1
|
Fu L, Pelosini L, Kopsachilis N, Foti R, D’Esposito F, Musa M, D’Amico A, Tognetto D, Gagliano C, Zeppieri M. Evaluating the efficacy of stem cells in treating severe dry eye disease. World J Stem Cells 2025; 17:101891. [DOI: 10.4252/wjsc.v17.i4.101891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 02/17/2025] [Accepted: 03/21/2025] [Indexed: 04/23/2025] Open
Abstract
Dry eye disease (DED) is a multifactorial disorder that disturbs ocular surface equilibrium, considerably diminishing quality of life. Present therapies only offer symptomatic alleviation. Stem cell treatment, especially mesenchymal stem cells (MSCs), has surfaced as a viable approach for tissue regeneration and immunological regulation in DED. Preclinical and early clinical investigations indicate that MSCs can improve lacrimal gland functionality, diminish inflammation, and facilitate corneal regeneration. Nonetheless, obstacles persist in enhancing MSC viability, determining the optimal MSC source, and guaranteeing sustained therapeutic effectiveness. Additional extensive randomized clinical trials are required to confirm the efficacy of MSC-based therapies for severe DED.
Collapse
Affiliation(s)
- Lanxing Fu
- Department of Ophthalmology, East Kent Hospitals University NHS Foundation Trust, Canterbury CT1 3NG, United Kingdom
| | - Lucia Pelosini
- Department of Ophthalmology, King’s College Hospital NHS Foundation Trust, London SE5 9RS, United Kingdom
| | - Nick Kopsachilis
- Department of Ophthalmology, East Kent Hospitals University NHS Foundation Trust, Canterbury CT1 3NG, United Kingdom
| | - Roberta Foti
- Division of Rheumatology, A.O.U. “Policlinico-San Marco,” Catania 95123, Italy
| | - Fabiana D’Esposito
- Imperial College Ophthalmic Research Group Unit, Imperial College, London NW1 5QH, United Kingdom
- Department of Neurosciences, Reproductive Sciences and Dentistry, University of Naples Federico II, Via Pansini 5, 80131 Napoli, Italy
| | - Mutali Musa
- Department of Optometry, University of Benin, Benin 300283, Nigeria
- Department of Ophthalmology, Centre for Sight Africa, Nkpor, Onitsha 434112, Nigeria
| | - Alberto D’Amico
- Academic Neurosurgery, Department of Neurosciences, University of Padova, Padova 35128, Italy
| | - Daniele Tognetto
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste 34129, Italy
| | - Caterina Gagliano
- Department of Medicine and Surgery, University of Enna “Kore,” Catania 94100, Italy
- Mediterranean Foundation "G.B. Morgagni", 95125 Catania, Italy
| | - Marco Zeppieri
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste 34129, Italy
- Department of Ophthalmology, University Hospital of Udine, Udine 33100, Italy
| |
Collapse
|
2
|
Gong B, Liu Y, Li H, Ju X, Li D, Zou Y, Guo X, Dong K, Xiao J, Wu W, Chai R, Zhang R, Yu M. A Silk Fibroin Nanoparticle Hydrogel Loaded With NK1R Antagonist Has Synergistic Anti-Inflammatory and Reparative Effects on Dry Eye Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2404835. [PMID: 39985258 PMCID: PMC12005769 DOI: 10.1002/advs.202404835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 01/28/2025] [Indexed: 02/24/2025]
Abstract
Dry eye disease (DED) is a multifactorial illness affecting tears and the ocular surface. The neurokinin 1 receptor (NK1R) is a target for controlling T helper 17 (Th17) and regulatory T cell (Treg) imbalances. This work creates a silk fibroin (SF) nanoparticle hydrogel that targets NK1R with CP-99,994 (CP). Combining CP and SF to generate stable nanoparticles while integrating a flexible hydrogel material results in a sustained-release ophthalmic drop formulation (SF@CP@Gel), which provides a long-lasting ocular formulation with anti-inflammatory and reparative properties. SF@CP@Gel could maintain a stable CP concentration for 25 h with detectable biological activity. The cell counting kit-8 and 2,7-DHL-DA results reveal that SF@CP@Gel has no cytotoxic effect on human corneal epithelial cells (HCECs) and decreases the reactive oxygen species level in oxidatively damaged HCECs. Cell scratch assays demonstrate that SF@CP@Gel can greatly increase HCEC migration and proliferation within 24 h. Furthermore, in vivo therapy with topical SF@CP@Gel twice daily markedly reduce clinical symptoms by reducing the amount of pathogenic Th17 cells while efficiently restoring Treg activity. In summary, this work reveals that SF@CP@Gel might attenuate DED by inhibiting NK1R-mediated SP signaling and thereby modulating the Th17/Treg ratio, a potential anti-inflammatory and repair treatment method for DED.
Collapse
Affiliation(s)
- Bo Gong
- Department of OphthalmologySichuan Academy of Medical Sciences & Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengdu610000China
- Human Disease Genes Key Laboratory of Sichuan Province and Institute of Laboratory MedicineSichuan Academy of Medical Sciences & Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengdu610000China
- Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026)Sichuan Academy of Medical Sciences & Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengdu610000China
| | - Yi Liu
- Department of OphthalmologyDeyang People's HospitalDeyang618000China
- School of MedicineUniversity of Electronic Science and Technology of ChinaChengdu610000China
| | - Huan Li
- Department of OphthalmologySichuan Academy of Medical Sciences & Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengdu610000China
- Department of OphthalmologyDeyang People's HospitalDeyang618000China
| | - Xueming Ju
- Human Disease Genes Key Laboratory of Sichuan Province and Institute of Laboratory MedicineSichuan Academy of Medical Sciences & Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengdu610000China
| | - Dongfeng Li
- Department of OphthalmologySichuan Academy of Medical Sciences & Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengdu610000China
| | - Yuhao Zou
- Department of OphthalmologySichuan Academy of Medical Sciences & Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengdu610000China
| | - Xiaoxin Guo
- Department of OphthalmologySichuan Academy of Medical Sciences & Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengdu610000China
- Human Disease Genes Key Laboratory of Sichuan Province and Institute of Laboratory MedicineSichuan Academy of Medical Sciences & Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengdu610000China
- Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026)Sichuan Academy of Medical Sciences & Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengdu610000China
| | - Kai Dong
- Department of OphthalmologySichuan Academy of Medical Sciences & Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengdu610000China
- Human Disease Genes Key Laboratory of Sichuan Province and Institute of Laboratory MedicineSichuan Academy of Medical Sciences & Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengdu610000China
- Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026)Sichuan Academy of Medical Sciences & Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengdu610000China
| | - Jialing Xiao
- Department of OphthalmologySichuan Academy of Medical Sciences & Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengdu610000China
- School of MedicineUniversity of Electronic Science and Technology of ChinaChengdu610000China
| | - Weijia Wu
- Department of OphthalmologySichuan Academy of Medical Sciences & Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengdu610000China
- School of MedicineUniversity of Electronic Science and Technology of ChinaChengdu610000China
| | - Renjie Chai
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology‐Head and Neck SurgeryZhongda HospitalSchool of Life Sciences and TechnologySchool of Medicine, Advanced Institute for Life and HealthJiangsu Province High‐Tech Key Laboratory for Bio‐Medical ResearchSoutheast UniversityNanjing210096China
- Co‐Innovation Center of NeuroregenerationNantong UniversityNantong226001China
- Department of NeurologyAerospace Center HospitalSchool of Life ScienceBeijing Institute of TechnologyBeijing100081China
- Department of Otolaryngology Head and Neck Surgery, Sichuan Provincial People's HospitalSchool of MedicineUniversity of Electronic Science and Technology of ChinaChengdu610072China
- Southeast University Shenzhen Research InstituteShenzhen518063China
| | - Ruifan Zhang
- Department of OphthalmologySichuan Academy of Medical Sciences & Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengdu610000China
| | - Man Yu
- Department of OphthalmologySichuan Academy of Medical Sciences & Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengdu610000China
| |
Collapse
|
3
|
Seto NA, Wong CW, Lu X, Watsky MA, Yee RW. Tolerability and Efficacy of Topical Methotrexate in Ocular Surface Disease. Cureus 2025; 17:e80958. [PMID: 40255803 PMCID: PMC12009605 DOI: 10.7759/cureus.80958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/20/2025] [Indexed: 04/22/2025] Open
Abstract
INTRODUCTION Topical immunomodulators have gained popularity in the treatment of inflammatory dry eye disease (DED). In our tertiary DED population, ocular surface inflammatory signs and symptoms refractory to conventional treatments remain a diagnostic and therapeutic challenge. This two-part in vitro toxicity and in vivo clinical study aims to determine the safety and therapeutic potential of off-label use of topical methotrexate (MTX) for the treatment of recalcitrant ocular surface disease (OSD). METHODS Aim 1 involved an in vitro toxicity assay where primary human corneal epithelial/stromal cells (HCEs and HCSCs, respectively) were established from de-identified donor corneal rims. Epithelial cells were separated and cultured in Dulbecco's modified Eagle's medium (DMEM), while stromal tissue was isolated, and the cells were sub-cultured in DMEM. HCEs and HCSCs were exposed to 1 mg/mL, 2 mg/mL, or 3 mg/mL of MTX. HCEs were also treated with 10 mg/mL of MTX. Cells were photographed at 24 and 96 hours post-treatment. Aim 2 involved MTX treatment for recalcitrant surface inflammation. A retrospective chart review was conducted on patients diagnosed with recalcitrant OSD who were treated with off-label topical 1 mg/mL MTX four times a day bilaterally. The ocular surface disease index (OSDI) and symptom assessment in dry eye (SANDE) were recorded to assess symptomatic changes. Changes in objective measurements were assessed by measuring Schirmer's test, corneal fluorescein staining (CFS), and palpebral conjunctival redness (PCR). Inferential statistical analysis was performed using paired t-test and Wilcoxon signed-rank test in STATA 16. RESULTS In vitro results for treated HCEs and HCSCs showed minimal changes relative to control across 1 mg/mL, 2 mg/mL, and 3 mg/mL MTX at 24 hours. Decreased cell survival of HCEs was noted after 96 hours of 1 mg/mL, 2 mg/mL, and 3 mg/mL MTX exposure. Cell survival of HCSCs at 96 hours in 1 mg/mL and 2 mg/mL MTX exposure was similar to control; however, at 3 mg/mL MTX exposure, decreased cell survival was noted. At 10 mg/mL MTX, loss of HCE cells was noted at 72 hours. Clinically, 19 patients were studied consecutively. Study times ranged from six to eight weeks, with a median follow-up time of two weeks between visits. Compared to before treatment, patients showed improvement in PCR (p=<0.01), OSDI (p=0.02), and CFS (p=0.03). SANDE and Schirmer's 1 were also improved but not statistically significant. Visual acuity, intraocular pressure (IOP), and visual analog scale (VAS) showed no statistical change across the treatment period. CONCLUSIONS Topical MTX demonstrated minimal cytotoxicity in vitro and appears to be well tolerated in our patient cohort. Topical MTX may play a role in the treatment of recalcitrant DED and OSD signs and symptoms, and further study with larger sample sizes is in progress.
Collapse
Affiliation(s)
- Nathan A Seto
- Ophthalmology, MD Anderson Cancer Center, Houston, USA
| | - Calvin W Wong
- Ophthalmology and Visual Science, McGovern Medical School, University of Texas Health Science Center, Houston, USA
| | - Xiaowen Lu
- Cellular Biology and Anatomy, Augusta University Medical College of Georgia, Augusta, USA
| | - Mitchell A Watsky
- Cellular Biology and Anatomy, Augusta University Medical College of Georgia, Augusta, USA
| | - Richard W Yee
- Head and Neck Surgery, MD Anderson Cancer Center, Houston, USA
| |
Collapse
|
4
|
Li Y, Luo Z, Liu Z, Zhu X, Reinach PS, Li L, Chen W. IL-23 Promotes γδT Cell Activity in Dry Eye Disease Progression. Invest Ophthalmol Vis Sci 2025; 66:10. [PMID: 39903182 PMCID: PMC11801388 DOI: 10.1167/iovs.66.2.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 01/10/2025] [Indexed: 02/06/2025] Open
Abstract
Purpose Conjunctival-resident γδT cells, the predominant ocular source of interleukin-17A (IL-17A), play crucial roles in dry eye disease (DED) pathogenesis. The upstream regulators of these cells are unknown. This study evaluated the role of conjunctival IL-23 expression in mediating γδT cell generation and elucidated its contribution to dry eye inflammatory responses. Methods Single-cell RNA sequencing (scRNA-seq) was used to identify and quantify conjunctival mRNA molecules in γδT cells in mice. The IL-23 level increased in wild-type (WT) and decreased in γδT-deficient (TCRδ-/-) mice after dry eye was induced via an intelligently controlled environmental system (ICES). Flow cytometry and transcriptome sequencing were used to investigate the impact of the changes in IL-23 expression on human γδT cells. Results The expression of the IL-23 receptor (IL-23R) was greater in γδT cells than in other conjunctival cell types, such as CD4+ T cells, CD8+ T cells and epithelial cells. An increase in IL-23 led to an increase in γδT cell density, which was proportional to dry eye severity. However, in the TCRδ-/- mice, the upregulation of IL-23 failed to increase the expression level of IL-17A and the severity of dry eye. Furthermore, increases in the expression of IL-23 and the number of γδT cells were evident in the ocular surface cells of patients who developed visual display terminal syndrome. Conclusions An increase in conjunctival IL-23 expression contributes to the induction of the DED inflammatory response through interactions with its cognate receptor on γδT cells and the promotion of their proliferation. The findings of this study suggest that the suppression of IL-17A through the blockade of IL-23R activation may be a viable target for improving the management of inflammation in DED patients.
Collapse
Affiliation(s)
- Yanxiao Li
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Zan Luo
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Zihao Liu
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Xinhao Zhu
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Peter S. Reinach
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Ling Li
- Ningbo Eye Institute, Ningbo Eye Hospital, Wenzhou Medical University, Ningbo, China
| | - Wei Chen
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- Ningbo Eye Institute, Ningbo Eye Hospital, Wenzhou Medical University, Ningbo, China
| |
Collapse
|
5
|
Fan NW, Yu M, Wang S, Blanco T, Luznik Z, Chauhan SK, Viswanath V, Gil D, Held K, Chen Y, Dana R. Activation of α2B/2C adrenergic receptor ameliorates ocular surface inflammation through enhancing regulatory T cell function. Mucosal Immunol 2025; 18:176-187. [PMID: 39522611 DOI: 10.1016/j.mucimm.2024.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 11/01/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
There is an unmet need for effectively treating dry eye disease (DED), a T cell-mediated chronic, inflammatory ocular surface disorder. Given the potential of nonneuronal adrenergic system in modulating T cell response, we herein investigated the therapeutic efficacy and the underlying mechanisms of a specific alpha 2 adrenergic receptor agonist (AGN-762, selective for α2B/2C receptor subtypes) in a mouse model of DED. Experimental DED was treated with the AGN-762 by oral gavage, either at disease induction or after disease establishment, and showed sustained amelioration, along with reduced expression of DED-pathogenic cytokines in ocular surface tissues, decreased corneal MHC-II+CD11b+ cells and lymphoid Th17 cells, and higher function of regulatory T cells (Treg). In vitro culture of DED-derived effector T helper cells (Teff) with AGN-762 failed to suppress Th17 response, while culture of DED-Treg with AGN-762 led to enhanced suppressive function of Treg and their IL-10 production. Adoptive transfer of AGN-762-pretreated DED-Treg in syngeneic B6.Rag1-/- mice effectively suppressed DED Teff-mediated disease and Th17 response, and the effect was abolished by the neutralization of IL-10. In conclusion, our findings demonstrate that α2B/2C adrenergic receptor agonism effectively ameliorates persistent corneal epitheliopathy in DED by enhancing IL-10 production from Treg and thus restoring their immunoregulatory function.
