1
|
Lyu Y, Yin Q, Liao X, Xie Y, Yang H, Cui Y, Han Y, Yao K, Wang C, Shentu X. Aminated fullerene for comprehensive dry eye therapy: Promoting epithelial-barrier reconstruction and nerve regeneration by suppressing oxidation and inflammation. Biomaterials 2025; 321:123329. [PMID: 40273474 DOI: 10.1016/j.biomaterials.2025.123329] [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: 11/20/2024] [Revised: 03/14/2025] [Accepted: 04/07/2025] [Indexed: 04/26/2025]
Abstract
Dry eye disease (DED) affects up to 50 % of the global population, leading to serious discomforts that affect patients' quality of life. In the multifactorial etiology of DED, oxidative stress is at the core, initiating a sequence of inflammatory responses and surface damage via a vicious cycle. However, current therapies merely have a narrow focus on inflammation. In this study, we developed a novel antioxidative eye drop, ethylenediamine (EDA)-modified C70 fullerene derivatives (abbreviated as FN-EDA), to break this vicious cycle. FN-EDA was successfully synthesized by modifying C70 fullerene with multiple ethylenediamine (EDA) groups, resulting in enhanced water solubility and a positive charge. This modification significantly improved ocular surface retention time, cellular uptake, and lysosomal escape in vitro. Therapeutically, FN-EDA significantly alleviated dry eye disease (DED) in a mouse model. It reduced corneal epithelial damage by 3.8-fold compared to 0.05 % cyclosporine A (CsA) and restored tear secretion to approximately 65 % of the normal level. Mechanistically, both in vivo and in vitro results demonstrate that FN-EDA is endowed with superior biological activity in effectively scavenging excessive oxidative stress, down-regulating proinflammatory cytokines expression, and promoting epithelial barrier reconstruction, even recovering corneal innervation. Thus, our findings open an avenue to make this multi-functional eye drop a promising candidate for DED.
Collapse
Affiliation(s)
- Ying Lyu
- Zhejiang University, Eye Center of Second Affiliated Hospital, School of Medicine, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, 310009, China.
| | - Qichuan Yin
- Zhejiang University, Eye Center of Second Affiliated Hospital, School of Medicine, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, 310009, China.
| | - Xiaodan Liao
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China.
| | - Youxuan Xie
- Zhejiang University, Eye Center of Second Affiliated Hospital, School of Medicine, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, 310009, China.
| | - Hao Yang
- Zhejiang University, Eye Center of Second Affiliated Hospital, School of Medicine, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, 310009, China.
| | - Yilei Cui
- Zhejiang University, Eye Center of Second Affiliated Hospital, School of Medicine, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, 310009, China.
| | - Yuqi Han
- Zhejiang University, Eye Center of Second Affiliated Hospital, School of Medicine, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, 310009, China.
| | - Ke Yao
- Zhejiang University, Eye Center of Second Affiliated Hospital, School of Medicine, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, 310009, China.
| | - Chunru Wang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China.
| | - Xingchao Shentu
- Zhejiang University, Eye Center of Second Affiliated Hospital, School of Medicine, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, 310009, China.
