USA; University of Washington, Department of Laboratory Medicine and Pathology, Seattle, WA, 98195, USA; University of Washington, Roger and Angie Karalis Johnson Retina Center, Seattle, WA, 98109, USA
| |
Collapse
|
8
|
Song J, Guo D, Tian Q, Wu Q, Zhang X, Bi H. Stress Suppresses Systemic Th17/Treg Imbalance in Rats with Experimental Autoimmune Uveitis. Ocul Immunol Inflamm 2022; 30:1890-1900. [PMID: 34468261 DOI: 10.1080/09273948.2021.1970778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE To explore the effect of chronic unpredictable mild stress (CUMS) on the immune response involved in rats with experimental autoimmune uveitis (EAU). METHODS Lewis rats were randomly divided into control, EAU, CUMS, and EAU+CUMS groups and received relevant treatments. On days 7, 11, 14, 21 and 28, frequencies of Th17 and Treg cells and the related cytokines were analyzed. RESULTS The intraocular inflammation of EAU rats peaked between days 11 and 13, while the severity of inflammation of the rats in EAU+CUMS group fluctuated between 11 and 15 days. Both frequencies of Th17, Treg cells and the related cytokines exhibited a significant difference between the two groups on days 11 and 14. CONCLUSION CUMS may protect against the possible harmful effects of immune disorder in rats with EAU through suppressing the immune disorder of T lymphocyte and the related cytokine responses.
Collapse
Affiliation(s)
- Jike Song
- Medical College of Optometry and Ophthalmology, Shandong University of Traditional Chinese Medicine, Jinan, P. R. China.,Shandong Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Therapy of Ocular Diseases, Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Therapy of Ocular Diseases in Universities of Shandong, Eye Institute, Shandong University of Traditional Chinese Medicine, Jinan, P. R. China
| | - Dadong Guo
- Shandong Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Therapy of Ocular Diseases, Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Therapy of Ocular Diseases in Universities of Shandong, Eye Institute, Shandong University of Traditional Chinese Medicine, Jinan, P. R. China
| | - Qingmei Tian
- Shandong Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Therapy of Ocular Diseases, Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Therapy of Ocular Diseases in Universities of Shandong, Eye Institute, Shandong University of Traditional Chinese Medicine, Jinan, P. R. China.,Affiliated Eye Hospital, Shandong University of Traditional Chinese Medicine, Jinan, P. R. China
| | - Qiuxin Wu
- Shandong Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Therapy of Ocular Diseases, Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Therapy of Ocular Diseases in Universities of Shandong, Eye Institute, Shandong University of Traditional Chinese Medicine, Jinan, P. R. China.,Affiliated Eye Hospital, Shandong University of Traditional Chinese Medicine, Jinan, P. R. China
| | - Xiuyan Zhang
- Shandong Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Therapy of Ocular Diseases, Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Therapy of Ocular Diseases in Universities of Shandong, Eye Institute, Shandong University of Traditional Chinese Medicine, Jinan, P. R. China.,Affiliated Eye Hospital, Shandong University of Traditional Chinese Medicine, Jinan, P. R. China
| | - Hongsheng Bi
- Shandong Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Therapy of Ocular Diseases, Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Therapy of Ocular Diseases in Universities of Shandong, Eye Institute, Shandong University of Traditional Chinese Medicine, Jinan, P. R. China.,Affiliated Eye Hospital, Shandong University of Traditional Chinese Medicine, Jinan, P. R. China
| |
Collapse
|
9
|
JAK-STAT Signaling Pathway in Non-Infectious Uveitis. Biochem Pharmacol 2022; 204:115236. [PMID: 36041544 DOI: 10.1016/j.bcp.2022.115236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/21/2022] [Accepted: 08/23/2022] [Indexed: 11/22/2022]
Abstract
Non-infectious uveitis (NIU) refers to various intraocular inflammatory disorders responsible for severe visual loss. Cytokines participate in the regulation of ocular homeostasis and NIU pathological processes. Cytokine receptors transmit signals by activating Janus kinase (JAK) and signal transducer and activator of transcription (STAT) proteins. Increasing evidence from human NIU and experimental models reveals the involvement of the JAK-STAT signaling pathway in NIU pathogenesis. Several small-molecule drugs that potentially inhibit multiple cytokine-dependent pathways are under investigation for treating autoimmune diseases, implicating possible applications for NIU treatment. This review summarizes the current understanding of the diverse roles of the JAK-STAT signaling pathway in ocular homeostasis and NIU pathology, providing a rationale for targeting JAKs and STATs for NIU treatment. Moreover, available evidence for the safety and efficacy of JAK inhibitors for refractory uveitis and potential approaches for treatment optimization are discussed.