Collapse
Affiliation(s)
- Nai-Wen Fan
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA; Department of Ophthalmology, Taipei Veterans General Hospital, Taiwan; School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Man Yu
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA; Department of Ophthalmology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, PR China
| | - Shudan Wang
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Tomas Blanco
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Zala Luznik
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Sunil K Chauhan
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Veena Viswanath
- (Former) Development Sciences, AbbVie Inc., Irvine, CA 92612, USA
| | - Daniel Gil
- (Former) Biological Research, Allergan plc, Irvine, CA 92612, USA
| | - Katherine Held
- Ophthalmology Discovery Research, AbbVie Inc., Irvine, CA 92612, USA
| | - Yihe Chen
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA.
| | - Reza Dana
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
6
|
Huang SC, Lei YP, Hsiao MC, Hsieh YK, Tang QP, Chen C, Hsu MY. Multicomponent Dietary Supplementation: Impact on Tear Secretion and Ocular Surface Inflammation in Dry Eye Syndrome Patients. Antioxidants (Basel) 2025; 14:103. [PMID: 39857437 PMCID: PMC11760835 DOI: 10.3390/antiox14010103] [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: 10/20/2024] [Revised: 01/05/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
Dry eye syndrome (DES) is a prevalent ocular condition characterized by tear film instability, inflammation, and discomfort, affecting millions worldwide. DES is related to oxidative stress imbalance and ocular surface inflammation, which are important factors in the development of the condition. Recent studies have demonstrated that fish oil, lutein, and zeaxanthin possess anti-inflammatory and antioxidant properties. This study investigated the efficacy of a multicomponent dietary supplement in improving tear secretion and mitigating ocular surface inflammation in patients with DES. It was an open-label intervention trial. In total, 52 participants were randomly assigned to control (n = 23) and supplement (45 mg/day eicosapentaenoic acid, 30 mg/day docosahexaenoic acid, 30 mg/day lutein, and 1.8 mg/day zeaxanthin; n = 29) groups for 12 weeks. The participants were evaluated using Schirmer's test and the ocular surface disease index (OSDI) as ocular surface parameters. Moreover, blood or tear oxidative stress, antioxidant capacities, and tear inflammatory indicators were measured at weeks 0 and 12. The results indicated a significant increase in tear secretion and a significant reduction in OSDI scores in the supplement group. Additionally, inflammatory markers, such as interleukin (IL)-6 and IL-8, significantly decreased after the intervention. However, the OSDI of the supplement group significantly improved by 6.60 points (β = -6.60, p = 0.01). These findings support the potential of targeted nutritional supplementation as a safe and effective strategy for alleviating DES symptoms, offering an alternative to conventional treatments that exclusively focus on symptom management. This study highlights the role of specific nutrients in modulating tear production and inflammation, thereby providing a foundation for dietary approaches to DES treatment. Future research should explore the long-term benefits of such interventions and their impact on overall ocular health.
Collapse
Affiliation(s)
- Shih-Chien Huang
- Department of Nutrition, Chung Shan Medical University, Taichung City 402, Taiwan;
- Department of Nutrition, Chung Shan Medical University Hospital, Taichung City 402, Taiwan
| | - Yen-Ping Lei
- Department of Nursing, National Yang Ming Chiao Tung University, Hsinchu City 300, Taiwan;
| | - Min-Chien Hsiao
- Department of Medical Education, Changhua Christian Hospital, Changhua City 500, Taiwan;
| | - Yu-Kai Hsieh
- School of Medicine, Taipei Medical University, Taipei City 110, Taiwan;
| | - Quei-Ping Tang
- Department of Nutrition, Wei Gong Memorial Hospital, Toufen City 351, Taiwan;
| | - Connie Chen
- Department of Ophthalmology, Chung Shan Medical University Hospital, Taichung City 402, Taiwan;
- Department of Optometry, Chung Shan Medical University, Taichung City 402, Taiwan
- Institute of Optometry, Chung Shan Medical University, Taichung City 402, Taiwan
| | - Min-Yen Hsu
- Department of Ophthalmology, Chung Shan Medical University Hospital, Taichung City 402, Taiwan;
- School of Medicine, Chung Shan Medical University, Taichung City 402, Taiwan
| |
Collapse
|
7
|
Li J, Liu Y, Liu Z, Li X. Tear cytokine levels in Sjogren's syndrome-related dry eye disease compared with non-Sjogren's syndrome-related dry eye disease patients: A meta-analysis. Medicine (Baltimore) 2024; 103:e40669. [PMID: 39654248 PMCID: PMC11631033 DOI: 10.1097/md.0000000000040669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/01/2024] [Accepted: 11/06/2024] [Indexed: 12/12/2024] Open
Abstract
BACKGROUND As a common complication of Sjogren syndrome (SS), SS-related dry eye disease (SS-DED) significantly affects the patients' quality of life. Pro-inflammatory cytokines in tears are widely believed to play a crucial role in the pathogenesis of SS-DED. A systematic literature review with meta-analyses was conducted to provide a quantitative summary of tear cytokine levels in SS-DED compared with non-SS-DED and healthy controls. METHODS The PubMed, Embase, Web of Science, Ovid, and Scopus databases were searched until June 2022. Original case-control studies investigating tear cytokines in SS-DED patients compared with non-SS-DED or healthy individuals were included. Differences of cytokines levels were compared with random-effects standardized mean differences ± 95% confidence intervals calculated as the effect size. RESULTS A total of 15 articles, 809 subjects (302 for SS-DED, 220 for non-SS-DED, and 287 for healthy controls) were included in the study. SS-DED patients had higher tear levels of interferon-gamma, interleukin (IL)-17, IL-1β, IL-2, IL-4, IL-6, and IL-8 as compared to healthy controls. As for comparison between SS-DED and non-SS-DED group, the levels of IL-12p70, IL-17, IL-4, IL-6, IL-8, and tumor necrosis factor-alpha were significantly higher in SS-DED patients compared with the non-SS-DED group. The level of epidermal growth factor was significantly lower in SS-DED patients compared with both non-SS-DED patients and healthy controls. CONCLUSION The findings from this study provide evidence for levels of tear cytokines in SS-DED to become potential diagnostic biomarkers or therapeutic targets. Further studies with a higher number of subjects and improved quality are necessary.
Collapse
Affiliation(s)
- Jiaxi Li
- Department of Ophthalmology, Peking University Third Hospital, Beijing, China
| | - Yihe Liu
- Department of Ophthalmology, Peking University Third Hospital, Beijing, China
| | - Ziyuan Liu
- Department of Ophthalmology, Peking University Third Hospital, Beijing, China
| | - Xuemin Li
- Department of Ophthalmology, Peking University Third Hospital, Beijing, China
| |
Collapse
|
8
|
Wang S, Naderi A, Kahale F, Ortiz G, Forouzanfar K, Chen Y, Dana R. Substance P regulates memory Th17 cell generation and maintenance in chronic dry eye disease. J Leukoc Biol 2024; 116:1446-1453. [PMID: 38916986 PMCID: PMC11599119 DOI: 10.1093/jleuko/qiae142] [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: 01/03/2024] [Revised: 05/13/2024] [Accepted: 05/24/2024] [Indexed: 06/27/2024] Open
Abstract
Substance P is a neuropeptide expressed by nerves and an array of cells that serves as a critical mediator of neuroinflammation. Our recent work has demonstrated that blocking the preferred receptor for substance P, neurokinin 1 receptor, effectively suppresses the induction of acute dry eye disease by preserving regulatory T-cell function, while inhibiting antigen-presenting cell maturation and subsequent generation of effector Th17 cells. Clinically, dry eye disease is a chronic disorder characterized by sustained ocular surface inflammation, which is mediated by long-lived memory Th17 cells demonstrated in our well-established chronic dry eye disease model. The present study aimed to further understand the function of substance P in the chronic phase of dry eye disease and its role in regulating the underlying pathogenic memory Th17. In vitro culture of effector T cells isolated from acute dry eye disease with substance P led to an enhanced conversion of effector Th17 to memory Th17, while culturing memory T cells isolated from chronic dry eye disease with substance P effectively preserved the memory Th17 cells. In contrast, the addition of a neurokinin 1 receptor antagonist in the cultures abolished the substance P-mediated effects. Furthermore, in vivo treatment with the neurokinin 1 receptor antagonist during the resolution phase of acute dry eye disease significantly suppressed memory Th17 generation, and treatment in the chronic phase of dry eye disease disrupted the maintenance of memory Th17. Taken together, our results demonstrate that increased expression of substance P promotes memory Th17 generation and maintenance in chronic dry eye disease, and thus blockade of substance P represents a novel promising memory Th17-targeting strategy in treating chronic ocular surface inflammation.
Collapse
Affiliation(s)
- Shudan Wang
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, 20 Staniford Street, Boston, MA 02114, United States
- Department of Ophthalmology, The First Affiliated Hospital of Harbin Medical University, 143 Yiman Street, Harbin, Heilongjiang Province, 150001, P.R. China
| | - Amirreza Naderi
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, 20 Staniford Street, Boston, MA 02114, United States
| | - Francesca Kahale
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, 20 Staniford Street, Boston, MA 02114, United States
| | - Gustavo Ortiz
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, 20 Staniford Street, Boston, MA 02114, United States
| | - Katayoon Forouzanfar
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, 20 Staniford Street, Boston, MA 02114, United States
| | - Yihe Chen
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, 20 Staniford Street, Boston, MA 02114, United States
| | - Reza Dana
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, 20 Staniford Street, Boston, MA 02114, United States
| |
Collapse
|
9
|
Ortiz G, Blanco T, Singh RB, Kahale F, Wang S, Chen Y, Dana R. IL-6 induces Treg dysfunction in desiccating stress-induced dry eye disease. Exp Eye Res 2024; 246:110006. [PMID: 39009059 PMCID: PMC11332651 DOI: 10.1016/j.exer.2024.110006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 06/03/2024] [Accepted: 07/12/2024] [Indexed: 07/17/2024]
Abstract
Regulatory T cells (Tregs) play a critical role in maintaining immune homeostasis, and their dysfunction is implicated in the pathogenesis of various autoimmune disorders, including dry eye disease (DED). Treg dysfunction in DED allows T-helper cell 17 (Th17) mediated chronic inflammation at the ocular surface. In this study, the factors causing Treg dysfunction in DED were investigated. We observed reduced expression of Treg functional markers - FoxP3, CD25, and CTLA-4 in the cells isolated from DED mice (DED Tregs). Additionally, DED Tregs showed increased expression levels of receptors for pro-inflammatory cytokine receptors, namely IL-6R, IL-17RA, and IL-23R. An increased expression level of pro-inflammatory cytokine receptors was observed on exposing Tregs isolated from naïve mice (NTregs) to IL-6 or IL-17, but not IL-23, with a concomitant downregulation of FoxP3, CD25, and CTLA-4 in these cells. Furthermore, among these cytokines, IL-6 induced the most pronounced loss of Treg mediated suppression of Th17 proliferation and IL-10 secretion. In vitro and in vivo blockade of IL-6 effectively restored function in DED Tregs, leading to enhanced suppressive function against proliferating Th17 cells and ameliorating disease severity. In conclusion, this study provides insights into mechanisms of Treg dysregulation in DED, specifically delineating the effect of Th17-associated cytokines, with IL-6 emerging as the critical factor inducing Treg dysfunctionality. These findings highlight the potential for developing novel therapeutic interventions for DED through restoration of immunosuppressive function of Tregs.
Collapse
Affiliation(s)
- Gustavo Ortiz
- Laboratory of Ocular Immunology, Transplantation, and Regeneration, Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Tomas Blanco
- Laboratory of Ocular Immunology, Transplantation, and Regeneration, Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Rohan Bir Singh
- Laboratory of Ocular Immunology, Transplantation, and Regeneration, Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Francesca Kahale
- Laboratory of Ocular Immunology, Transplantation, and Regeneration, Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Shudan Wang
- Laboratory of Ocular Immunology, Transplantation, and Regeneration, Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Yihe Chen
- Laboratory of Ocular Immunology, Transplantation, and Regeneration, Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Reza Dana
- Laboratory of Ocular Immunology, Transplantation, and Regeneration, Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
10
|
Ganugula R, Babalola KT, Heyns IM, Arora M, Agarwal SK, Mohan C, Kumar MNVR. Lymph node targeting of cyclosporine ameliorates ocular manifestations in a mouse model of systemic lupus erythematosus (SLE) via PD-L1. NANO TODAY 2024; 57:102359. [PMID: 38911970 PMCID: PMC11192230 DOI: 10.1016/j.nantod.2024.102359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/25/2024]
Abstract
One-third of systemic lupus erythematosus (SLE) patients experience various degrees of ocular manifestations, with immunosuppressants recommended as a treatment option. Targeted immune suppression via oral administration is challenging due to the harsh gastrointestinal tract environment combined with complex physiological barriers. Here, we report the efficacy of orally administered cyclosporine (CsA)-laden polymer nanoparticles decorated with the ligand - Gambogic Acid (P2Ns-GA-CsA) in sustained lymph node delivery. This is the first report demonstrating the CD71 specificity of P2Ns-GA-CsA in the CD71 knockout mouse model and the influence of spacer length in achieving target tissue bioavailability in a lupus mouse model. P2Ns-GA-CsA effectively regulates T-cell chemotaxis by PD-L1 at a 50 % lower dose compared to conventional CsA in a mouse model exhibiting lupus-associated corneal inflammation. Collectively, these results suggest the possibility for further development of P2Ns-GA to target a diverse range of lymphatic disorders.
Collapse
Affiliation(s)
- Raghu Ganugula
- The Center for Convergent Bioscience and Medicine (CCBM), The University of Alabama, Tuscaloosa, AL, USA
- Department of Translational Science and Medicine, College of Community Health Sciences, The University of Alabama, Tuscaloosa, AL, USA
- Alabama Life Research Institute, The University of Alabama, Tuscaloosa, AL, USA
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL, USA
| | - Kabirat T. Babalola
- The Center for Convergent Bioscience and Medicine (CCBM), The University of Alabama, Tuscaloosa, AL, USA
- Department of Translational Science and Medicine, College of Community Health Sciences, The University of Alabama, Tuscaloosa, AL, USA
- Alabama Life Research Institute, The University of Alabama, Tuscaloosa, AL, USA
| | - Ingrid M. Heyns
- The Center for Convergent Bioscience and Medicine (CCBM), The University of Alabama, Tuscaloosa, AL, USA
- Department of Translational Science and Medicine, College of Community Health Sciences, The University of Alabama, Tuscaloosa, AL, USA
- Alabama Life Research Institute, The University of Alabama, Tuscaloosa, AL, USA
| | - Meenakshi Arora
- The Center for Convergent Bioscience and Medicine (CCBM), The University of Alabama, Tuscaloosa, AL, USA
- Department of Translational Science and Medicine, College of Community Health Sciences, The University of Alabama, Tuscaloosa, AL, USA
- Alabama Life Research Institute, The University of Alabama, Tuscaloosa, AL, USA
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL, USA
| | - Sandeep. K. Agarwal
- Section of Immunology, Allergy and Rheumatology, Department of Medicine, Biology of Inflammation Baylor College of Medicine, One Baylor Plaza, Houston, TX, USA
| | - Chandra Mohan
- Department of Biomedical Engineering, University of Houston, Houston, TX, USA
| | - M. N. V. Ravi Kumar
- The Center for Convergent Bioscience and Medicine (CCBM), The University of Alabama, Tuscaloosa, AL, USA
- Department of Translational Science and Medicine, College of Community Health Sciences, The University of Alabama, Tuscaloosa, AL, USA
- Alabama Life Research Institute, The University of Alabama, Tuscaloosa, AL, USA
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL, USA
- Chemical and Biological Engineering, University of Alabama, Tuscaloosa, AL, USA
- Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, USA
- Nephrology Research and Training Center, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
11
|
Song J, Dong H, Wang T, Yu H, Yu J, Ma S, Song X, Sun Q, Xu Y, Liu M. What is the impact of microbiota on dry eye: a literature review of the gut-eye axis. BMC Ophthalmol 2024; 24:262. [PMID: 38898418 PMCID: PMC11186098 DOI: 10.1186/s12886-024-03526-2] [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: 02/29/2024] [Accepted: 06/14/2024] [Indexed: 06/21/2024] Open
Abstract
BACKGROUND Dry eye is a chronic and multifactorial ocular surface disease caused by tear film instability or imbalance in the microenvironment of the ocular surface. It can lead to various discomforts such as inflammation of the ocular surface and visual issues. However, the mechanism of dry eye is not clear, which results in dry eye being only relieved but not cured in clinical practice. Finding multiple environmental pathways for dry eye and exploring the pathogenesis of dry eye have become the focus of research. Studies have found that changes in microbiota may be related to the occurrence and development of dry eye disease. METHODS Entered the keywords "Dry eye", "Microbiota", "Bacteria" through PUBMED, summarised the articles that meet the inclusion criteria and then filtered them while the publication time range of the literature was defined in the past 5 years, with a deadline of 2023.A total of 13 clinical and 1 animal-related research articles were screened out and included in the summary. RESULTS Study found that different components of bacteria can induce ocular immune responses through different receptors present on the ocular surface, thereby leading to an imbalance in the ocular surface microenvironment. Changes in the ocular surface microbiota and gut microbiota were also found when dry eye syndrome occurs, including changes in diversity, an increase in pro-inflammatory bacteria, and a decrease in short-chain fatty acid-related bacterial genera that produce anti-inflammatory effects. Fecal microbiota transplantation or probiotic intervention can alleviate signs of inflammation on the ocular surface of dry eye animal models. CONCLUSIONS By summarizing the changes in the ocular surface and intestinal microbiota when dry eye occurs, it is speculated and concluded that the intestine may affect the occurrence of eye diseases such as dry eye through several pathways and mechanisms, such as the occurrence of abnormal immune responses, microbiota metabolites- intervention of short-chain fatty acids, imbalance of pro-inflammatory and anti-inflammatory factors, and release of neurotransmitters, etc. Analyzing the correlation between the intestinal tract and the eyes from the perspective of microbiota can provide a theoretical basis and a new idea for relieving dry eyes in multiple ways in the future.