| |
Collapse
|
2
|
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. [PMID: 40308890 PMCID: PMC12038461 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
|
3
|
Gehlsen U, Maass M, Stary D, Wagener-Ryczek S, Musial G, Pasparakis M, de Paiva CS, Stern ME, Steven P. Desiccation stress triggers and exacerbates experimental ocular Graft-versus-host-disease. Ocul Surf 2025; 37:236-246. [PMID: 40287060 DOI: 10.1016/j.jtos.2025.04.008] [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: 02/06/2025] [Revised: 04/10/2025] [Accepted: 04/13/2025] [Indexed: 04/29/2025]
Abstract
INTRODUCTION Chronic ocular graft-versus-host disease (oGVHD) is one of the most common complications after allogeneic hematopoietic stem cell transplantation (aHSCT). Recent studies indicate that desiccating stress by air-conditioning in transplantation wards increases the incidence of oGVHD. To test the hypothesis that experimental desiccating stress is a risk factor for oGVHD a mouse model of oGVHD was subjected to experimental desiccating stress. MATERIALS/METHODS A previously established chemo-induced minor-mismatch mouse model of oGVHD was used. One group was challenged with desiccating stress for 18 days and compared to non-desiccated GVHD animals. Clinical phenotyping was performed weekly and ocular tissue and regional lymph nodes were collected on days 7 and 28 for flow-cytometry, tear film cytokine analysis, histology for corneal lymphatics and dendritic cell counts, and corneal gene expression. RESULTS Desiccating stress leads to significant earlier and more severe systemic and oGVHD accompanied by higher numbers of activated corneal dendritic cells, higher expression of TNF in tear film and earlier corneal lymphangiogenesis. Gene expression analysis suggests that systemic GVHD severity may influence oGVHD. Different inflammatory pathways are upregulated at d28 following desiccating stress in contrast to non-desiccated GVHD. CONCLUSIONS The data presented strengthens the hypothesis, that desiccating stress during aHSCT is a risk factor for oGVHD. Together with already published clinical data, there is increasing evidence that implicates protecting patients from desiccation during the engraftment of allogeneic hematopoietic stem cells. Furthermore, specific prophylactic therapies should be developed and tested to reduce the incidence and severity of oGVHD.
Collapse
Affiliation(s)
- Uta Gehlsen
- Competence Center for Ocular GVHD, Center of Integrated Oncology, Department I of Internal Medicine and Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, Germany
| | - Martina Maass
- Competence Center for Ocular GVHD, Center of Integrated Oncology, Department I of Internal Medicine and Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, Germany
| | - Daniela Stary
- Competence Center for Ocular GVHD, Center of Integrated Oncology, Department I of Internal Medicine and Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, Germany
| | - Svenja Wagener-Ryczek
- Institute of Pathology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Germany
| | - Gwen Musial
- Competence Center for Ocular GVHD, Center of Integrated Oncology, Department I of Internal Medicine and Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, Germany
| | - Manolis Pasparakis
- Institute for Genetics, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany
| | - Cintia S de Paiva
- Ocular Surface Center at the Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, TX, USA
| | - Michael E Stern
- Competence Center for Ocular GVHD, Center of Integrated Oncology, Department I of Internal Medicine and Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, Germany
| | - Philipp Steven
- Competence Center for Ocular GVHD, Center of Integrated Oncology, Department I of Internal Medicine and Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, Germany.
| |
Collapse
|
4
|
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
|
5
|
Zhang M, Liang Y, Wu H, Zong R, Zhang X, He H, Reinach PS, Liu Z, Shen L, Li W. Ocular Surface Involvements in the Development of Sjögren's Syndrome-Associated Dry Eye in the IL14α Transgenic Mouse. Invest Ophthalmol Vis Sci 2025; 66:2. [PMID: 40029244 PMCID: PMC11887930 DOI: 10.1167/iovs.66.3.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 01/23/2025] [Indexed: 03/05/2025] Open
Abstract
Purpose To investigate the ocular surface changes during progress of the Sjögren's Syndrome (SS), using a previously described IL14α transgenic mice (IL14α TG) SS model. Methods The ocular surface of IL14α TG and C57BL/6 wild-type (WT) female mice were evaluated at the age of six, nine, 12, 15, and 18 months. Slit lamp microscopy observation, Oregon green dextran staining, Schirmer test, and periodic-acid-Schiff staining were assessed. Immunohistochemistry, immunofluorescence, and associated gene expression analysis by qPCR and ELISA were performed in cornea, conjunctiva, and lacrimal grand at different ages of the mice. Masson's trichome staining was conducted on lacrimal gland cryosections. Results Compared with C57BL/6 WT mice, IL14α TG mice showed corneal barrier function damage and losses in conjunctival goblet cell density starting at nine months, whereas decreases in tear secretion started at 18 months of age. Significant increases in CD4+ T cell infiltration in the conjunctiva of IL14α TG mice was first observed at 6 months. Higher expression levels of inflammatory cytokines IL-17A, IFN-γ, IL-1β, and TNF-α in the conjunctiva, whereas MUC5AC and MUC5B had lower expression levels at nine months in the IL14α TG mice. However, lacrimal gland function-associated gene expression levels mostly decreased in IL14α TG mice at 12 months of age. Conclusions Ocular surface tissue changes were involved in SS-like dry eye in a time-dependent manner in IL14α TG mice, and conjunctival T-cell infiltration may contribute to ocular surface pathological changes in an early stage of SS-related dry eye.