Collapse
|
10
|
Hoffmann ALC, Hauck SM, Deeg CA, Degroote RL. Pre-Activated Granulocytes from an Autoimmune Uveitis Model Show Divergent Pathway Activation Profiles upon IL8 Stimulation In Vitro. Int J Mol Sci 2022; 23:ijms23179555. [PMID: 36076947 PMCID: PMC9455241 DOI: 10.3390/ijms23179555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 08/17/2022] [Accepted: 08/18/2022] [Indexed: 11/25/2022] Open
Abstract
In the pathophysiology of autoimmune-mediated uveitis, granulocytes have emerged as possible disease mediators and were shown to be pre-activated in equine recurrent uveitis (ERU), a spontaneous disease model. We therefore used granulocytes from ERU horses to identify early molecular mechanisms involved in this dysregulated innate immune response. Primary granulocytes from healthy and ERU horses were stimulated with IL8, and cellular response was analyzed with differential proteomics, which revealed significant differences in protein abundance of 170 proteins in ERU. Subsequent ingenuity pathway analysis identified three activated canonical pathways “PKA signaling”, “PTEN signaling” and “leukocyte extravasation”. Clustered to the leukocyte extravasation pathway, we found the membrane-type GPI-anchored protease MMP25, which was increased in IL8 stimulated ERU granulocytes. These findings point to MMP25 as a possible regulator of granulocyte extravasation in uveitis and a role of this molecule in the impaired integrity of the blood-retina-barrier. In conclusion, our analyses show a clearly divergent reaction profile of pre-activated granulocytes upon IL8 stimulation and provide basic information for further in-depth studies on early granulocyte activation in non-infectious ocular diseases. This may be of interest for the development of new approaches in uveitis diagnostics and therapy. Raw data are available via ProteomeXchange with identifier PXD013648.
Collapse
Affiliation(s)
- Anne L. C. Hoffmann
- Chair of Physiology, Department of Veterinary Sciences, LMU Munich, D-82152 Martinsried, Germany
| | - Stefanie M. Hauck
- Research Unit Protein Science, Helmholtz Center Munich, German Research Center for Environmental Health, D-80939 Munich, Germany
| | - Cornelia A. Deeg
- Chair of Physiology, Department of Veterinary Sciences, LMU Munich, D-82152 Martinsried, Germany
| | - Roxane L. Degroote
- Chair of Physiology, Department of Veterinary Sciences, LMU Munich, D-82152 Martinsried, Germany
- Correspondence:
| |
Collapse
|
11
|
DeDreu J, Pal-Ghosh S, Mattapallil MJ, Caspi RR, Stepp MA, Menko AS. Uveitis-mediated immune cell invasion through the extracellular matrix of the lens capsule. FASEB J 2021; 36:e21995. [PMID: 34874579 PMCID: PMC9300120 DOI: 10.1096/fj.202101098r] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 09/20/2021] [Accepted: 10/04/2021] [Indexed: 12/05/2022]
Abstract
While the eye is considered an immune privileged site, its privilege is abrogated when immune cells are recruited from the surrounding vasculature in response to trauma, infection, aging, and autoimmune diseases like uveitis. Here, we investigate whether in uveitis immune cells become associated with the lens capsule and compromise its privilege in studies of C57BL/6J mice with experimental autoimmune uveitis. These studies show that at D14, the peak of uveitis in these mice, T cells, macrophages, and Ly6G/Ly6C+ immune cells associate with the lens basement membrane capsule, burrow into the capsule matrix, and remain integrated with the capsule as immune resolution is occurring at D26. 3D surface rendering image analytics of confocal z‐stacks and scanning electron microscopy imaging of the lens surface show the degradation of the lens capsule as these lens‐associated immune cells integrate with and invade the lens capsule, with a subset infiltrating both epithelial and fiber cell regions of lens tissue, abrogating its immune privilege. Those immune cells that remain on the surface often become entwined with a fibrillar net‐like structure. Immune cell invasion of the lens capsule in uveitis has not been described previously and may play a role in induction of lens and other eye pathologies associated with autoimmunity.