Collapse
Affiliation(s)
- Jiaping Song
- Department of Clinical Medical Laboratory, The Third People's Hospital of Dalian, No. 40, Qianshan Road, Ganjingzi District, Dalian City, Liaoning Province, 116033, China
| | - He Dong
- Department of Ophthalmology, The Third People's Hospital of Dalian, Dalian, Liaoning, 116033, China
| | - Tingting Wang
- Department of Clinical Medical Laboratory, The Third People's Hospital of Dalian, No. 40, Qianshan Road, Ganjingzi District, Dalian City, Liaoning Province, 116033, China
| | - He Yu
- Department of Ophthalmology, The Third People's Hospital of Dalian, Dalian, Liaoning, 116033, China
| | - Jian Yu
- Department of Clinical Medical Laboratory, The Third People's Hospital of Dalian, No. 40, Qianshan Road, Ganjingzi District, Dalian City, Liaoning Province, 116033, China
| | - Shaokang Ma
- Department of Clinical Medical Laboratory, The Third People's Hospital of Dalian, No. 40, Qianshan Road, Ganjingzi District, Dalian City, Liaoning Province, 116033, China
| | - Xiaohai Song
- Department of Clinical Medical Laboratory, The Third People's Hospital of Dalian, No. 40, Qianshan Road, Ganjingzi District, Dalian City, Liaoning Province, 116033, China
| | - Qianhui Sun
- Department of Clinical Medical Laboratory, The Third People's Hospital of Dalian, No. 40, Qianshan Road, Ganjingzi District, Dalian City, Liaoning Province, 116033, China
| | - Yongcheng Xu
- Department of Clinical Medical Laboratory, The Third People's Hospital of Dalian, No. 40, Qianshan Road, Ganjingzi District, Dalian City, Liaoning Province, 116033, China.
| | - Mingkai Liu
- Department of Clinical Laboratory, Dalian University Affiliated Xinhua Hospital, Dalian, Liaoning, 116021, China.
| |
Collapse
|
12
|
Fang X, Lan G, Lin Y, Xie Z, Zhong Y, Luo S, Xiao X, Luo L, Zhang Y, Li H, Wu H. Inflammation due to ocular surface homeostasis imbalance caused by pterygia: tear lymphotoxin-alpha study and a literature review. J Ophthalmic Inflamm Infect 2024; 14:28. [PMID: 38874736 PMCID: PMC11178703 DOI: 10.1186/s12348-024-00413-1] [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: 03/10/2024] [Accepted: 06/10/2024] [Indexed: 06/15/2024] Open
Abstract
OBJECTIVE To estimate the pterygium ocular surface state, and compare with healthy eyes and dry eyes. To investigate the inflammation due to pterygia growth by tear Lymphotoxin-alpha (LT α) test. DESIGN Prospective, single-center study. PARTICIPANTS 400 patients, divided into 100 pterygium group, 100 mild dry eye group, 100 moderate dry eye group, and 100 age-and sex-matched normal controls. METHODS The non-invasive break-up time (NIBUT), tear meniscus height (TMH) test, corneal fluorescein staining (CFS), meibomian gland loss score (MGs), and lipid layer thickness (LLT) were evaluated in all patients. Pterygium status and ocular status in the pterygium group were collected. The tear LT α test was conducted in the pterygium patients group. RESULT Pterygium can affect the ocular surface, leading to decreased tear film stability. The TMH, NIBUT, CFS, MGs, and lipid layer thickness can provide insights into this phenomenon. The presence of pterygium can change the structure and condition of the ocular surface. Tear LT α testing shows an abnormal decrease in LT α levels in pterygium patients. This indicates an immune-inflammation microenvironment that causes tissue repair deficiency. CONCLUSION The dry eye triggered by the growth of pterygium may originate from the tear film instability due to pterygia. As an inflammatory index, LT α in the development of pterygium and the aggravation of dry eye patients can indicate that the ocular surface is in different inflammatory states. Future tear testing in LT α may be a potential indicator to assess the inflammatory status of the dry eye.
Collapse
Affiliation(s)
- Xie Fang
- Xiamen Eye Center and Eye Institute of Xiamen University, Xiamen, China
- Xiamen Clinical Research Center for Eye Diseases, Xiamen, Fujian, China
- Xiamen Key Laboratory of Ophthalmology, Xiamen, Fujian, China
- Fujian Key Laboratory of Corneal & Ocular Surface Diseases, Xiamen, Fujian, China
- Xiamen Key Laboratory of Corneal & Ocular Surface Diseases, Xiamen, Fujian, China
- Translational Medicine Institute of Xiamen Eye Center of Xiamen University, Xiamen, Fujian, China
| | - Guoli Lan
- Xiamen Eye Center and Eye Institute of Xiamen University, Xiamen, China
- Xiamen Clinical Research Center for Eye Diseases, Xiamen, Fujian, China
- Xiamen Key Laboratory of Ophthalmology, Xiamen, Fujian, China
- Fujian Key Laboratory of Corneal & Ocular Surface Diseases, Xiamen, Fujian, China
- Xiamen Key Laboratory of Corneal & Ocular Surface Diseases, Xiamen, Fujian, China
- Translational Medicine Institute of Xiamen Eye Center of Xiamen University, Xiamen, Fujian, China
| | - Yuan Lin
- Xiamen Eye Center and Eye Institute of Xiamen University, Xiamen, China.
- Xiamen Clinical Research Center for Eye Diseases, Xiamen, Fujian, China.
- Xiamen Key Laboratory of Ophthalmology, Xiamen, Fujian, China.
- Fujian Key Laboratory of Corneal & Ocular Surface Diseases, Xiamen, Fujian, China.
- Xiamen Key Laboratory of Corneal & Ocular Surface Diseases, Xiamen, Fujian, China.
- Translational Medicine Institute of Xiamen Eye Center of Xiamen University, Xiamen, Fujian, China.
| | - Zhiwen Xie
- Xiamen Eye Center and Eye Institute of Xiamen University, Xiamen, China
- Xiamen Clinical Research Center for Eye Diseases, Xiamen, Fujian, China
- Xiamen Key Laboratory of Ophthalmology, Xiamen, Fujian, China
- Fujian Key Laboratory of Corneal & Ocular Surface Diseases, Xiamen, Fujian, China
- Xiamen Key Laboratory of Corneal & Ocular Surface Diseases, Xiamen, Fujian, China
- Translational Medicine Institute of Xiamen Eye Center of Xiamen University, Xiamen, Fujian, China
| | - Yanlin Zhong
- Xiamen Eye Center and Eye Institute of Xiamen University, Xiamen, China
- Xiamen Clinical Research Center for Eye Diseases, Xiamen, Fujian, China
- Xiamen Key Laboratory of Ophthalmology, Xiamen, Fujian, China
- Fujian Key Laboratory of Corneal & Ocular Surface Diseases, Xiamen, Fujian, China
- Xiamen Key Laboratory of Corneal & Ocular Surface Diseases, Xiamen, Fujian, China
- Translational Medicine Institute of Xiamen Eye Center of Xiamen University, Xiamen, Fujian, China
| | - Shunrong Luo
- Xiamen Eye Center and Eye Institute of Xiamen University, Xiamen, China
- Xiamen Clinical Research Center for Eye Diseases, Xiamen, Fujian, China
- Xiamen Key Laboratory of Ophthalmology, Xiamen, Fujian, China
- Fujian Key Laboratory of Corneal & Ocular Surface Diseases, Xiamen, Fujian, China
- Xiamen Key Laboratory of Corneal & Ocular Surface Diseases, Xiamen, Fujian, China
- Translational Medicine Institute of Xiamen Eye Center of Xiamen University, Xiamen, Fujian, China
| | - Xianwen Xiao
- Xiamen Eye Center and Eye Institute of Xiamen University, Xiamen, China
- Xiamen Clinical Research Center for Eye Diseases, Xiamen, Fujian, China
- Xiamen Key Laboratory of Ophthalmology, Xiamen, Fujian, China
- Fujian Key Laboratory of Corneal & Ocular Surface Diseases, Xiamen, Fujian, China
- Xiamen Key Laboratory of Corneal & Ocular Surface Diseases, Xiamen, Fujian, China
- Translational Medicine Institute of Xiamen Eye Center of Xiamen University, Xiamen, Fujian, China
| | - Lianghuan Luo
- Xiamen Eye Center and Eye Institute of Xiamen University, Xiamen, China
- Xiamen Clinical Research Center for Eye Diseases, Xiamen, Fujian, China
- Xiamen Key Laboratory of Ophthalmology, Xiamen, Fujian, China
- Fujian Key Laboratory of Corneal & Ocular Surface Diseases, Xiamen, Fujian, China
- Xiamen Key Laboratory of Corneal & Ocular Surface Diseases, Xiamen, Fujian, China
- Translational Medicine Institute of Xiamen Eye Center of Xiamen University, Xiamen, Fujian, China
| | - Yiqiu Zhang
- Xiamen Eye Center and Eye Institute of Xiamen University, Xiamen, China
- Xiamen Clinical Research Center for Eye Diseases, Xiamen, Fujian, China
- Xiamen Key Laboratory of Ophthalmology, Xiamen, Fujian, China
- Fujian Key Laboratory of Corneal & Ocular Surface Diseases, Xiamen, Fujian, China
- Xiamen Key Laboratory of Corneal & Ocular Surface Diseases, Xiamen, Fujian, China
- Translational Medicine Institute of Xiamen Eye Center of Xiamen University, Xiamen, Fujian, China
| | - Hanqiao Li
- Xiamen Eye Center and Eye Institute of Xiamen University, Xiamen, China
- Xiamen Clinical Research Center for Eye Diseases, Xiamen, Fujian, China
- Xiamen Key Laboratory of Ophthalmology, Xiamen, Fujian, China
- Fujian Key Laboratory of Corneal & Ocular Surface Diseases, Xiamen, Fujian, China
- Xiamen Key Laboratory of Corneal & Ocular Surface Diseases, Xiamen, Fujian, China
- Translational Medicine Institute of Xiamen Eye Center of Xiamen University, Xiamen, Fujian, China
| | - Huping Wu
- Xiamen Eye Center and Eye Institute of Xiamen University, Xiamen, China.
- Xiamen Clinical Research Center for Eye Diseases, Xiamen, Fujian, China.
- Xiamen Key Laboratory of Ophthalmology, Xiamen, Fujian, China.
- Fujian Key Laboratory of Corneal & Ocular Surface Diseases, Xiamen, Fujian, China.
- Xiamen Key Laboratory of Corneal & Ocular Surface Diseases, Xiamen, Fujian, China.
- Translational Medicine Institute of Xiamen Eye Center of Xiamen University, Xiamen, Fujian, China.
| |
Collapse
|
13
|
Surico PL, Lee S, Singh RB, Naderi A, Bhullar S, Blanco T, Chen Y, Dana R. Local administration of myeloid-derived suppressor cells prevents progression of immune-mediated dry eye disease. Exp Eye Res 2024; 242:109871. [PMID: 38527580 PMCID: PMC11055659 DOI: 10.1016/j.exer.2024.109871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 02/12/2024] [Accepted: 03/18/2024] [Indexed: 03/27/2024]
Abstract
Myeloid derived suppressor cells (MDSCs) are a heterogenous population of immature hematopoietic precursors with known immunoregulatory functions. The immunosuppressive role of MDSCs has been highlighted in several inflammatory ophthalmic disorders; however, their therapeutic application in suppressing the immune-mediated changes in dry eye disease (DED) has not been studied. We observed significant reduction in antigen presenting cell (APC) frequencies and their maturation in the presence of MDSCs. Moreover, co-culturing MDSCs with T helper 17 cells (Th17) resulted in reduced Th17 frequencies and their IL-17 expression. On the contrary, MDSCs maintained regulatory T cell frequencies and enhanced their function in-vitro. Furthermore, we delineated the role of interleukin-10 (IL-10) secreted by MDSCs in their immunoregulatory functions. We confirmed these results by flow cytometry analysis and observed that treatment with MDSCs in DED mice effectively suppressed the maturation of APCs, pathogenic Th17 response, and maintained Treg function and significantly ameliorated the disease. The results in this study highlight the potential therapeutic application of MDSCs in treating refractory DED.
Collapse
Affiliation(s)
- Pier Luigi Surico
- Laboratory of Ocular Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Seokjoo Lee
- Laboratory of Ocular Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Rohan Bir Singh
- Laboratory of Ocular Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Amirreza Naderi
- Laboratory of Ocular Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Shilpy Bhullar
- Laboratory of Ocular Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Tomas Blanco
- Laboratory of Ocular Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Yihe Chen
- Laboratory of Ocular Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Reza Dana
- Laboratory of Ocular Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
14
|
Surico PL, Scarabosio A, Miotti G, Grando M, Salati C, Parodi PC, Spadea L, Zeppieri M. Unlocking the versatile potential: Adipose-derived mesenchymal stem cells in ocular surface reconstruction and oculoplastics. World J Stem Cells 2024; 16:89-101. [PMID: 38455097 PMCID: PMC10915950 DOI: 10.4252/wjsc.v16.i2.89] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/06/2024] [Accepted: 01/29/2024] [Indexed: 02/26/2024] Open
Abstract
This review comprehensively explores the versatile potential of mesenchymal stem cells (MSCs) with a specific focus on adipose-derived MSCs. Ophthalmic and oculoplastic surgery, encompassing diverse procedures for ocular and periocular enhancement, demands advanced solutions for tissue restoration, functional and aesthetic refinement, and aging. Investigating immunomodulatory, regenerative, and healing capacities of MSCs, this review underscores the potential use of adipose-derived MSCs as a cost-effective alternative from bench to bedside, addressing common unmet needs in the field of reconstructive and regenerative surgery.
Collapse
Affiliation(s)
- Pier Luigi Surico
- Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA 02114, United States
- Department of Ophthalmology, Campus Bio-Medico University, Rome 00128, Italy
| | - Anna Scarabosio
- Department of Plastic Surgery, University Hospital of Udine, Udine 33100, Italy
| | - Giovanni Miotti
- Department of Plastic Surgery, University Hospital of Udine, Udine 33100, Italy
| | - Martina Grando
- Department of Internal Medicine, Azienda Sanitaria Friuli Occidentale, San Vito al Tagliamento 33078, Italy
| | - Carlo Salati
- Department of Ophthalmology, University Hospital of Udine, Udine 33100, Italy
| | - Pier Camillo Parodi
- Department of Plastic Surgery, University Hospital of Udine, Udine 33100, Italy
| | - Leopoldo Spadea
- Eye Clinic, Policlinico Umberto I, "Sapienza" University of Rome, Rome 00142, Italy
| | - Marco Zeppieri
- Department of Ophthalmology, University Hospital of Udine, Udine 33100, Italy.
| |
Collapse
|
15
|
Musa M, Chukwuyem E, Ojo OM, Topah EK, Spadea L, Salati C, Gagliano C, Zeppieri M. Unveiling Ocular Manifestations in Systemic Lupus Erythematosus. J Clin Med 2024; 13:1047. [PMID: 38398361 PMCID: PMC10889738 DOI: 10.3390/jcm13041047] [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: 12/04/2023] [Revised: 01/29/2024] [Accepted: 02/09/2024] [Indexed: 02/25/2024] Open
Abstract
Systemic Lupus Erythematosus (SLE) is a complex autoimmune disorder characterized by immune dysregulation and multi-organ involvement. In this concise brief review, we highlight key insights into Ocular Systemic Lupus Erythematosus (SLE), an intricate autoimmune disorder with diverse organ involvement. Emphasizing the formation of autoantibodies and immune complex deposition, we delve into the inflammation and damage affecting ocular structures. Clinical presentations, ranging from mild dry eye syndrome to severe conditions like retinal vasculitis, necessitate a comprehensive diagnostic approach, including clinical exams, serological testing, and imaging studies. Differential diagnosis involves distinguishing SLE-related ocular manifestations from other autoimmune and non-inflammatory ocular conditions. The multidisciplinary management approach, involving rheumatologists, ophthalmologists, and immunologists, tailors treatment based on ocular involvement severity, encompassing corticosteroids, immunosuppressive agents, and biologics. Follow-up is crucial for monitoring disease progression and treatment response. Future perspectives revolve around advancing molecular understanding, refining diagnostic tools, and exploring targeted therapies. Novel research areas include genetic factors, microbiome composition, and biotechnology for tailored and effective SLE ocular treatments.