Collapse
Affiliation(s)
- Minjie Zhang
- Xiamen University affiliated Xiamen Eye Center; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science; Eye Institute of Xiamen University; School of Medicine, Xiamen University, Xiamen, Fujian, China
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, China
- Department of Ophthalmology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Yichen Liang
- Oncology Department, Northern Jiangsu People's Hospital, Yangzhou, Jiangsu, China
- Cancer Institute affiliated to Northern Jiangsu People's Hospital, Yangzhou, Jiangsu, China
- Medical College, Yangzhou University, Yangzhou, Jiangsu, China
| | - Han Wu
- Xiamen University affiliated Xiamen Eye Center; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science; Eye Institute of Xiamen University; School of Medicine, Xiamen University, Xiamen, Fujian, China
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, China
| | - Rongrong Zong
- Xiamen University affiliated Xiamen Eye Center; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science; Eye Institute of Xiamen University; School of Medicine, Xiamen University, Xiamen, Fujian, China
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, China
| | - Xiaobo Zhang
- C-MER (Shenzhen) Dennis Lam Eye Hospital, Shenzhen, Guangdong, China
| | - Hui He
- Xiamen University affiliated Xiamen Eye Center; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science; Eye Institute of Xiamen University; School of Medicine, Xiamen University, Xiamen, Fujian, China
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, China
| | - Peter Sol Reinach
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zuguo Liu
- Xiamen University affiliated Xiamen Eye Center; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science; Eye Institute of Xiamen University; School of Medicine, Xiamen University, Xiamen, Fujian, China
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, China
| | - Long Shen
- Oncology Department, Northern Jiangsu People's Hospital, Yangzhou, Jiangsu, China
- Cancer Institute affiliated to Northern Jiangsu People's Hospital, Yangzhou, Jiangsu, China
- Medical College, Yangzhou University, Yangzhou, Jiangsu, China
| | - Wei Li
- Xiamen University affiliated Xiamen Eye Center; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science; Eye Institute of Xiamen University; School of Medicine, Xiamen University, Xiamen, Fujian, China
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, China
| |
Collapse
|
6
|
Truong DV, Yang BS, Song C. Anti-inflammatory effects of LCB 03-0110 on human corneal epithelial and murine T helper 17 cells. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2025; 29:205-214. [PMID: 39539175 PMCID: PMC11842296 DOI: 10.4196/kjpp.24.166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 08/08/2024] [Accepted: 08/27/2024] [Indexed: 11/16/2024]
Abstract
Dry eye disease (DED) is a complicated disorder that impacts ocular surface and tear-film stability. Inflammation has recently been reported as the core mechanism and main therapeutic target of DED. Although anti-inflammatory drugs have been developed, they still have limited efficacy and various side effects. Recent reports have suggested that kinase inhibitors are beneficial for relieving inflammation. Therefore, this study aimed to investigate the anti-inflammatory effects of LCB 03-0110, a multi-tyrosine kinase inhibitor, on representative cell-based models (HCE- 2 and Th17 cells) of DED. While tacrolimus and tofacitinib, two different anti-inflammatory drugs that have entered clinical trials for DED treatment, did not induce any anti-inflammatory responses in HCE-2 cells, LCB 03-0110 significantly suppressed the phosphorylation of P38 and ERK and reduced the expression levels of IL-6 and IL-8 in HCE-2 cells treated with either LPS or poly(I:C). Moreover, LCB 03-0110 notably decreased the expression level of IL-17A in Th17 cells in a dose-dependent manner, whereas tofacitinib promoted IL-17A production at low concentrations but inhibited its expression at concentrations greater than 1 μM. In addition, LCB 03-0110 was found to be non-toxic to both HCE-2 and Th17 cells. In conclusion, these results suggest that LCB 03-0110 would be a promising drug candidate for the treatment of DED because of its advantages over tacrolimus and tofacitinib.