Collapse
Affiliation(s)
- JodiRae DeDreu
- Department of Pathology, Anatomy and Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Sonali Pal-Ghosh
- Department of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, USA
| | - Mary J Mattapallil
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Rachel R Caspi
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Mary Ann Stepp
- Department of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, USA.,Department of Ophthalmology, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, USA
| | - A Sue Menko
- Department of Pathology, Anatomy and Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.,Department of Ophthalmology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
12
|
McDonald T, Muhammad F, Peters K, Lee DJ. Combined Deficiency of the Melanocortin 5 Receptor and Adenosine 2A Receptor Unexpectedly Provides Resistance to Autoimmune Disease in a CD8 + T Cell-Dependent Manner. Front Immunol 2021; 12:742154. [PMID: 34867964 PMCID: PMC8634946 DOI: 10.3389/fimmu.2021.742154] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 10/28/2021] [Indexed: 12/15/2022] Open
Abstract
Regulatory immunity that provides resistance to relapse emerges during resolution of experimental autoimmune uveitis (EAU). This post-EAU regulatory immunity requires a melanocortin 5 receptor (MC5r)-dependent suppressor antigen presenting cell (APC), as shown using a MC5r single knock-out mouse. The MC5r-dependent APC activates an adenosine 2A receptor (A2Ar)-dependent regulatory Treg cell, as shown using an A2Ar single knock-out mouse. Unexpectedly, when MC5r-/- post-EAU APC were used to activate A2Ar-/- post-EAU T cells the combination of cells significantly suppressed EAU, when transferred to EAU mice. In contrast, transfer of the reciprocal activation scheme did not suppress EAU. In order to explain this finding, MC5r-/-A2Ar-/- double knock-out (DKO) mice were bred. Naïve DKO mice had no differences in the APC populations, or inflammatory T cell subsets, but did have significantly more Treg cells. When we examined the number of CD4 and CD8 T cell subsets, we found significantly fewer CD8 T cells in the DKO mice compared to WT and both single knock-out mice. DKO mice also had significantly reduced EAU severity and accelerated resolution. In order to determine if the CD8 T cell deficiency contributed to the resistance to EAU in the DKO mice, we transferred naïve CD8 T cells from WT mice, that were immunized for EAU. Susceptibility to EAU was restored in DKO mice that received a CD8 T cell transfer. While the mechanism that contributed to the CD8 T cell deficiency in the DKO mice remains to be determined, these observations indicate an importance of CD8 T cells in the initiation of EAU. The involvement of CD4 and CD8 T cells suggests that both class I and class II antigen presentation can trigger an autoimmune response, suggesting a much wider range of antigens may trigger autoimmune disease.