Collapse
Affiliation(s)
- Mutali Musa
- Department of Optometry, University of Benin, Benin City 300238, Nigeria;
- Centre for Sight Africa, Onitsha 434112, Nigeria
| | | | - Oluwasola Michael Ojo
- School of Optometry and Vision Sciences, College of Health Sciences, University of Ilorin, Ilorin 240003, Nigeria
| | - Efioshiomoshi Kings Topah
- Department of Optometry, Faculty of Allied Health Sciences, College of Health Sciences, Bayero University, Kano 700006, Nigeria
| | - Leopoldo Spadea
- Eye Clinic, Policlinico Umberto I, “Sapienza” University of Rome, 00142 Rome, Italy
| | - Carlo Salati
- Department of Ophthalmology, University Hospital of Udine, 33100 Udine, Italy
| | - Caterina Gagliano
- Faculty of Medicine and Surgery, University of Enna “Kore”, Piazza Dell’Università, 94100 Enna, Italy
- Eye Clinic, Catania University San Marco Hospital, Viale Carlo Azeglio Ciampi, 95121 Catania, Italy
| | - Marco Zeppieri
- Department of Ophthalmology, University Hospital of Udine, 33100 Udine, Italy
| |
Collapse
|
16
|
Chen J, Gong Y, Sun X, Chen N, Zhao Z, Zhang W, Zheng Y. Prostaglandin E2 may clinically alleviate dry eye disease by inducing Th17 cell differentiation. Chem Biol Drug Des 2024; 103:e14477. [PMID: 38361150 DOI: 10.1111/cbdd.14477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 01/12/2024] [Accepted: 01/22/2024] [Indexed: 02/17/2024]
Abstract
Dry eye (DE) is a multifactorial ocular surface disease characterised by an imbalance in tear homeostasis. The pathogenesis of DE is complex and related to environmental, immunological (e.g., T helper 17 cells) and other factors. However, the DE disease pathogenesis remains unclear, thereby affecting its clinical treatment. This study aimed to explore the mechanism through which prostaglandin E2 (PGE2) affects DE inflammation by regulating Th17. The DE mouse model was established through subcutaneous injection of scopolamine hydrobromide. The tear secretion test and break-up time (BUT) method were used to detect tear secretion and tear film BUT, respectively. Enzyme-linked immunosorbent assay (ELISA) was used to detect the concentrations of PGE2, interleukin (IL)-17, IL-6 and tumour necrosis factor (TNF-α) in tear fluid and those of PGE2 and IL-17 in the serum. RT-qPCR and western blotting were used to test the mRNA and protein expression levels of IL-17 and retinoid-related orphan receptor-γt (RORγt). PGE2 was highly expressed in the DE mouse model. The mRNA and protein levels of IL-17 and the key Th17 transcription factor RORγt were increased in tissues of the DE mice. Moreover, PGE2 promoted tear secretion, reduced the BUT, increased the IL-17 concentration in tears and increased the Th17 cell proportion in DE, whereas the PGE2 receptor inhibitor AH6809 reversed the effects of PGE2 on tear secretion, BUT, and the Th17 cell proportion in draining lymph node (DLN) cells. Taken together, the study findings indicate that PGE2 could induce DE-related symptoms by promoting Th17 differentiation.
Collapse
Affiliation(s)
- Jingyao Chen
- Department of Ophthalmology, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, China
- Ophthalmology Department of Kunming First People's Hospital, Kunming, China
| | - Yu Gong
- Kunming Medical University, Kunming, China
| | - Xiaoyu Sun
- Kunming Medical University, Kunming, China
| | - Nuo Chen
- Kunming Medical University, Kunming, China
| | - Zijun Zhao
- Department of Ophthalmology, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, China
| | - Weijia Zhang
- Department of Ophthalmology, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, China
| | - Yixin Zheng
- Department of Ophthalmology, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, China
| |
Collapse
|
17
|
Lee KE, Oh S, Bhujel B, Kim CM, Lee H, Park JH, Kim JY. Effect of Topical Programmed Death-Ligand1 on Corneal Epithelium in Dry Eye Mouse. Biomolecules 2024; 14:68. [PMID: 38254668 PMCID: PMC10812943 DOI: 10.3390/biom14010068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 12/26/2023] [Accepted: 01/02/2024] [Indexed: 01/24/2024] Open
Abstract
Dry eye disease (DED) is a growing health concern that impacts millions of individuals every year, and is associated with corneal injury, excessive oxidative stress and inflammation. Current therapeutic strategies, including artificial tears and anti-inflammatory agents, are unable to achieve a permanent clinical cure due to their temporary nature or adverse side effects. Therefore, here, we investigated the effectiveness of the topical administration of programmed death-ligand 1 (PD-L1) in the mouse model of DED. The model was generated in C57BL/6 mice by excising the extra orbital lacrimal gland and causing desiccation stress with scopolamine injections. Subsequently, either phosphate-buffered saline (3 µL/eye) or PD-L1 (0.5 µg/mL) was topically administered for 10 days. Tear volume was evaluated with phenol red thread, and corneal fluorescein staining was observed to quantify the corneal epithelial defect. Corneas were collected for histological analysis, and the expression levels of inflammatory signaling proteins such as CD4, CD3e, IL-17, IL-1β, pIkB-α, pNF-kB and pERK1/2 were assessed through immunofluorescence and Western blot techniques. Our results demonstrate that desiccating stress-induced corneal epithelial defect and tear secretion were significantly improved by topical PD-L1 and could reduce corneal CD4+ T cell infiltration, inflammation and apoptosis in a DED mouse model by downregulating IL-17 production and ERK1/2-NFkB pathways.
Collapse
Affiliation(s)
- Ko Eun Lee
- Moon’s Eye Clinic, Suwon 16200, Republic of Korea;
- Department of Ophthalmology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Republic of Korea; (B.B.); (H.L.)
| | - Seheon Oh
- Research Institute for Biomacromolecules, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Republic of Korea; (S.O.); (C.M.K.)
- Department of Medical Science, University of Ulsan Graduate School, Seoul 05505, Republic of Korea
| | - Basanta Bhujel
- Department of Ophthalmology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Republic of Korea; (B.B.); (H.L.)
- Research Institute for Biomacromolecules, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Republic of Korea; (S.O.); (C.M.K.)
| | - Chang Min Kim
- Research Institute for Biomacromolecules, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Republic of Korea; (S.O.); (C.M.K.)
- Department of Medical Science, University of Ulsan Graduate School, Seoul 05505, Republic of Korea
| | - Hun Lee
- Department of Ophthalmology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Republic of Korea; (B.B.); (H.L.)
- Research Institute for Biomacromolecules, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Republic of Korea; (S.O.); (C.M.K.)
| | - Jin Hyoung Park
- Research Institute for Biomacromolecules, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Republic of Korea; (S.O.); (C.M.K.)
- MS Eye Clinic, Seongnam 13640, Republic of Korea
| | - Jae Yong Kim
- Department of Ophthalmology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Republic of Korea; (B.B.); (H.L.)
- Research Institute for Biomacromolecules, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Republic of Korea; (S.O.); (C.M.K.)
| |
Collapse
|
18
|
Zhou Y, Ma B, Liu Q, Duan H, Huo Y, Zhao L, Chen J, Han W, Qi H. Transmembrane Protein CMTM6 Alleviates Ocular Inflammatory Response and Improves Corneal Epithelial Barrier Function in Experimental Dry Eye. Invest Ophthalmol Vis Sci 2024; 65:4. [PMID: 38165704 PMCID: PMC10768713 DOI: 10.1167/iovs.65.1.4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 12/04/2023] [Indexed: 01/04/2024] Open
Abstract
Purpose To investigate the impact of transmembrane protein CMTM6 on the pathogenesis of dry eye disease (DED) and elucidate its potential mechanisms. Methods CMTM6 expression was confirmed by database analysis, real-time polymerase chain reaction (RT-PCR), western blot, and immunohistochemistry. Tear secretion was measured using the phenol red thread test. Immune cell infiltration was assessed through flow cytometry. Barrier function was evaluated by fluorescein sodium staining, immunofluorescence staining of zonula occludens 1 (ZO-1), and electric cell-substrate impedance sensing (ECIS) assessment. For silencing CMTM6 expression, siRNA and shRNA were employed, along with lentiviral vector-mediated overexpression of CMTM6. Proinflammatory cytokine levels were analyzed by RT-PCR and cytometric bead array (CBA) analysis. Results CMTM6 showed high expression in healthy human and mouse corneal and conjunctival epithelium but was notably reduced in DED. Notably, this downregulation was correlated with disease severity. Cmtm6-/- dry eye (DE) mice displayed reduced tear secretion, severe corneal epithelial defects, decreased conjunctival goblet cell density, and upregulated inflammatory response. Additionally, Cmtm6-/- DE mice and CMTM6 knockdown human corneal epithelial cell-transformed (HCE-T) cells showed more severe barrier disruption and reduced expression of ZO-1. Knockdown of CMTM6 in HCE-T cells increased inflammatory responses induced by hyperosmotic stress, which was significantly mitigated by CMTM6 overexpression. Moreover, the level of phospho-p65 in hyperosmolarity-stimulated HCE-T cells increased after silencing CMTM6. Nuclear factor kappa B (NF-κB) p65 inhibition (JSH-23) reversed the excessive inflammatory responses caused by hyperosmolarity in CMTM6 knockdown HCE-T cells. Conclusions The reduction in CMTM6 expression on the ocular surface contributes to the pathogenesis of DED. The CMTM6-NF-κB p65 signaling pathway may serve as a promising therapeutic target for DED.
Collapse
Affiliation(s)
- Yifan Zhou
- Department of Ophthalmology, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing, China
| | - Baikai Ma
- Department of Ophthalmology, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing, China
| | - Qiyao Liu
- Department of Immunology, Peking University Health Science Center, NHC Key Laboratory of Medical Immunology, Beijing, China
- Peking University Center for Human Disease Genomics, Beijing, China
| | - Hongyu Duan
- Department of Ophthalmology, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing, China
| | - Yangbo Huo
- Department of Immunology, Peking University Health Science Center, NHC Key Laboratory of Medical Immunology, Beijing, China
- Peking University Center for Human Disease Genomics, Beijing, China
| | - Lu Zhao
- Department of Ophthalmology, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing, China
| | - Jiawei Chen
- Institute of Medical Technology, Peking University Health Science Center, Beijing, China
| | - Wenling Han
- Department of Immunology, Peking University Health Science Center, NHC Key Laboratory of Medical Immunology, Beijing, China
- Peking University Center for Human Disease Genomics, Beijing, China
| | - Hong Qi
- Department of Ophthalmology, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing, China
- Institute of Medical Technology, Peking University Health Science Center, Beijing, China
| |
Collapse
|
19
|
Bhujel B, Oh SH, Kim CM, Yoon YJ, Chung HS, Ye EA, Lee H, Kim JY. Current Advances in Regenerative Strategies for Dry Eye Diseases: A Comprehensive Review. Bioengineering (Basel) 2023; 11:39. [PMID: 38247916 PMCID: PMC10813666 DOI: 10.3390/bioengineering11010039] [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: 11/20/2023] [Revised: 12/17/2023] [Accepted: 12/26/2023] [Indexed: 01/23/2024] Open
Abstract
Dry eye disease (DED) is an emerging health issue affecting millions of individuals annually. Ocular surface disorders, such as DED, are characterized by inflammation triggered by various factors. This condition can lead to tear deficiencies, resulting in the desiccation of the ocular surface, corneal ulceration/perforation, increased susceptibility to infections, and a higher risk of severe visual impairment and blindness. Currently, the clinical management of DED primarily relies on supportive and palliative measures, including the frequent and lifelong use of different lubricating agents. While some advancements like punctal plugs, non-steroidal anti-inflammatory drugs, and salivary gland autografts have been attempted, they have shown limited effectiveness. Recently, there have been promising developments in the treatment of DED, including biomaterials such as nano-systems, hydrogels, and contact lenses for drug delivery, cell-based therapies, biological approaches, and tissue-based regenerative therapy. This article specifically explores the different strategies reported so far for treating DED. The aim is to discuss their potential as long-term cures for DED while also considering the factors that limit their feasibility and effectiveness. These advancements offer hope for more effective and sustainable treatment options in the future.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Jae-Yong Kim
- Department of Ophthalmology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea; (B.B.); (S.-H.O.); (C.-M.K.); (Y.-J.Y.); (H.-S.C.); (E.-A.Y.); (H.L.)
| |
Collapse
|
20
|
Zhou Z, Long H, Zhou L, Xu X, Zhang R, Tu N, Liu F, Xiong J. Visual analysis of autoimmune dry eye studies based on bibliometrics. Medicine (Baltimore) 2023; 102:e36028. [PMID: 38013305 PMCID: PMC10681533 DOI: 10.1097/md.0000000000036028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 10/18/2023] [Indexed: 11/29/2023] Open
Abstract
BACKGROUND Dry eye is a chronic ocular surface disease caused by the instability of tear film or the imbalance of the ocular surface microenvironment which can lead to a diverse range of ocular discomfort symptoms. At present, the relevant mechanism of autoimmunity and treatment of dry eye is still unclear. Due to the proliferation of research papers in this field, visual analysis of existing papers can provide reference for future research. METHODS The academic papers of Web of Science were searched with the topics of "autoimmunity" and "dry eye," and the countries, institutions and keywords of the literatures selected in this domain were visualized by Citespace and Vosviewer software. RESULTS A total of 787 valid international papers were detected, and the publication count exhibited a consistent upward trend year by year. Within this field, the US has produced the highest number of papers (363), with Baylor College of Medicine being the most prolific institution (28 publications). High-producing authors in this field include Artemis P. Simopoulos and Stephen C. Pflugfelder. CONCLUSION International research in this field has focused on the pathogenesis, symptoms, and treatment of dry eye. It is predicted that the future international research hotspots will be the pathophysiology of autoimmune dry eye disease, data analysis of artificial intelligence-related diseases, and research on improving patients' quality of life.
Collapse
Affiliation(s)
- Zhenfeng Zhou
- Ophthalmology, Yueyang Hospital of Traditional Chinese Medicine, Yueyang, China
| | - Hui Long
- The First Clinical Medical School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Li Zhou
- Ophthalmology, Yueyang Hospital of Traditional Chinese Medicine, Yueyang, China
| | - Xing Xu
- Ophthalmology, Yueyang Hospital of Traditional Chinese Medicine, Yueyang, China
| | - Rong Zhang
- Ophthalmology, Yueyang Hospital of Traditional Chinese Medicine, Yueyang, China
| | - Ning Tu
- Ophthalmology, Yueyang Hospital of Traditional Chinese Medicine, Yueyang, China
| | - Fen Liu
- Ophthalmology, Yueyang Hospital of Traditional Chinese Medicine, Yueyang, China
| | - Jing Xiong
- Ophthalmology, Yueyang Hospital of Traditional Chinese Medicine, Yueyang, China
| |
Collapse
|
21
|
Kawahara A. Treatment of Dry Eye Disease (DED) in Asia: Strategies for Short Tear Film Breakup Time-Type DED. Pharmaceutics 2023; 15:2591. [PMID: 38004570 PMCID: PMC10674215 DOI: 10.3390/pharmaceutics15112591] [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: 09/20/2023] [Revised: 10/19/2023] [Accepted: 11/03/2023] [Indexed: 11/26/2023] Open
Abstract
Dry eye disease (DED) is a multifactorial disorder in which tear fluid homeostasis is lost, resulting in increased tear film osmolarity and ocular surface irritation. In Asia, the short tear film breakup time-type DED, which has become a global problem in recent years, is common. While the mainstay of DED treatment in the West is the suppression of inflammation, the first goal of treatment is the stabilization of the tear film in Asia. To date, artificial tears and steroid eye drops have been the main treatment for DED. However, artificial tears require frequent administration of eye drops and thus pose adherence problems, while steroids have problems with side-effects (cataracts, increased intraocular pressure). This review evaluates the new generation therapies in Asia based on what is known about them and demonstrates that they are more effective for DED than traditional therapies such as artificial tears and steroids. Based on considerations, it is proposed that the optimal treatment for the short tear film breakup time-type DED is the initial application of mucin-secretion-enhancing eye drops (long-acting diquafosol) and oral supplements; and if additional treatment is needed, cyclosporine eye drops and the adjunctive therapies presented in this review are added.