Collapse
Affiliation(s)
- Trisha McDonald
- Dean McGee Eye Institute, Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Fauziyya Muhammad
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Kayleigh Peters
- Dean McGee Eye Institute, Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Darren J. Lee
- Dean McGee Eye Institute, Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States,Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States,*Correspondence: Darren J. Lee,
| |
Collapse
|
13
|
Chen YH, Lightman S, Calder VL. CD4 + T-Cell Plasticity in Non-Infectious Retinal Inflammatory Disease. Int J Mol Sci 2021; 22:9584. [PMID: 34502490 PMCID: PMC8431487 DOI: 10.3390/ijms22179584] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/27/2021] [Accepted: 08/31/2021] [Indexed: 01/14/2023] Open
Abstract
Non-infectious uveitis (NIU) is a potentially sight-threatening disease. Effector CD4+ T cells, especially interferon-γ-(IFNγ) producing Th1 cells and interleukin-17-(IL-17) producing Th17 cells, are the major immunopathogenic cells, as demonstrated by adoptive transfer of disease in a model of experimental autoimmune uveitis (EAU). CD4+FoxP3+CD25+ regulatory T cells (Tregs) were known to suppress function of effector CD4+ T cells and contribute to resolution of disease. It has been recently reported that some CD4+ T-cell subsets demonstrate shared phenotypes with another CD4+ T-cell subset, offering the potential for dual function. For example, Th17/Th1 (co-expressing IFNγ and IL-17) cells and Th17/Treg (co-expressing IL-17 and FoxP3) cells have been identified in NIU and EAU. In this review, we have investigated the evidence as to whether these 'plastic CD4+ T cells' are functionally active in uveitis. We conclude that Th17/Th1 cells are generated locally, are resistant to the immunosuppressive effects of steroids, and contribute to early development of EAU. Th17/Treg cells produce IL-17, not IL-10, and act similar to Th17 cells. These cells were considered pathogenic in uveitis. Future studies are needed to better clarify their function, and in the future, these cell subsets may in need to be taken into consideration for designing treatment strategies for disease.
Collapse
Affiliation(s)
- Yi-Hsing Chen
- UCL Institute of Ophthalmology, University College London, London EC1V 9EL, UK; (Y.-H.C.); (S.L.)
- Department of Ophthalmology, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Sue Lightman
- UCL Institute of Ophthalmology, University College London, London EC1V 9EL, UK; (Y.-H.C.); (S.L.)
| | - Virginia L. Calder
- UCL Institute of Ophthalmology, University College London, London EC1V 9EL, UK; (Y.-H.C.); (S.L.)
- NIHR Biomedical Research Centre, Moorfields Eye Hospital NHS Foundation Trust, London EC1V 2PD, UK
| |
Collapse
|
14
|
Zhu L, Chen B, Su W. A Review of the Various Roles and Participation Levels of B-Cells in Non-Infectious Uveitis. Front Immunol 2021; 12:676046. [PMID: 34054864 PMCID: PMC8160461 DOI: 10.3389/fimmu.2021.676046] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 04/28/2021] [Indexed: 12/30/2022] Open
Abstract
Non-infectious uveitis is an inflammatory disorder of the eye that accounts for severe visual loss without evident infectious agents. While T cells are supposed to dominate the induction of inflammation in non-infectious uveitis, the role of B cells in the pathogenesis of this disease is obscure. Therefore, this review aimed to discuss diverse B-cell participation in different non-infectious uveitides and their roles in the pathogenesis of this disease as well as the mechanism of action of rituximab. Increasing evidence from experimental models and human non-infectious uveitis has suggested the participation of B cells in non-infectious uveitis. The participation levels vary in different uveitides. Furthermore, B cells play multiple roles in the pathogenic mechanisms. B cells produce autoantibodies, regulate T cell responses via antibody-independent functions, and constitute ectopic lymphoid structures. Regulatory B cells perform pivotal anti-inflammatory functions in non-infectious uveitis. Rituximab may work by depleting pro-inflammatory B cells and restoring the quantity and function of regulatory B cells in this disease. Identifying the levels of B-cell participation and the associated roles is beneficial for optimizing therapy. Diversified experimental model choices and emerging tools and/or methods are conducive for future studies on this topic.