Collapse
Affiliation(s)
- Atsushi Kawahara
- Yoshida Eye Hospital, 2-31-8, Hondori, Hakodate 041-0851, Hokkaido, Japan
| |
Collapse
|
22
|
An Q, Zou H. Ocular surface microbiota dysbiosis contributes to the high prevalence of dry eye disease in diabetic patients. Crit Rev Microbiol 2023; 49:805-814. [PMID: 36409575 DOI: 10.1080/1040841x.2022.2142090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 10/02/2022] [Accepted: 10/26/2022] [Indexed: 11/23/2022]
Abstract
People with diabetes mellitus (DM) are at an increased risk for developing dry eye disease (DED). However, the mechanisms underlying this phenomenon remain unclear. Recent studies have found that the ocular surface microbiota (OSM) differs significantly between patients with DED and healthy people, suggesting that OSM dysbiosis may contribute to the pathogenesis of DED. This hypothesis provides a new possible explanation for why diabetic patients have a higher prevalence of DED than healthy people. The high-glucose environment and the subsequent pathological changes on the ocular surface can cause OSM dysbiosis. The unbalanced microbiota then promotes ocular surface inflammation and alters tear composition, which disturbs the homeostasis of the ocular surface. This "high glucose-OSM dysbiosis" pathway in the pathogenesis of DED with DM (DM-DED) is discussed in this review.
Collapse
Affiliation(s)
- Qingyu An
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Eye Diseases Prevention & Treatment Center, Shanghai Eye Hospital, Shanghai, China
| | - Haidong Zou
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Eye Diseases Prevention & Treatment Center, Shanghai Eye Hospital, Shanghai, China
- Shanghai Key Laboratory of Fundus Diseases, Shanghai, China
- National Clinical Research Center for Eye Diseases, Shanghai, China
- Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| |
Collapse
|
23
|
Cui T, Sun H, Hu Z, Shi Y, Zhu J, Jin M, Qin B. Optical Coherence Tomography Angiography for Evaluation of Conjunctival Vessels in Dry Eyes. J Ophthalmol 2023; 2023:1609332. [PMID: 37868692 PMCID: PMC10590264 DOI: 10.1155/2023/1609332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 08/22/2023] [Accepted: 09/15/2023] [Indexed: 10/24/2023] Open
Abstract
Objective This study aimed to evaluate conjunctival vessels in patients with dry eye disease (DED) using optical coherence tomography angiography (OCTA). Methods This was a cross-sectional, observational clinical study. Twenty-three eyes of 18 patients with DED and 28 eyes of 23 healthy controls were included for examination in this study. The evaluation included the application of an Ocular Surface Disease Index Questionnaire, Schirmer Basic Secretion Test, and anterior OCTA targeting the temporal conjunctiva. AngioTool software was used to quantify the total vessel length and vessel density in the 3 × 3 mm temporal region of interest. Results Blood vessel density measurements were compared across the OCTA systems. The total vessel length within the conjunctiva of the DED group (4799.34 ± 834.36) exceeded that of the control eye (3864.89 ± 1455.70) group (P < 0.05). However, the difference in vessel density between the two groups was not statistically significant. Conclusion Measurement and analysis of conjunctival blood vessels using OCTA exhibited robust repeatability. In dry eyes, the total number of conjunctival blood vessels increased in accordance with disease severity. Hypoxia of conjunctival tissue may be an important cause of dry eye disease.
Collapse
Affiliation(s)
- TongFeng Cui
- Department of Ophthalmology, Suqian First Hospital, Suqian, China
| | - HongYan Sun
- Department of Ophthalmology, Suqian First Hospital, Suqian, China
| | - ZiZhong Hu
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - YaBo Shi
- Department of Ophthalmology, Suqian First Hospital, Suqian, China
| | - Jiang Zhu
- Department of Ophthalmology, Suqian First Hospital, Suqian, China
| | - ManMan Jin
- Department of Ophthalmology, Suqian First Hospital, Suqian, China
| | - Bing Qin
- Department of Ophthalmology, Suqian First Hospital, Suqian, China
| |
Collapse
|
24
|
Kuklinski EJ, Yu Y, Ying GS, Asbell PA. Association of Ocular Surface Immune Cells With Dry Eye Signs and Symptoms in the Dry Eye Assessment and Management (DREAM) Study. Invest Ophthalmol Vis Sci 2023; 64:7. [PMID: 37669063 PMCID: PMC10484021 DOI: 10.1167/iovs.64.12.7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 07/27/2023] [Indexed: 09/06/2023] Open
Abstract
Purpose Dry eye disease (DED) is a multifactorial, heterogeneous disease of the ocular surface with one etiology being ocular surface inflammation. Studies using animal models demonstrate the role of ocular surface immune cells in the inflammatory pathway leading to DED, but few have evaluated humans. This study described the white blood cell population from the ocular surface of patients with DED and assessed its association with DED signs and symptoms in participants of the Dry Eye Assessment and Management (DREAM) study. Methods Participants were assessed for symptoms using the Ocular Surface Disease Index, signs via corneal staining, conjunctival staining, tear break-up time, and Schirmer test, and Sjögren's syndrome (SS) based on the 2012 American College of Rheumatology classification criteria. Impression cytology of conjunctival cells from each eye was evaluated using flow cytometry: T cells, helper T cells (Th), regulatory T cells (Tregs), cytotoxic T cells, and dendritic cells. Results We assessed 1049 eyes from 527 participants. White blood cell subtype percentages varied widely across participants. Significant positive associations were found for Th and conjunctival staining (mean score of 2.8 for 0% Th and 3.1 for >4.0% Th; P = 0.007), and corneal staining (mean score of 3.5 for 0% Th and 4.3 for >4.0% Th; P = 0.01). SS was associated with higher percent of Tregs (median 0.1 vs. 0.0; P = 0.01). Conclusions Th were associated with more severe conjunctival and corneal staining, possibly indicating their role in inflammation leading to damage of the ocular surface. There is no consistent conclusion about Tregs in SS, but these results support that Tregs are elevated in SS.
Collapse
Affiliation(s)
- Eric J. Kuklinski
- Rutgers New Jersey Medical School, Newark, New Jersey, United States
| | - Yinxi Yu
- Perelman School of Medicine at University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Gui-Shuang Ying
- Perelman School of Medicine at University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | | | - for the DREAM Study Research Group
- Rutgers New Jersey Medical School, Newark, New Jersey, United States
- Perelman School of Medicine at University of Pennsylvania, Philadelphia, Pennsylvania, United States
- University of Memphis, Memphis, Tennessee, United States
| |
Collapse
|
25
|
Zhao L, Zhang Y, Duan H, Yang T, Zhou Y, Ma B, Chen Y, Qi H. Clinical Characteristics and Tear Film Biomarkers in Patients With Chronic Dry Eye Disease After Femtosecond Laser-Assisted Laser in Situ Keratomileusis. J Refract Surg 2023; 39:556-563. [PMID: 37578178 DOI: 10.3928/1081597x-20230717-02] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
PURPOSE To investigate clinical characteristics and tear film biomarkers of patients with chronic dry eye disease (DED) following femtosecond laser-assisted laser in situ keratomileusis (FS-LASIK). METHODS Patients were divided into the chronic DED after FS-LASIK (n = 36), DED without FS-LASIK (n = 39), and normal control (without FS-LASIK; n = 34) groups. Dry eye, pain, and psychological-related symptoms were evaluated using the Ocular Surface Disease Index (OSDI), Numerical Rating Scale (NRS), Neuropathic Pain Symptom Inventory Modified for the Eye (NPSI-Eye), and Hamilton Anxiety Rating Scale (HAMA) questionnaires. Ocular surface parameters, tear cytokines, and neuropeptide concentrations were evaluated with specific tests. RESULTS The DED after FS-LASIK group showed higher corneal fluorescein staining scores, but lower OSDI and NPSI-Eye scores than the DED without FS-LASIK group (all P < .05). Corneal sensitivity and nerve density decreased in the DED after FS-LASIK group (all P < .01). Granulocyte-macrophage colony-stimulating factor (GM-CSF), interleukin (IL)-17A, IL-23, alpha-melanocyte stimulating hormone (α-MSH), oxytocin, and substance P levels were highest in the DED after FS-LASIK group, followed by the DED without FS-LASIK and normal control groups (all P < .05). Interferon-γ and neurotensin levels were only significantly higher in the DED after FS-LASIK group (all P < .05). CONCLUSIONS Patients with chronic DED after FS-LASIK showed milder ocular symptoms, greater epithelial damage, and higher levels of tear inflammatory cytokines and neuropeptides than patients with DED without FS-LASIK, indicating that the nervous and immune systems may play significant roles in FS-LASIK-related chronic DED development. [J Refract Surg. 2023;39(8):556-563.].
Collapse
|
26
|
Compagnone A, Matheeussen A, De Vooght L, Cos P. Development and validation of a multiplex electrochemiluminescence immunoassay to evaluate dry eye disease in rat tear fluids. Sci Rep 2023; 13:12203. [PMID: 37500810 PMCID: PMC10374623 DOI: 10.1038/s41598-023-39397-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 07/25/2023] [Indexed: 07/29/2023] Open
Abstract
Dry eye disease (DED) is a challenge in ophthalmology. Rat models represent valuable tools to study the pathophysiology and to develop novel treatments. A major challenge in DED research is detecting multiple biomarkers in a low tear volume sample. Multiplex immunoassays for DED rat research are missing. We have developed a multiplex electrochemiluminescence immunoassay (ECLIA) to detect three biomarkers for DED: MMP-9, IL-17 and ICAM-1. Tears, used as matrix, were collected from six healthy Wistar rats. Assays were run based on the U-Plex Meso Scale Diagnostics (MSD) platform, by two independent operators according to the EMA guideline on bioanalytical method validation. Linear mixed, regression models were fit to perform the statistical analysis on the range of concentrations for the chosen analytes. During optimization, it has observed that incubation time, temperature and agitation affected the robustness of the protocol. ECLIA optimum conditions include the use of antibodies at 0.5 µg/ml concentration and 1 h incubation at room temperature with shaking. Precision met the acceptance criteria in the chosen range: 1062-133 pg/ml for ICAM-1, 275-34.4 pg/ml for IL-17, 1750-219 pg/ml for MMP-9. Accuracy and linearity were acceptable for a broader range. This is the first report of a validated ECLIA that allows measurements of three relevant DED biomarkers in rat tear fluids.
Collapse
Affiliation(s)
- Agnese Compagnone
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Department of Pharmaceutical Sciences, University of Antwerp, Campus Drie Eiken D.S.723, FFBD-FDFAR-LMPH, Universiteitsplein 1, 2610, Wilrijk, Antwerp, Belgium.
| | - An Matheeussen
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Department of Pharmaceutical Sciences, University of Antwerp, Campus Drie Eiken D.S.723, FFBD-FDFAR-LMPH, Universiteitsplein 1, 2610, Wilrijk, Antwerp, Belgium
| | - Linda De Vooght
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Department of Pharmaceutical Sciences, University of Antwerp, Campus Drie Eiken D.S.723, FFBD-FDFAR-LMPH, Universiteitsplein 1, 2610, Wilrijk, Antwerp, Belgium
| | - Paul Cos
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Department of Pharmaceutical Sciences, University of Antwerp, Campus Drie Eiken D.S.723, FFBD-FDFAR-LMPH, Universiteitsplein 1, 2610, Wilrijk, Antwerp, Belgium
| |
Collapse
|
27
|
Vereertbrugghen A, Pizzano M, Sabbione F, Keitelman IA, Shiromizu CM, Aguilar DV, Fuentes F, de Paiva CS, Giordano M, Trevani A, Galletti JG. An ocular Th1 immune response promotes corneal nerve damage independently of the development of corneal epitheliopathy. J Neuroinflammation 2023; 20:120. [PMID: 37217914 PMCID: PMC10201717 DOI: 10.1186/s12974-023-02800-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 05/09/2023] [Indexed: 05/24/2023] Open
Abstract
Proper sight is not possible without a smooth, transparent cornea, which is highly exposed to environmental threats. The abundant corneal nerves are interspersed with epithelial cells in the anterior corneal surface and are instrumental to corneal integrity and immunoregulation. Conversely, corneal neuropathy is commonly observed in some immune-mediated corneal disorders but not in others, and its pathogenesis is poorly understood. Here we hypothesized that the type of adaptive immune response may influence the development of corneal neuropathy. To test this, we first immunized OT-II mice with different adjuvants that favor T helper (Th)1 or Th2 responses. Both Th1-skewed mice (measured by interferon-γ production) and Th2-skewed (measured by interleukin-4 production) developed comparable ocular surface inflammation and conjunctival CD4+ T cell recruitment but no appreciable corneal epithelial changes upon repeated local antigenic challenge. Th1-skewed mice showed decreased corneal mechanical sensitivity and altered corneal nerve morphology (signs of corneal neuropathy) upon antigenic challenge. However, Th2-skewed mice also developed milder corneal neuropathy immediately after immunization and independently of ocular challenge, suggestive of adjuvant-induced neurotoxicity. All these findings were confirmed in wild-type mice. To circumvent unwanted neurotoxicity, CD4+ T cells from immunized mice were adoptively transferred to T cell-deficient mice. In this setup, only Th1-transferred mice developed corneal neuropathy upon antigenic challenge. To further delineate the contribution of each profile, CD4+ T cells were polarized in vitro to either Th1, Th2, or Th17 cells and transferred to T cell-deficient mice. Upon local antigenic challenge, all groups had commensurate conjunctival CD4+ T cell recruitment and macroscopic ocular inflammation. However, none of the groups developed corneal epithelial changes and only Th1-transferred mice showed signs of corneal neuropathy. Altogether, the data show that corneal nerves, as opposed to corneal epithelial cells, are sensitive to immune-driven damage mediated by Th1 CD4+ T cells in the absence of other pathogenic factors. These findings have potential therapeutic implications for ocular surface disorders.
Collapse
Affiliation(s)
- Alexia Vereertbrugghen
- Innate Immunity Laboratory, Institute of Experimental Medicine (CONICET/National Academy of Medicine of Buenos Aires), Buenos Aires, Argentina
| | - Manuela Pizzano
- Innate Immunity Laboratory, Institute of Experimental Medicine (CONICET/National Academy of Medicine of Buenos Aires), Buenos Aires, Argentina
| | - Florencia Sabbione
- Innate Immunity Laboratory, Institute of Experimental Medicine (CONICET/National Academy of Medicine of Buenos Aires), Buenos Aires, Argentina
| | - Irene Angelica Keitelman
- Innate Immunity Laboratory, Institute of Experimental Medicine (CONICET/National Academy of Medicine of Buenos Aires), Buenos Aires, Argentina
| | - Carolina Maiumi Shiromizu
- Innate Immunity Laboratory, Institute of Experimental Medicine (CONICET/National Academy of Medicine of Buenos Aires), Buenos Aires, Argentina
| | - Douglas Vera Aguilar
- Innate Immunity Laboratory, Institute of Experimental Medicine (CONICET/National Academy of Medicine of Buenos Aires), Buenos Aires, Argentina
| | - Federico Fuentes
- Confocal Microscopy Unit, Institute of Experimental Medicine (CONICET/National Academy of Medicine of Buenos Aires), Buenos Aires, Argentina
| | - Cintia S de Paiva
- Department of Ophthalmology, Ocular Surface Center, Cullen Eye Institute, Baylor College of Medicine, Houston, TX, USA
| | - Mirta Giordano
- Innate Immunity Laboratory, Institute of Experimental Medicine (CONICET/National Academy of Medicine of Buenos Aires), Buenos Aires, Argentina
| | - Analía Trevani
- Innate Immunity Laboratory, Institute of Experimental Medicine (CONICET/National Academy of Medicine of Buenos Aires), Buenos Aires, Argentina
| | - Jeremías G Galletti
- Innate Immunity Laboratory, Institute of Experimental Medicine (CONICET/National Academy of Medicine of Buenos Aires), Buenos Aires, Argentina.
| |
Collapse
|
28
|
Shanks RMQ, Romanowski EG, Romanowski JE, Davoli K, McNamara NA, Klarlund JK. Extending the use of biologics to mucous membranes by attachment of a binding domain. Commun Biol 2023; 6:477. [PMID: 37130912 PMCID: PMC10154311 DOI: 10.1038/s42003-023-04801-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 04/03/2023] [Indexed: 05/04/2023] Open
Abstract
Biologics are almost exclusively administered systemically, but localized delivery is preferable as it minimizes off-target exposure and allows more aggressive treatments. Topical application of biologics to epithelia is generally ineffective because most are covered with fluids and biologics are washed out too quickly to have significant therapeutic effects. Here we explore the idea that attaching a binding domain can serve as an "anchor" to extend the residency time of biologics on wet epithelia, allowing their effective use even with infrequent applications. We use topical application to the ocular surface as a challenging test since foreign substances are washed out especially efficiently by tear flow and blinking. Our results demonstrate that conjugation of antibodies to wheat germ agglutinin, which binds GlcNAc and sialic acid that are ubiquitously present in tissues, increases their half-life 350-fold upon application to the ocular surface in a mouse model of dry eye, a common and onerous disease in humans. Importantly, antibodies to IL-17A, IL-23, and IL-1β conjugated to the agglutinin reduces manifestations of dry eye, even when applied just once daily. In contrast, unconjugated antibodies are ineffective. Attaching an anchor to biologics is a simple means to overcome washout and to extend their therapeutic use.
Collapse
Affiliation(s)
- Robert M Q Shanks
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Charles T. Campbell Laboratory of Ophthalmic Microbiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Eric G Romanowski
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Charles T. Campbell Laboratory of Ophthalmic Microbiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - John E Romanowski
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Charles T. Campbell Laboratory of Ophthalmic Microbiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Katherine Davoli
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Nancy A McNamara
- School of Optometry and Vision Science Graduate Program, University of California, Berkeley, CA, USA
| | - Jes K Klarlund
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
29
|
Kate A, Shanbhag SS, Donthineni PR, Amescua G, Quinones VLP, Basu S. Role of topical and systemic immunosuppression in aqueous-deficient dry eye disease. Indian J Ophthalmol 2023; 71:1176-1189. [PMID: 37026249 PMCID: PMC10276741 DOI: 10.4103/ijo.ijo_2818_22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 12/19/2022] [Accepted: 01/27/2023] [Indexed: 04/08/2023] Open
Abstract
Immunosuppression in aqueous-deficient dry eye disease (ADDE) is required not only to improve the symptoms and signs but also to prevent further progression of the disease and its sight-threatening sequelae. This immunomodulation can be achieved through topical and/or systemic medications, and the choice of one drug over the other is determined by the underlying systemic disease. These immunosuppressive agents require a minimum of 6-8 weeks to achieve their beneficial effect, and during this time, the patient is usually placed on topical corticosteroids. Antimetabolites such as methotrexate, azathioprine, and mycophenolate mofetil, along with calcineurin inhibitors, are commonly used as first-line medications. The latter have a pivotal role in immunomodulation since T cells contribute significantly to the pathogenesis of ocular surface inflammation in dry eye disease. Alkylating agents are largely limited to controlling acute exacerbations with pulse doses of cyclophosphamide. Biologic agents, such as rituximab, are particularly useful in patients with refractory disease. Each group of drugs has its own side-effect profiles and requires a stringent monitoring schedule that must be followed to prevent systemic morbidity. A customized combination of topical and systemic medications is usually required to achieve adequate control, and this review aims to help the clinician choose the most appropriate modality and monitoring regimen for a given case of ADDE.
Collapse
Affiliation(s)
- Anahita Kate
- Shantilal Shanghvi Cornea Institue, LV Prasad Eye Institute, Vijayawada, Andhra Pradesh, India
| | - Swapna S Shanbhag
- Shantilal Shanghvi Cornea Institue, LV Prasad Eye Institute, Hyderabad, Telengana, India
| | - Pragnya R Donthineni
- Shantilal Shanghvi Cornea Institue, LV Prasad Eye Institute, Hyderabad, Telengana, India
| | - Guillermo Amescua
- Department of Ophthalmology, Duke Eye Center, Duke University School of Medicine, Durham 27705, NC, USA
| | - Victor L Perez Quinones
- Foster Center for Ocular Immunology, Department of Ophthalmology, Duke Eye Center, Duke University School of Medicine, Durham, North Carolina, USA
| | - Sayan Basu
- Shantilal Shanghvi Cornea Institue, LV Prasad Eye Institute, Hyderabad, Telengana, India
- Center for Ocular Regeneration (CORE), L. V. Prasad Eye Institute, Hyderabad, Telangana, India
- Brien Holden Eye Research Centre, L. V. Prasad Eye Institute, Hyderabad, Telangana, India
| |
Collapse
|
30
|
Bao X, Zhong Y, Yang C, Chen Y, Han Y, Lin X, Huang C, Wang K, Liu Z, Li C. T-Cell Repertoire Analysis in the Conjunctiva of Murine Dry Eye Model. Invest Ophthalmol Vis Sci 2023; 64:14. [PMID: 36877515 PMCID: PMC10007900 DOI: 10.1167/iovs.64.3.14] [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: 03/07/2023] Open
Abstract
Purpose Dry eye is closely related to the activation and proliferation of immune cells, especially T cells. However, the determination of the preferential T-cell clonotypes is technically challenging. This study aimed to investigate the characterization of T-cell receptor (TCR) repertoire in the conjunctiva during dry eye. Methods A desiccating stress animal model was established using C57/BL6 mice (8-10 weeks, female). After 7 days of stress stimulation, the slit-lamp image and Oregon-green-dextran staining were used to evaluate the ocular surface injury. Periodic acid-Schiff staining was used to measure the number of goblet cells. Flow cytometry was used to detect the activation and proliferation of T cells in the conjunctiva and cervical lymph nodes. Next-generation sequencing was used to detect the αβ TCR repertoire of the conjunctiva. Results The αβ TCR diversity increased significantly in the dry eye group, including the higher CDR3 amino acid length, marked gene usage on TCR V and J gene segments, extensive V(D)J recombination, and distinct CDR3 aa motifs. More important, several T-cell clonotypes were uniquely identified in dry eye. Furthermore, these perturbed rearrangements were reversed after glucocorticoid administration. Conclusions A comprehensive analysis of the αβ TCR repertoire in the conjunctiva of the dry eye mouse model was performed. Data in this study contributed significantly to the research on dry eye pathogenesis by demonstrating the TCR gene distribution and disease-specific TCR signatures. This study further provided some potential predictive T-cell biomarkers for future studies.
Collapse
Affiliation(s)
- Xiaorui Bao
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Eye Institute & Affiliated Xiamen Eye Center, School of Medicine, Xiamen University, Xiamen, Fujian, China.,Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, China
| | - Yanlin Zhong
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Eye Institute & Affiliated Xiamen Eye Center, School of Medicine, Xiamen University, Xiamen, Fujian, China.,Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, China
| | - Chunyan Yang
- School of Life Science, Xiamen University, Xiamen, Fujian, China
| | - Yujie Chen
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Eye Institute & Affiliated Xiamen Eye Center, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Yi Han
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Eye Institute & Affiliated Xiamen Eye Center, School of Medicine, Xiamen University, Xiamen, Fujian, China.,Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, China
| | - Xiang Lin
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, China
| | - Caihong Huang
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Eye Institute & Affiliated Xiamen Eye Center, School of Medicine, Xiamen University, Xiamen, Fujian, China.,Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, China
| | - Kejia Wang
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Eye Institute & Affiliated Xiamen Eye Center, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Zuguo Liu
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Eye Institute & Affiliated Xiamen Eye Center, School of Medicine, Xiamen University, Xiamen, Fujian, China.,Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, China.,The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Cheng Li
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Eye Institute & Affiliated Xiamen Eye Center, School of Medicine, Xiamen University, Xiamen, Fujian, China.,Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, China
| |
Collapse
|
31
|
Perez VL, Mah FS, Willcox M, Pflugfelder S. Anti-Inflammatories in the Treatment of Dry Eye Disease: A Review. J Ocul Pharmacol Ther 2023; 39:89-101. [PMID: 36796014 DOI: 10.1089/jop.2022.0133] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2023] Open
Abstract
Inflammation is an important driver of dry eye disease (DED) pathogenesis. An initial insult that results in the loss of tear film homeostasis can initiate a nonspecific innate immune response that leads to a chronic and self-sustaining inflammation of the ocular surface, which results in classic symptoms of dry eye. This initial response is followed by a more prolonged adaptive immune response, which can perpetuate and aggravate inflammation and result in a vicious cycle of chronic inflammatory DED. Effective anti-inflammatory therapies can help patients exit this cycle, and effective diagnosis of inflammatory DED and selection of the most appropriate treatment are therefore key to successful DED management and treatment. This review explores the cellular and molecular mechanisms of the immune and inflammatory components of DED, and examines the evidence base for the use of currently available topical treatment options. These agents include topical steroid therapy, calcineurin inhibitors, T cell integrin antagonists, antibiotics, autologous serum/plasma therapy, and omega-3 fatty acid dietary supplements.
Collapse
Affiliation(s)
- Victor L Perez
- Department of Ophthalmology, Foster Center for Ocular Immunology at Duke Eye Center, Duke University School of Medicine, Durham, North Carolina. USA
| | - Francis S Mah
- Scripps Clinic Torrey Pines, La Jolla, California, USA
| | - Mark Willcox
- School of Optometry and Vision Science, UNSW Sydney, Sydney, New South Wales, Australia
| | - Stephen Pflugfelder
- Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
32
|
Paik B, Tong L. Polymorphisms in Lymphotoxin-Alpha as the "Missing Link" in Prognosticating Favourable Response to Omega-3 Supplementation for Dry Eye Disease: A Narrative Review. Int J Mol Sci 2023; 24:ijms24044236. [PMID: 36835647 PMCID: PMC9965360 DOI: 10.3390/ijms24044236] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 02/08/2023] [Accepted: 02/16/2023] [Indexed: 02/23/2023] Open
Abstract
Elements of inflammation are found in almost all chronic ocular surface disease, such as dry eye disease. The chronicity of such inflammatory disease speaks to the dysregulation of innate and adaptive immunity. There has been a rising interest in omega-3 fatty acids to attenuate inflammation. While many cell-based (in vitro) studies verify the anti-inflammatory effects of omega-3, different human trials report discordant outcomes after supplementation. This may be due to underlying inter-individual differences in inflammatory cytokine metabolism (such as tumor necrosis factor alpha (TNF-α)), in which genetic differences might play a role, such as polymorphisms in the lymphotoxin alpha (LT-α) gene. Inherent TNF-α production affects omega-3 response and is also associated with LT-α genotype. Therefore, LT-α genotype might predict omega-3 response. Using the NIH dbSNP, we analyzed the relative frequency of LT-α polymorphisms among various ethnicities, each weighted by the genotype's probability of positive response. While the probability of response for unknown LT-α genotypes are 50%, there is greater distinction in response rates between various genotypes. Hence, there is value in genetic testing to prognosticate an individual's response to omega-3.
Collapse
Affiliation(s)
- Benjamin Paik
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | - Louis Tong
- Department of Cornea and External Eye Disease, Singapore National Eye Center, Singapore 168751, Singapore
- Ocular Surface Research Group, Singapore Eye Research Institute, Singapore 169856, Singapore
- Ophthalmology and Visual Sciences Academic Clinical Program, Duke-NUS Medical School, Singapore 169857, Singapore
- Correspondence: ; Tel.: +65-6227-7255
| |
Collapse
|
33
|
Kim BY, Oh C, Jeon D, Jun I, Lee HK, Kim BR, Park J, Seo KY, Kim KA, Lim D, Lee S, Lee J, Yoon H, Kim TI, Namkung W. Synthetic Strategies for Improving Solubility: Optimization of Novel Pyrazolo[1,5- a]pyrimidine CFTR Activator That Ameliorates Dry Eye Disease. J Med Chem 2023; 66:413-434. [PMID: 36573286 PMCID: PMC9841530 DOI: 10.1021/acs.jmedchem.2c01382] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Indexed: 12/28/2022]
Abstract
Dry eye disease (DED) is one of the most prevalent ocular diseases but has limited treatment options. Cystic fibrosis transmembrane conductance regulator (CFTR), a major chloride channel that stimulates fluid secretion in the ocular surface, may pave the way for new therapeutic strategies for DED. Herein, we report the optimization of Cact-3, a potent CFTR activator with poor solubility, to 16d, a potent CFTR activator with suitable solubility for eye drop formulation. Notably, 16d was well distributed in target tissues including cornea and conjunctiva with minimal systemic exposure in rabbit. Topical ocular instillation of 16d significantly enhanced tear secretion and improved corneal erosion in a mouse model of DED. In addition, 16d significantly reduced mRNA expression of pro-inflammatory cytokines including IL-1β, IL-17, and TNF-α and MMP2 in cornea and conjunctiva of DED mice.
Collapse
Affiliation(s)
- Bo Yi Kim
- The
Institute of Vision Research, Department of Ophthalmology, Severance
Hospital, Yonsei University College of Medicine, 50-1 Yonsei-ro,
Seodaemungu, Seoul 03722, Korea
| | - Changmok Oh
- Research
Laboratories, ILDONG Pharmaceutical Co.,
Ltd., 20, Samsung 1-ro
1-gil, Hwaseong 18449, Korea
| | - Dongkyu Jeon
- College
of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Korea
| | - Ikhyun Jun
- The
Institute of Vision Research, Department of Ophthalmology, Severance
Hospital, Yonsei University College of Medicine, 50-1 Yonsei-ro,
Seodaemungu, Seoul 03722, Korea
| | - Ho K. Lee
- College
of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Korea
| | - Bo-Rahm Kim
- The
Institute of Vision Research, Department of Ophthalmology, Severance
Hospital, Yonsei University College of Medicine, 50-1 Yonsei-ro,
Seodaemungu, Seoul 03722, Korea
| | - Jinhong Park
- College
of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Korea
| | - Kyoung Yul Seo
- The
Institute of Vision Research, Department of Ophthalmology, Severance
Hospital, Yonsei University College of Medicine, 50-1 Yonsei-ro,
Seodaemungu, Seoul 03722, Korea
| | - Kyeong-A Kim
- Research
Laboratories, ILDONG Pharmaceutical Co.,
Ltd., 20, Samsung 1-ro
1-gil, Hwaseong 18449, Korea
| | - Dami Lim
- Research
Laboratories, ILDONG Pharmaceutical Co.,
Ltd., 20, Samsung 1-ro
1-gil, Hwaseong 18449, Korea
| | - Seolhee Lee
- Research
Laboratories, ILDONG Pharmaceutical Co.,
Ltd., 20, Samsung 1-ro
1-gil, Hwaseong 18449, Korea
| | - Jooyun Lee
- Research
Laboratories, ILDONG Pharmaceutical Co.,
Ltd., 20, Samsung 1-ro
1-gil, Hwaseong 18449, Korea
| | - Hongchul Yoon
- Research
Laboratories, ILDONG Pharmaceutical Co.,
Ltd., 20, Samsung 1-ro
1-gil, Hwaseong 18449, Korea
| | - Tae-im Kim
- The
Institute of Vision Research, Department of Ophthalmology, Severance
Hospital, Yonsei University College of Medicine, 50-1 Yonsei-ro,
Seodaemungu, Seoul 03722, Korea
| | - Wan Namkung
- College
of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Korea
| |
Collapse
|
34
|
Jiang Y, Lin S, Gao Y. Mesenchymal Stromal Cell-Based Therapy for Dry Eye: Current Status and Future Perspectives. Cell Transplant 2022; 31:9636897221133818. [PMID: 36398793 PMCID: PMC9679336 DOI: 10.1177/09636897221133818] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Dry eye is one of the most common chronic diseases in ophthalmology. It affects quality of life and has become a public health problem that cannot be ignored. The current treatment methods mainly include artificial tear replacement therapy, anti-inflammatory therapy, and local immunosuppressive therapy. These treatments are mainly limited to improvement of ocular surface discomfort and other symptoms. In recent years, regenerative medicine has developed rapidly, and ophthalmologists are working on new methods to treat dry eye. Mesenchymal stromal cells (MSCs) have anti-inflammatory, tissue repair, and immune regulatory effects, and have become a promising tool for the treatment of dry eye. These effects can also be produced by MSC-derived exosomes (MSC-Exos). As a cell-free therapy, MSC-Exos are hypoimmunogenic, serve more stable entities, and compared with MSCs, reduce the safety risks associated with the injection of live cells. This article reviews current knowledge about MSCs and MSC-Exos, and highlights the latest progress and future prospects of MSC-based therapy in dry eye treatment.
Collapse
Affiliation(s)
- Yuting Jiang
- Department of Ophthalmology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Shu Lin
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Yingying Gao
- Department of Ophthalmology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China,Yingying Gao, The Second Affiliated Hospital of Fujian Medical University, No. 34 North Zhongshan Road, Quanzhou 362000, Fujian, China.
| |
Collapse
|
35
|
Li L, Li Y, Zhu X, Wu B, Tang Z, Wen H, Yuan J, Zheng Q, Chen W. Conjunctiva Resident γδ T Cells Expressed High Level of IL-17A and Promoted the Severity of Dry Eye. Invest Ophthalmol Vis Sci 2022; 63:13. [PMID: 36350619 PMCID: PMC9652718 DOI: 10.1167/iovs.63.12.13] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Purpose Conjunctival inflammation promotes ocular surface disorders in dry eye disease (DED). Here we identified γδ T cells as the predominant source of IL-17A in the murine conjunctiva and assessed their contribution to the pathogenesis of DED. Methods We enrolled 22 patients with DED, and analyzed the proportion of γδ T cells in the conjunctival epithelial samples by flow cytometry. Adult C57Bl/6 wild-type and TCRδ−/− mice were used to induce DED models to investigate the role of γδ T cells. The characteristics of immune cell infiltration and the expression of immune-related cytokines or markers in mouse conjunctiva were analyzed by flow cytometry, Western blot, and quantitative polymerase chain reaction. Results The proportion of γδ T cells in the human DED conjunctiva is significantly higher in patients with severe corneal epithelial defects than in mild ones, which is consistently observed in the murine DED model. Further, a high level of IL-17A but not IFN-γ is detected in the conjunctiva of mice. The increased murine IL-17A–producing cells on the conjunctiva are identified as γδ T cells predominantly and Th17 cells to a lesser extent. Ablation of γδ T cells by antibody depletion or genetic deletion of TCRδ alleviates ocular surface damage in the murine DED model. Conclusions Our studies evaluate human and experimental murine DED for evidence of γδ T-cell–mediated inflammation and highlight a potential therapeutic synergy by targeting IL-17 and γδ T cells in DED treatment.