Collapse
Affiliation(s)
- Lei Zhu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Binyao Chen
- 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
|
15
|
Degroote RL, Deeg CA. Immunological Insights in Equine Recurrent Uveitis. Front Immunol 2021; 11:609855. [PMID: 33488614 PMCID: PMC7821741 DOI: 10.3389/fimmu.2020.609855] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 11/30/2020] [Indexed: 12/05/2022] Open
Abstract
Horses worldwide suffer from equine recurrent uveitis (ERU), an organ-specific, immune-mediated disease with painful, remitting-relapsing inflammatory attacks alternating with periods of quiescence, which ultimately leads to blindness. In course of disease, both eyes can eventually be affected and since blind horses pose a threat to themselves and their surroundings, these animals have to be killed. Therefore, this disease is highly relevant for veterinary medicine. Additionally, ERU shows strong clinical and pathological resemblance to autoimmune uveitis in man. The exact cause for the onset of ERU is unclear to date. T cells are believed to be the main effector cells in this disease, as they overcome the blood retinal barrier to invade the eye, an organ physiologically devoid of peripheral immune cells. These cells cause severe intraocular inflammation, especially in their primary target, the retina. With every inflammatory episode, retinal degeneration increases until eyesight is completely lost. In ERU, T cells show an activated phenotype, with enhanced deformability and migration ability, which is reflected in the composition of their proteome and downstream interaction pathways even in quiescent stage of disease. Besides the dysregulation of adaptive immune cells, emerging evidence suggests that cells of the innate immune system may also directly contribute to ERU pathogenesis. As investigations in both the target organ and the periphery have rapidly evolved in recent years, giving new insights on pathogenesis-associated processes on cellular and molecular level, this review summarizes latest developments in ERU research.
Collapse
Affiliation(s)
- Roxane L Degroote
- Chair of Physiology, Department of Veterinary Sciences, LMU Munich, Munich, Germany
| | - Cornelia A Deeg
- Chair of Physiology, Department of Veterinary Sciences, LMU Munich, Munich, Germany
| |
Collapse
|
16
|
Deviant proteome profile of equine granulocytes associates to latent activation status in organ specific autoimmune disease. J Proteomics 2020; 230:103989. [PMID: 32977044 DOI: 10.1016/j.jprot.2020.103989] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 08/26/2020] [Accepted: 09/14/2020] [Indexed: 02/06/2023]
Abstract
Equine recurrent uveitis (ERU) is a spontaneous, remitting-relapsing autoimmune disease driven by the adaptive immune system. Although T cells are described as the main effector cells in pathogenesis, granulocytes have also emerged as possible disease mediators. To explore the role of these innate immune cells, we investigated the whole cell proteome of granulocytes from equine recurrent uveitis cases and healthy controls. Among the 2362 proteins identified by mass spectrometry, we found 96 proteins with significantly changed abundance between groups (p < 0.05, fold change >1.2), representing 4.1% of total granulocyte proteome. Within these differential identifications, calgranulin B, a protein associated with pathogenesis in other autoimmune diseases, showed highest abundance in equine recurrent uveitis (18 fold). For a better interpretation of the results from our hypothesis-generating approach, we added a threshold for biological significance (ratio ERU/controls >2: 36 proteins) to the proteins with increased abundance in equine recurrent uveitis and analyzed their allocation to the subsets within the Immune System superpathway. The 36 differentially abundant proteins predominantly associated to RAF/MAP kinase cascade, MHC-I-mediated antigen presentation and neutrophil degranulation, suggesting a latently activated phenotype of these innate immune cells in disease. Raw data are available via ProteomeXchange with identifier PXD013648. SIGNIFICANCE: Our study provides new insights into the protein repertoire of primary equine granulocytes and identifies protein abundance changes associated to equine recurrent uveitis (ERU), an organ specific, spontaneously occurring autoimmune disease. We show that granulocyte proteins with increased abundance in ERU strongly associate to RAF/MAP kinase signaling, MHC-I antigen presentation and neutrophil degranulation, pointing to a more activated state of these cells in ERU cases. Since cells were obtained in quiescent stage of disease, latent activation of granulocytes underlines the role of these innate immune cells in ERU. These findings are highly relevant for veterinary medicine, further establishing the importance of granulocytes in this T cell-driven autoimmune disease. Moreover, they have translational quality for autoimmune uveitis in man, due to strong similarity in disease occurrence, progression and pathogenesis.