Collapse
Affiliation(s)
- Ling Li
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- The Affiliated Ningbo Eye Hospital of Wenzhou Medical University, Ningbo, Zhejiang, China
| | - Yanxiao Li
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xinhao Zhu
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Biao Wu
- Shaoxing people's hospital, Shaoxing, Zhejiang, China
| | - Zhuo Tang
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Han Wen
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jianshu Yuan
- The Affiliated Ningbo Eye Hospital of Wenzhou Medical University, Ningbo, Zhejiang, China
| | - Qinxiang Zheng
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- The Affiliated Ningbo Eye Hospital of Wenzhou Medical University, Ningbo, Zhejiang, China
| | - Wei Chen
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- The Affiliated Ningbo Eye Hospital of Wenzhou Medical University, Ningbo, Zhejiang, China
| |
Collapse
|
36
|
Therapeutic Potential of d-MAPPS™ for Ocular Inflammatory Diseases and Regeneration of Injured Corneal and Retinal Tissue. Int J Mol Sci 2022; 23:ijms232113528. [DOI: 10.3390/ijms232113528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 11/01/2022] [Accepted: 11/03/2022] [Indexed: 11/06/2022] Open
Abstract
The invasion of microbial pathogens and/or sterile inflammation caused by physical/chemical injuries, increased ocular pressure, oxidative stress, and ischemia could lead to the generation of detrimental immune responses in the eyes, which result in excessive tissue injury and vision loss. The bioavailability of eye drops that are enriched with immunoregulatory and trophic factors which may concurrently suppress intraocular inflammation and promote tissue repair and regeneration is generally low. We recently developed “derived- Multiple Allogeneic Proteins Paracrine Signaling regenerative biologics platform technology d-MAPPS™”, a bioengineered biological product which is enriched with immunomodulatory and trophic factors that can efficiently suppress detrimental immune responses in the eye and promote the repair and regeneration of injured corneal and retinal tissues. The results obtained in preclinical and clinical studies showed that d-MAPPS™ increased the viability of injured corneal cells, inhibited the production of inflammatory cytokines in immune cells, alleviated inflammation, and restored vision loss in patients suffering from meibomian gland dysfunction and dry eye disease. Herewith, we emphasized molecular mechanisms responsible for the therapeutic efficacy of d-MAPPS™ and we presented the main beneficial effects of d-MAPPS™ in clinical settings, indicating that the topical administration of d-MAPPS™ could be considered a new therapeutic approach for the treatment of ocular inflammatory diseases and for the repair and regeneration of injured corneal and retinal tissues.
Collapse
|
37
|
Asiedu K. Role of ocular surface neurobiology in neuronal-mediated inflammation in dry eye disease. Neuropeptides 2022; 95:102266. [PMID: 35728484 DOI: 10.1016/j.npep.2022.102266] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 06/07/2022] [Accepted: 06/08/2022] [Indexed: 01/18/2023]
Abstract
Inflammation is the consequence of dry eye disease regardless of its etiology. Several injurious or harmless processes to the ocular surface neurons promote ocular surface neurogenic inflammation, leading to the vicious cycle of dry eye disease. These processes include the regular release of neuromediators during the conduction of ocular surface sensations, hyperosmolarity-induced ocular surface neuronal damage, neuro-regenerative activities, and neuronal-mediated dendritic cell activities. Neurogenic inflammation appears to be the main culprit, instigating the self-perpetuating inflammation observed in patients with dry eye disease.
Collapse
Affiliation(s)
- Kofi Asiedu
- School of Optometry & Vision Science, Faculty of Medicine and Health, University of New South Wales, Sydney, Australia.
| |
Collapse
|
38
|
Soifer M, Azar NS, Mousa HM, Perez VL. Ocular Surface Inflammatory Disorders (OSID): A Collective of Systemic Etiologies Which Cause or Amplify Dry Eye Syndrome. Front Med (Lausanne) 2022; 9:949202. [PMID: 35872765 PMCID: PMC9301237 DOI: 10.3389/fmed.2022.949202] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 06/20/2022] [Indexed: 11/16/2022] Open
Abstract
The ocular surface inflammatory disorders (OSID) are caused by systemic disorders that conduct a persistent inflammatory reaction in the ocular adnexal connective tissues, such as the conjunctiva, lacrimal gland (LG) and meibomian glands (MGs), which cause an inflammatory dry eye. The etiologies of OSID are a subset of systemic pathologies such as graft versus host disease, Sjögren’s syndrome, allergies, cicatrizing conjunctivitis, and more. These cause a purely inflammatory dry eye syndrome as a consequence of the persistent surrounding inflammation in the adnexal tissues, which is distinct from the age-related dry eye disease. A limitation toward management of these conditions is the lack of available biomarkers that can detect presence of inflammation and quantify damage on the conjunctiva and LG, even though these are considered to be drivers of the inflammatory milieu. The OSID and dry eye syndrome are caused by different immune cells which are not exclusively limited to T cell lymphocytes, but rather derive from an orchestrated multicellular immunologic response. Recognition of this syndrome is crucial to direct research in a direction that clarifies the potential role of inflammation and its associated immune phenotype on the conjunctiva and adnexal ocular tissues in OSID and dry eye syndrome. On this paper, we review the basic and clinical research evidence for the existence of OSID with focus on the different immune cells involved, the target tissues and potential consequences and OSIDs diagnostic and therapeutic implications.
Collapse
Affiliation(s)
- Matias Soifer
- Foster Center for Ocular Immunology, Duke Eye Institute, Durham, NC, United States
- Department of Ophthalmology, Duke University Medical Center, Durham, NC, United States
| | - Nadim S. Azar
- Foster Center for Ocular Immunology, Duke Eye Institute, Durham, NC, United States
- Department of Ophthalmology, Duke University Medical Center, Durham, NC, United States
| | - Hazem M. Mousa
- Foster Center for Ocular Immunology, Duke Eye Institute, Durham, NC, United States
- Department of Ophthalmology, Duke University Medical Center, Durham, NC, United States
| | - Victor L. Perez
- Foster Center for Ocular Immunology, Duke Eye Institute, Durham, NC, United States
- Department of Ophthalmology, Duke University Medical Center, Durham, NC, United States
- *Correspondence: Victor L. Perez,
| |
Collapse
|
39
|
Wang HH, Chen WY, Huang YH, Hsu SM, Tsao YP, Hsu YH, Chang MS. Interleukin-20 is involved in dry eye disease and is a potential therapeutic target. J Biomed Sci 2022; 29:36. [PMID: 35681232 PMCID: PMC9178884 DOI: 10.1186/s12929-022-00821-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 05/29/2022] [Indexed: 11/23/2022] Open
Abstract
Background Dry eye disease (DED) is a common disease in ophthalmology, affecting millions of people worldwide. Recent studies have shown that inflammation is the core mechanism of DED. IL-20 is a proinflammatory cytokine involved in various inflammatory diseases. Therefore, we aimed to explore the role of this cytokine in the pathogenesis of DED and evaluate the therapeutic potential of the anti-IL-20 monoclonal antibody (mAb) 7E for DED treatment. Methods Clinical tear samples from patients with DED and non-DED controls were collected and their IL-20 protein levels were determined. We established three DED animal models to explore the role of IL-20 and the efficacy of IL-20 antibody in DED. Benzalkonium chloride (BAC)-induced over-evaporative DED, extra-orbital lacrimal gland excision (LGE)-induced aqueous tear-deficient DED, and desiccating stress (DS)-induced combined over-evaporative and aqueous tear-deficient DED animal models were established to investigate the role of IL-20. The anti-IL-20 antibody 7E was established to neutralize IL-20 activity. The effects of IL-20 or 7E on human corneal epithelial cells and macrophages under hyperosmotic stress were analyzed. 7E was topically applied to eyes to evaluate the therapeutic effects in the DED animal models. Results IL-20 was significantly upregulated in the tears of patients with DED and in the tears and corneas of DED animal models. Under hyperosmotic stress, IL-20 expression was induced via NFAT5 activation in corneal epithelial cells. 7E suppressed hyperosmotic stress-induced activation of macrophages. IL-20 induced cell death in corneal epithelial cells and 7E protected cells from hyperosmotic stress-induced cell death. Blocking IL-20 signaling with 7E protected mice from BAC-induced, LGE-induced, and DS-induced DED by reducing DED symptoms and inhibiting inflammatory responses, macrophage infiltration, apoptosis, and Th17 populations in the conjunctiva and draining lymph nodes. Conclusions Our results demonstrated the functions of IL-20 in DED and presented a potential therapeutic option for this condition. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12929-022-00821-2.
Collapse
Affiliation(s)
- Hsiao-Hsuan Wang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Wei-Yu Chen
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Hsun Huang
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Ophthalmology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Sheng-Min Hsu
- Department of Ophthalmology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yeou-Ping Tsao
- Department of Medical Research, Mackay Memorial Hospital, Taipei, Taiwan.,Department of Ophthalmology, Mackay Memorial Hospital, Taipei, Taiwan
| | - Yu-Hsiang Hsu
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Research Center of Clinical Medicine, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Ming-Shi Chang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan. .,Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
40
|
Mittal SK, Cho W, Elbasiony E, Guan Y, Foulsham W, Chauhan SK. Mesenchymal stem cells augment regulatory T cell function via CD80-mediated interactions and promote allograft survival. Am J Transplant 2022; 22:1564-1577. [PMID: 35170213 PMCID: PMC11261724 DOI: 10.1111/ajt.17001] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 01/25/2022] [Accepted: 02/09/2022] [Indexed: 02/05/2023]
Abstract
Mesenchymal stem cells (MSCs) and regulatory T cells (Tregs) both have been shown to modulate the alloimmune response and promote transplant survival. Mounting evidence suggests that MSCs augment Treg function, but the mechanisms underlying this phenomenon have not been fully deciphered. Here, we identified that MSCs express substantial levels of CD80 and evaluated its immunoregulatory function using in vivo and in vitro experiments. Our in vitro culture assays demonstrated that MSCs induce expression of FoxP3 in Tregs in a contact-dependent manner, and the blockade of CD80 abrogates this FoxP3 induction and Treg-mediated suppression of T cell proliferation. Moreover, supplementation of soluble CD80 significantly upregulated FoxP3 expression. Using a well-characterized murine model of corneal transplantation, we show that silencing CD80 in MSCs diminishes the capacity of MSCs to promote selective graft infiltration of Tregs, promote FoxP3 expression and upregulate suppressive function of Tregs. Consequently, MSCs, following CD80 knockdown, failed to promote corneal allograft survival.
Collapse
Affiliation(s)
- Sharad K Mittal
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, USA
| | - WonKyung Cho
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, USA
| | - Elsayed Elbasiony
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, USA
| | - Yilin Guan
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, USA
| | - William Foulsham
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, USA
| | - Sunil K Chauhan
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
41
|
A new non-human primate model of desiccating stress-induced dry eye disease. Sci Rep 2022; 12:7957. [PMID: 35562371 PMCID: PMC9106732 DOI: 10.1038/s41598-022-12009-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 04/28/2022] [Indexed: 11/17/2022] Open
Abstract
Dry eye disease (DED), a multifactorial ocular surface disease, is estimated to affect up to 34% of individuals over 50 years old. Although numerous animal models, including rodents and rabbits, have been developed to mimic the pathophysiologic mechanisms involved in dry eye, there is a lack of non-human primate (NHP) models, critical for translational drug studies. Here, we developed a novel desiccating stress-induced dry eye disease model using Rhesus macaque monkeys. The monkeys were housed in a controlled environment room for 21 to 36 days under humidity, temperature, and airflow regulation. Following desiccating stress, NHPs demonstrated clinical symptoms similar to those of humans, as shown by increased corneal fluorescein staining (CFS) and decreased tear-film breakup time (TFBUT). Moreover, corticosteroid treatment significantly reduced CFS scoring, restored TFBUT, and prevented upregulation of tear proinflammatory cytokines as observed in dry eye patients following steroid treatment. The close resemblance of clinical symptoms and treatment responses to those of human DED patients provides great translational value to the NHP model, which could serve as a clinically relevant animal model to study the efficacy of new potential treatments for DED.
Collapse
|
42
|
Jeon D, Jun I, Lee HK, Park J, Kim BR, Ryu K, Yoon H, Kim TI, Namkung W. Novel CFTR Activator Cact-3 Ameliorates Ocular Surface Dysfunctions in Scopolamine-Induced Dry Eye Mice. Int J Mol Sci 2022; 23:5206. [PMID: 35563597 PMCID: PMC9101838 DOI: 10.3390/ijms23095206] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 05/02/2022] [Accepted: 05/05/2022] [Indexed: 02/04/2023] Open
Abstract
Cystic fibrosis transmembrane conductance regulator (CFTR) is highly expressed on the ocular epithelium and plays a pivotal role in the fluid secretion driven by chloride transport. Dry eye disease is one of the most common diseases with limited therapeutic options. In this study, a high-throughput screening was performed to identify novel CFTR activators capable of inducing chloride secretion on the ocular surface. The screening of 50,000 small molecules revealed three novel CFTR activators. Among them, the most potent CFTR activator, Cact-3 (7-(3,4-dimethoxyphenyl)-N-(4-ethoxyphenyl)pyrazolo [1,5-α]pyrimidine-2-carboxamide), produced large and sustained Cl- currents in WT-CFTR-expressing FRT cells with no alterations of ANO1 and hERG channel activity. The application of Cact-3 strongly activated CFTR in the ocular epithelia of mice and it also significantly increased CFTR-mediated Cl- transport in a primary cultured human conjunctival epithelium. Cact-3 strongly stimulated tear secretion in normal mice. In addition, Cact-3 significantly reduced ocular surface damage and the expression of proinflammatory factors, including interleukin (IL)-1β, IL-6, tumor necrosis factor (TNF)-α, and interferon (IFN)-γ in an experimental mouse model of dry eye disease. These results suggest that Cact-3, a novel CFTR activator, may be a potential development candidate for the treatment of dry eye disease.
Collapse
Affiliation(s)
- Dongkyu Jeon
- Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Korea; (D.J.); (H.K.L.); (J.P.); (K.R.)
| | - Ikhyun Jun
- Department of Ophthalmology, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemoon-Gu, Seoul 03722, Korea; (I.J.); (B.-R.K.)
| | - Ho K. Lee
- Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Korea; (D.J.); (H.K.L.); (J.P.); (K.R.)
| | - Jinhong Park
- Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Korea; (D.J.); (H.K.L.); (J.P.); (K.R.)
| | - Bo-Rahm Kim
- Department of Ophthalmology, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemoon-Gu, Seoul 03722, Korea; (I.J.); (B.-R.K.)
| | - Kunhi Ryu
- Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Korea; (D.J.); (H.K.L.); (J.P.); (K.R.)
| | - Hongchul Yoon
- Research Laboratories, ILDONG Pharmaceutical Co., Ltd., 20, Samsung 1-ro 1-gil, Hwaseong 18449, Korea;
| | - Tae-im Kim
- Department of Ophthalmology, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemoon-Gu, Seoul 03722, Korea; (I.J.); (B.-R.K.)
| | - Wan Namkung
- Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Korea; (D.J.); (H.K.L.); (J.P.); (K.R.)
- Department of Ophthalmology, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemoon-Gu, Seoul 03722, Korea; (I.J.); (B.-R.K.)
| |
Collapse
|
43
|
Tagami M, Kakehashi A, Katsuyama-Yoshikawa A, Misawa N, Sakai A, Wanibuchi H, Azumi A, Honda S. FOXP3 and CXCR4-positive regulatory T cells in the tumor stroma as indicators of tumor immunity in the conjunctival squamous cell carcinoma microenvironment. PLoS One 2022; 17:e0263895. [PMID: 35358193 PMCID: PMC8970378 DOI: 10.1371/journal.pone.0263895] [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: 07/02/2021] [Accepted: 01/22/2022] [Indexed: 11/29/2022] Open
Abstract
Conjunctival squamous cell carcinoma (SCC) is the most common ocular surface neoplasia. The purpose of this retrospective study was to examine the role of regulatory T cell (Treg) activity in tumor immunity and investigate the tumor microenvironment as a new treatment focus in conjunctival SCC. Cancer progression gene array and immunohistochemical analyses of FOXP3 as a Treg marker, CD8 as a tumor-infiltrating lymphocyte marker, and CXCR4 expression on activated Tregs were conducted in a series of 31 conjunctival SCC cases. The objective was to investigate the immunoreactive response in tumor cells and stromal cells in the cancer microenvironment. The stroma ratio in tumor cells was investigated by monitoring α-smooth muscle actine (SMA) expression between carcinoma in situ (Tis) and advanced carcinoma (Tadv) (P<0.01). No significant change in PD-L1 expression was observed in this study (P = 0.15). Staining patterns of FOXP3, CD8, and CXCR4 were examined separately between tumor cells and stromal cells in SCC tumors. Differences in staining of FOXP3 in Tregs and CD8 in tumor-infiltrating lymphocytes in tumor stroma in the Tis group were observed compared with the Tadv group (each P<0.01). In addition, double immunostaining of CXCR4/FOXP3 was correlated with progression-free survival (P = 0.049). Double immunostaining of CXCR4/FOXP3 correlated with American Joint Committee on Cancer T-stage, independent of age or Ki67 index (P<0.01). Our results show that FOXP3 and the CXCR4/FOXP3 axis are important pathologic and prognostic factors of ocular surface neoplasia, including SCC. The tumor microenvironment of conjunctival SCC should be considered in the future development of treatment options.
Collapse
Affiliation(s)
- Mizuki Tagami
- Department of Ophthalmology and Visual Sciences, Graduate School of Medicine, Osaka City University, Osaka, Japan
- Ophthalmology Department and Eye Center, Kobe Kaisei Hospital, Kobe, Hyogo, Japan
- * E-mail:
| | - Anna Kakehashi
- Department of Molecular Pathology, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | - Atsuko Katsuyama-Yoshikawa
- Ophthalmology Department and Eye Center, Kobe Kaisei Hospital, Kobe, Hyogo, Japan
- Division of Ophthalmology, Department of Surgery, Kobe University Graduate School of Medicine, Hyogo, Japan
| | - Norihiko Misawa
- Department of Ophthalmology and Visual Sciences, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | - Atsushi Sakai
- Department of Ophthalmology and Visual Sciences, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | - Hideki Wanibuchi
- Department of Molecular Pathology, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | - Atsushi Azumi
- Ophthalmology Department and Eye Center, Kobe Kaisei Hospital, Kobe, Hyogo, Japan
| | - Shigeru Honda
- Department of Ophthalmology and Visual Sciences, Graduate School of Medicine, Osaka City University, Osaka, Japan
| |
Collapse
|
44
|
Alam J, Yazdanpanah G, Ratnapriya R, Borcherding N, de Paiva CS, Li D, Guimaraes de Souza R, Yu Z, Pflugfelder SC. IL-17 Producing Lymphocytes Cause Dry Eye and Corneal Disease With Aging in RXRα Mutant Mouse. Front Med (Lausanne) 2022; 9:849990. [PMID: 35402439 PMCID: PMC8983848 DOI: 10.3389/fmed.2022.849990] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 02/24/2022] [Indexed: 11/22/2022] Open
Abstract
Purpose To investigate IL-17 related mechanisms for developing dry eye disease in the Pinkie mouse strain with a loss of function RXRα mutation. Methods Measures of dry eye disease were assessed in the cornea and conjunctiva. Expression profiling was performed by single-cell RNA sequencing (scRNA-seq) to compare gene expression in conjunctival immune cells. Conjunctival immune cells were immunophenotyped by flow cytometry and confocal microscopy. The activity of RXRα ligand 9-cis retinoic acid (RA) was evaluated in cultured monocytes and γδ T cells. Results Compared to wild type (WT) C57BL/6, Pinkie has increased signs of dry eye disease, including decreased tear volume, corneal barrier disruption, corneal/conjunctival cornification and goblet cell loss, and corneal vascularization, opacification, and ulceration with aging. ScRNA-seq of conjunctival immune cells identified γδ T cells as the predominant IL-17 expressing population in both strains and there is a 4-fold increased percentage of γδ T cells in Pinkie. Compared to WT, IL-17a, and IL-17f significantly increased in Pinkie with conventional T cells and γδ T cells as the major producers. Flow cytometry revealed an increased number of IL-17+ γδ T cells in Pinkie. Tear concentration of the IL-17 inducer IL-23 is significantly higher in Pinkie. 9-cis RA treatment suppresses stimulated IL-17 production by γδ T and stimulatory activity of monocyte supernatant on γδ T cell IL-17 production. Compared to WT bone marrow chimeras, Pinkie chimeras have increased IL-17+ γδ T cells in the conjunctiva after desiccating stress and anti-IL-17 treatment suppresses dry eye induced corneal MMP-9 production/activity and conjunctival goblet cell loss. Conclusion These findings indicate that RXRα suppresses generation of dry eye disease-inducing IL-17 producing lymphocytes s in the conjunctiva and identifies RXRα as a potential therapeutic target in dry eye.
Collapse
Affiliation(s)
- Jehan Alam
- Department of Ophthalmology, Ocular Surface Center, Baylor College of Medicine, Houston, TX, United States
| | - Ghasem Yazdanpanah
- Department of Ophthalmology, Ocular Surface Center, Baylor College of Medicine, Houston, TX, United States
| | - Rinki Ratnapriya
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX, United States
| | - Nicholas Borcherding
- Department of Pathology, Washington University School of Medicine, St. Louis, MO, United States
| | - Cintia S. de Paiva
- Department of Ophthalmology, Ocular Surface Center, Baylor College of Medicine, Houston, TX, United States
| | - DeQuan Li
- Department of Ophthalmology, Ocular Surface Center, Baylor College of Medicine, Houston, TX, United States
| | - Rodrigo Guimaraes de Souza
- Department of Ophthalmology, Ocular Surface Center, Baylor College of Medicine, Houston, TX, United States
- Department of Ophthalmology, University of São Paulo, São Paulo, Brazil
| | - Zhiyuan Yu
- Department of Ophthalmology, Ocular Surface Center, Baylor College of Medicine, Houston, TX, United States
| | - Stephen C. Pflugfelder
- Department of Ophthalmology, Ocular Surface Center, Baylor College of Medicine, Houston, TX, United States
- *Correspondence: Stephen C. Pflugfelder
| |
Collapse
|
45
|
Gut-derived butyrate suppresses ocular surface inflammation. Sci Rep 2022; 12:4512. [PMID: 35296712 PMCID: PMC8927112 DOI: 10.1038/s41598-022-08442-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 02/23/2022] [Indexed: 12/16/2022] Open
Abstract
Dry eye is a common ocular inflammatory disorder characterized by tear film instability and reduced tear production. There is increasing evidence that homeostasis of the ocular surface is impacted by the intestinal microbiome. We are interested in investigating the potential role of microbially produced small molecules in mediating the interaction between the intestinal microbiota and the ocular surface. One such molecule is butyrate, a short-chain fatty acid (SCFA) produced by certain members of the gut microbiota through fermentation of dietary fiber. Here we show that SCFA transporter SLC5A8 is expressed in vivo in murine conjunctival and corneal epithelium. Pre-treatment of in vitro corneal epithelial cultures or bone marrow-derived dendritic cells (BMDCs) with phenylbutyrate (PBA) reduces lipopolysaccharide-induced pro-inflammatory Tnf expression. Corneal epithelial cultures and BMDCs isolated from Slc5a8 knockout mice are unable to respond to PBA pre-treatment, suggesting that SLC5A8 is required for the protective effect of PBA. The treatment of mice undergoing desiccating stress (DS) with oral tributyrin, a prodrug form of butyrate, reduces inflammation at the ocular surface in vivo, and this effect partially requires SLC5A8. Finally, expression analysis on conjunctival tissue isolated from mice subjected to DS with and without tributyrin treatment revealed that treatment downregulated genes involved in Type I interferon signaling. Together these data support our hypothesis that SCFAs produced in the gut participate in the maintenance of ocular surface homeostasis.
Collapse
|
46
|
ŞİMŞEK C. Sjögren sendromuna bağlı gelişen kuru göz hastalığında meydana gelen oküler yüzey değişikliklerinin konfokal mikroskopi ile değerlendirilmesi. EGE TIP DERGISI 2022. [DOI: 10.19161/etd.1086117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
47
|
Trujillo-Vargas CM, Mauk KE, Hernandez H, de Souza RG, Yu Z, Galletti JG, Dietrich J, Paulsen F, de Paiva CS. Immune phenotype of the CD4 + T cells in the aged lymphoid organs and lacrimal glands. GeroScience 2022; 44:2105-2128. [PMID: 35279788 DOI: 10.1007/s11357-022-00529-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 02/11/2022] [Indexed: 02/07/2023] Open
Abstract
Aging is associated with a massive infiltration of T lymphocytes in the lacrimal gland. Here, we aimed to characterize the immune phenotype of aged CD4+ T cells in this tissue as compared with lymphoid organs. To perform this, we sorted regulatory T cells (Tregs, CD4+CD25+GITR+) and non-Tregs (CD4+CD25negGITRneg) in lymphoid organs from female C57BL/6J mice and subjected these cells to an immunology NanoString® panel. These results were confirmed by flow cytometry, live imaging, and tissue immunostaining in the lacrimal gland. Importantly, effector T helper 1 (Th1) genes were highly upregulated on aged Tregs, including the master regulator Tbx21. Among the non-Tregs, we also found a significant increase in the levels of EOMESmed/high, TbetnegIFN-γ+, and CD62L+CD44negCD4+ T cells with aging, which are associated with cell exhaustion, immunopathology, and the generation of tertiary lymphoid tissue. At the functional level, aged Tregs from lymphoid organs are less able to decrease proliferation and IFN-γ production of T responders at any age. More importantly, human lacrimal glands (age range 55-81 years) also showed the presence of CD4+Foxp3+ cells. Further studies are needed to propose potential molecular targets to avoid immune-mediated lacrimal gland dysfunction with aging.
Collapse
Affiliation(s)
- Claudia M Trujillo-Vargas
- Grupo de Inmunodeficiencias Primarias, Facultad de Medicina, Universidad de Antioquia, UdeA, Medellín, Colombia.,Department of Ophthalmology, Ocular Surface Center, Cullen Eye Institute, Baylor College of Medicine, 6565 Fannin Street, Houston, TX, NC 505G, USA
| | - Kelsey E Mauk
- Graduate Program in Immunology & Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Humberto Hernandez
- Department of Ophthalmology, Ocular Surface Center, Cullen Eye Institute, Baylor College of Medicine, 6565 Fannin Street, Houston, TX, NC 505G, USA
| | - Rodrigo G de Souza
- Department of Ophthalmology, Ocular Surface Center, Cullen Eye Institute, Baylor College of Medicine, 6565 Fannin Street, Houston, TX, NC 505G, USA
| | - Zhiyuan Yu
- Department of Ophthalmology, Ocular Surface Center, Cullen Eye Institute, Baylor College of Medicine, 6565 Fannin Street, Houston, TX, NC 505G, USA
| | - Jeremias G Galletti
- Institute of Experimental Medicine, CONICET-National Academy of Medicine of Buenos Aires, Buenos Aires, Argentina
| | - Jana Dietrich
- Institute of Functional and Clinical Anatomy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Friedrich Paulsen
- Institute of Functional and Clinical Anatomy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Cintia S de Paiva
- Department of Ophthalmology, Ocular Surface Center, Cullen Eye Institute, Baylor College of Medicine, 6565 Fannin Street, Houston, TX, NC 505G, USA.
| |
Collapse
|
48
|
Schaefer L, Trujillo-Vargas CM, Midani FS, Pflugfelder SC, Britton RA, de Paiva CS. Gut Microbiota From Sjögren syndrome Patients Causes Decreased T Regulatory Cells in the Lymphoid Organs and Desiccation-Induced Corneal Barrier Disruption in Mice. Front Med (Lausanne) 2022; 9:852918. [PMID: 35355610 PMCID: PMC8959809 DOI: 10.3389/fmed.2022.852918] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 02/11/2022] [Indexed: 12/12/2022] Open
Abstract
Sjögren syndrome (SS) is an autoimmune inflammatory disorder characterized by secretory dysfunction in the eye and mouth; in the eye, this results in tear film instability, reduced tear production, and corneal barrier disruption. A growing number of studies show that homeostasis of the ocular surface is impacted by the intestinal microbiome, and several 16S sequencing studies have demonstrated dysbiosis of the intestinal microbiota in SS patients. In this study, we utilized metagenomic sequencing to perform a deeper analysis of the intestinal microbiome using stools collected from sex- and age-matched healthy (n = 20), dry eye (n = 4) and SS (n = 7) subjects. The observed Operational Taxonomic Units (OTUs) and Shannon alpha diversity were significantly decreased in SS compared to healthy controls, and there was a significant inverse correlation between observed OTUs and ocular severity score. We also identified specific bacterial strains that are differentially modulated in SS vs. healthy subjects. To investigate if the differential composition of intestinal microbiome would have an impact on the immune and eye phenotype, we performed functional studies using germ-free mice colonized with human intestinal microbiota from SS patients and healthy controls. Flow cytometry analysis demonstrated reduced frequency of CD4+ FOXP3+ cells in ocular draining cervical lymph nodes (CLN) in mice colonized with SS patient intestinal microbiota 4 weeks post-colonization. We also found that offspring of SS-humanized mice also have fewer CD4+FOXP3+ cells in the CLN as well as spleen, demonstrating vertical transmission. SS-humanized mice subjected to desiccating stress exhibited greater corneal barrier disruption as compared to healthy control humanized mice under the same conditions. Taken together, these data support the hypothesis that the intestinal microbiota can modulate ocular surface health, possibly by influencing development of CD4+ FOXP3+ regulatory T cells (Tregs) in the ocular draining lymph nodes.
Collapse
Affiliation(s)
- Laura Schaefer
- Center of Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - Claudia M. Trujillo-Vargas
- Center of Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
- Department of Ophthalmology, Ocular Surface Center, Baylor College of Medicine, Cullen Eye Institute, Houston, TX, United States
- Grupo de Inmunodeficiencias Primarias, Facultad de Medicina, Universidad de Antioquia UdeA, Medellin, Colombia
| | - Firas S. Midani
- Center of Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - Stephen C. Pflugfelder
- Department of Ophthalmology, Ocular Surface Center, Baylor College of Medicine, Cullen Eye Institute, Houston, TX, United States
| | - Robert A. Britton
- Center of Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - Cintia S. de Paiva
- Department of Ophthalmology, Ocular Surface Center, Baylor College of Medicine, Cullen Eye Institute, Houston, TX, United States
| |
Collapse
|
49
|
Di Zazzo A, Coassin M, Surico PL, Bonini S. Age-related ocular surface failure: A narrative review. Exp Eye Res 2022; 219:109035. [DOI: 10.1016/j.exer.2022.109035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 02/28/2022] [Accepted: 03/13/2022] [Indexed: 12/26/2022]
|
50
|
Chen Y, Wang S, Alemi H, Dohlman T, Dana R. Immune regulation of the ocular surface. Exp Eye Res 2022; 218:109007. [PMID: 35257715 PMCID: PMC9050918 DOI: 10.1016/j.exer.2022.109007] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 02/10/2022] [Accepted: 02/20/2022] [Indexed: 01/01/2023]
Abstract
Despite constant exposure to various environmental stimuli, the ocular surface remains intact and uninflamed while maintaining the transparency of the cornea and its visual function. This 'immune privilege' of the ocular surface is not simply a result of the physical barrier function of the mucosal lining but, more importantly, is actively maintained through a variety of immunoregulatory mechanisms that prevent the disruption of immune homeostasis. In this review, we focus on essential molecular and cellular players that promote immune quiescence in steady-state conditions and suppress inflammation in disease-states. Specifically, we examine the interactions between the ocular surface and its local draining lymphoid compartment, by encompassing the corneal epithelium, corneal nerves and cornea-resident myeloid cells, conjunctival goblet cells, and regulatory T cells (Treg) in the context of ocular surface autoimmune inflammation (dry eye disease) and alloimmunity (corneal transplantation). A better understanding of the immunoregulatory mechanisms will facilitate the development of novel, targeted immunomodulatory strategies for a broad range of ocular surface inflammatory disorders.
Collapse
Affiliation(s)
- Yihe Chen
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA.
| | - Shudan Wang
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA
| | - Hamid Alemi
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA
| | - Thomas Dohlman
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA
| | - Reza Dana
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA
| |
Collapse
|