Collapse
|
17
|
John S, Rolnick K, Wilson L, Wong S, Van Gelder RN, Pepple KL. Bioluminescence for in vivo detection of cell-type-specific inflammation in a mouse model of uveitis. Sci Rep 2020; 10:11377. [PMID: 32647297 PMCID: PMC7347586 DOI: 10.1038/s41598-020-68227-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 06/01/2020] [Indexed: 11/26/2022] Open
Abstract
This study reports the use of cell-type-specific in vivo bioluminescence to measure intraocular immune cell population dynamics during the course of inflammation in a mouse model of uveitis. Transgenic lines expressing luciferase in inflammatory cell subsets (myeloid cells, T cells, and B cells) were generated and ocular bioluminescence was measured serially for 35 days following uveitis induction. Ocular leukocyte populations were identified using flow cytometry and compared to the ocular bioluminescence profile. Acute inflammation is neutrophilic (75% of ocular CD45 + cells) which is reflected by a significant increase in ocular bioluminescence in one myeloid reporter line on day 2. By day 7, the ocular T cell population increases to 50% of CD45 + cells, leading to a significant increase in ocular bioluminescence in the T cell reporter line. While initially negligible (< 1% of CD45 + cells), the ocular B cell population increases to > 4% by day 35. This change is reflected by a significant increase in the ocular bioluminescence of the B cell reporter line starting on day 28. Our data demonstrates that cell-type-specific in vivo bioluminescence accurately detects changes in multiple intraocular immune cell populations over time in experimental uveitis. This assay could also be useful in other inflammatory disease models.
Collapse
Affiliation(s)
- Sarah John
- Department of Ophthalmology, University of Washington, Seattle, WA, 98104, USA
| | - Kevin Rolnick
- Department of Ophthalmology, University of Washington, Seattle, WA, 98104, USA
| | - Leslie Wilson
- Department of Ophthalmology, University of Washington, Seattle, WA, 98104, USA
| | - Silishia Wong
- Department of Ophthalmology, University of Washington, Seattle, WA, 98104, USA
| | - Russell N Van Gelder
- Department of Ophthalmology, University of Washington, Seattle, WA, 98104, USA.,Department of Biological Structure, University of Washington, Seattle, WA, 98195, USA.,Department of Pathology, University of Washington, Seattle, WA, 98195, USA
| | - Kathryn L Pepple
- Department of Ophthalmology, University of Washington, Seattle, WA, 98104, USA.
| |
Collapse
|
18
|
Carreño E, Serrano C, Muñoz N, Romero-Bueno F, Sánchez Pernaute O, Alejandre N. Intraocular leucocyte subpopulations analysis by multiparametric flow cytometry in human uveitis. Br J Ophthalmol 2020; 105:322-327. [PMID: 32424059 DOI: 10.1136/bjophthalmol-2019-315511] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 04/08/2020] [Accepted: 04/29/2020] [Indexed: 11/04/2022]
Abstract
PURPOSE The aim of this study was to describe the cellular infiltrate in aqueous and vitreous samples of patients with uveitis analysed by multiparametric flow cytometry. METHODS This is a retrospective analysis of aqueous and vitreous samples analysed by flow cytometry for diagnostic purposes, in cases of masquerade syndromes and infectious and non-infectious uveitis. Data collected included demographics, anatomical classification of uveitis, phenotypic diagnosis, anterior chamber cells grading, vitreous haze and time of follow-up since presentation to sample obtained. RESULTS Thirty-one samples (17 aqueous and 14 vitreous fluids) from 31 patients, 18 men, were analysed. The mean age at the time of sample collection was 60.23±17.03 years. The most frequent anatomical classification was panuveitis (14 of 31). T cells accounted for the main cellular component in the majority of the samples (10 of 13 aqueous samples; 7 of 14 in vitreous samples). CD4:CD8 ratios ranged from 0.21 to 16.3 in the case of aqueous samples and from 0.5 to 9.7 in the case of vitreous samples. DISCUSSION Flow cytometry analysis of aqueous and vitreous samples from patients with uveitis could provide insight into the pathogenesis of human uveitis and help develop accurate animal models which better mimic human disease.
Collapse
Affiliation(s)
- Ester Carreño
- Ophthalmology Department, Hospital Universitario Fundacion Jimenez Diaz, Madrid, Spain .,Rheumatology Department, Hospital Universitario Rey Juan Carlos, Mostoles, Madrid, Spain
| | - Cristina Serrano
- Clinical Immunology Department, Hospital Universitario Fundacion Jimenez Diaz, Madrid, Spain
| | - Nelida Muñoz
- Ophthalmology Department, Hospital Universitario Fundacion Jimenez Diaz, Madrid, Spain
| | | | - Olga Sánchez Pernaute
- Rheumatology Department, Hospital Universitario Rey Juan Carlos, Mostoles, Madrid, Spain
| | - Nicolas Alejandre
- Ophthalmology Department, Hospital Universitario Fundacion Jimenez Diaz, Madrid, Spain
| |
Collapse
|
19
|
Wang B, Tian Q, Guo D, Lin W, Xie X, Bi H. Activated γδ T Cells Promote Dendritic Cell Maturation and Exacerbate the Development of Experimental Autoimmune Uveitis (EAU) in Mice. Immunol Invest 2020; 50:164-183. [PMID: 31985304 DOI: 10.1080/08820139.2020.1716786] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Our previous study reveals that gamma delta (γδ) T cells were activated and dendritic cells (DCs) underwent maturation during the inflammation phase in experimental autoimmune uveitis (EAU) mice, and the interaction between DCs and γδ T cells may significantly exacerbate the development of EAU. However, the interactions between DCs and γδ T cells that can affect DCs maturation to influence EAU development must be further addressed. In this study we showed that mature DC numbers in TCR-δ-/- (KO) EAU mice were lower than those in wild-type (WT) C57BL/6 (B6) mice. The γδ T cells harvested from WT EAU mice secreted more interferon-γ (IFN-γ), however, after blocking IFN-γ, the maturation of DCs was significantly downregulated. By contrast, the percentage of IFN-γ- and IL-17-producing CD4+ T cells in KO EAU mice decreased to a greater extent than that in WT EAU mice during the inflammatory phase. Additionally, the levels of IFN-γ/IL-17 in serum were in agreement with those of CD4+ T cells. Furthermore, after activated γδ T cells injection, the inflammatory symptoms of EAU mice were more aggravated. In vitro co-cultures of both cell types showed that activated γδ T cells may induce DCs to generate higher levels of intracellular cell adhesion molecule-1 (ICAM-1/CD54), CD80, CD83, and CD86. Moreover, co-culture of the two cells may induce the activation of CD4+ T cells. Taken together, our results demonstrated that activated γδ T cells may promote DCs maturation and further enhance the generation of Th1/Th17 cells in EAU mice, resulting in exacerbated EAU.
Collapse
Affiliation(s)
- Beibei Wang
- First Clinical College, Shandong University of Traditional Chinese Medicine , Jinan, P. R. China.,Department of Ophthalmology, Affiliated Eye Hospital of Shandong University of Traditional Chinese Medicine , Jinan, P. R. China
| | - Qingmei Tian
- First Clinical College, Shandong University of Traditional Chinese Medicine , Jinan, P. R. China.,Department of Ophthalmology, Affiliated Eye Hospital of Shandong University of Traditional Chinese Medicine , Jinan, P. R. China
| | - Dadong Guo
- Shandong Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Therapy of Ocular Diseases, Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Therapy of Ocular Diseases in Universities of Shandong, Eye Institute of Shandong University of Traditional Chinese Medicine , Jinan, P. R. China
| | - Wei Lin
- Department of Microbiology, Shandong Academy of Medical Sciences , Jinan, People's Republic of China
| | - Xiaofeng Xie
- Department of Ophthalmology, Affiliated Eye Hospital of Shandong University of Traditional Chinese Medicine , Jinan, P. R. China
| | - Hongsheng Bi
- First Clinical College, Shandong University of Traditional Chinese Medicine , Jinan, P. R. China.,Department of Ophthalmology, Affiliated Eye Hospital of Shandong University of Traditional Chinese Medicine , Jinan, P. R. China.,Shandong Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Therapy of Ocular Diseases, Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Therapy of Ocular Diseases in Universities of Shandong, Eye Institute of Shandong University of Traditional Chinese Medicine , Jinan, P. R. China
| |
Collapse
|