Collapse
Affiliation(s)
- Do Vinh Truong
- 1Division of Bio-Medical Science & Technology, KIST School, University of Science and Technology, Seoul 02792, Korea
- Institute of Biotechnology, Vietnam Academy of Science and Technology, Hanoi 10072, Vietnam
| | - Beom-Seok Yang
- 1Division of Bio-Medical Science & Technology, KIST School, University of Science and Technology, Seoul 02792, Korea
| | - Chiman Song
- 1Division of Bio-Medical Science & Technology, KIST School, University of Science and Technology, Seoul 02792, Korea
- Chemical & Biological Integrative Research Center, Korea Institute of Science and Technology, Seoul 02792, Korea
| |
Collapse
|
7
|
Moon J, Jiang E, Liu J, Jin H, Yoon HS, Choi HI, Choi JS, Qi H, Yoon HJ, Yoon KC. Therapeutic Potential of Combined 5% Lifitegrast and Tocopherol Eye Drops in Managing Inflammation and Oxidative Stress in Murine Dry Eye. Pharmaceuticals (Basel) 2025; 18:38. [PMID: 39861101 PMCID: PMC11769320 DOI: 10.3390/ph18010038] [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: 11/20/2024] [Revised: 12/27/2024] [Accepted: 12/28/2024] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: This study aimed to evaluate the therapeutic effects of combined 5% lifitegrast (LF) and tocopherol (TCP) eye drops in a murine experimental dry eye (EDE) model. Methods: Female C57BL/6 were divided into seven groups: untreated controls, EDE control, EDE + 0.05% cyclosporin A (CsA), EDE + tocopherol (TCP), EDE + 5% LF, EDE + 5% LF + TCP (once daily), and EDE + 5% LF + TCP (twice daily). Clinical parameters (tear volume, tear break-up time (TBUT), corneal fluorescein staining score (CFSS), tear film lipid layer grade (TFLLG)) were assessed on days 7 and 14. Goblet cell density in the conjunctiva, CD4+ IFN-γ+ T cells, interleukin levels, reactive oxygen species (ROS) levels, and corneal apoptotic cells were analyzed on day 14. Results: Monotherapy with 0.05% CsA and LF showed improvements in all clinical parameters compared to the EDE control (p < 0.05). Combination therapy groups demonstrated superior improvements in clinical parameters compared to the EDE control, 0.05% CsA, and 5% LF groups. CD4+ IFN-γ+ T cell percentages and ROS levels in the cornea and conjunctiva were markedly reduced in the combination groups compared with the 0.05% CsA and 5% LF groups (p < 0.01). Furthermore, corneal apoptotic cells significantly decreased in the combination groups compared to the 0.05% CsA and TCP groups (p < 0.05). Conclusions: Combined 5% LF and TCP eye drops improved tear film parameters and reduced inflammatory and oxidative stress markers. The combination therapy can mitigate ocular surface damage by managing inflammation and oxidative stress in dry eye.
Collapse
Affiliation(s)
- Jayoung Moon
- Department of Ophthalmology, Chonnam National University Medical School and Hospital, Gwangju 61469, Republic of Korea (J.L.); (H.S.Y.); (H.-I.C.); (J.S.C.); (H.-J.Y.)
| | - Enying Jiang
- Department of Ophthalmology, Chonnam National University Medical School and Hospital, Gwangju 61469, Republic of Korea (J.L.); (H.S.Y.); (H.-I.C.); (J.S.C.); (H.-J.Y.)
| | - Jingting Liu
- Department of Ophthalmology, Chonnam National University Medical School and Hospital, Gwangju 61469, Republic of Korea (J.L.); (H.S.Y.); (H.-I.C.); (J.S.C.); (H.-J.Y.)
| | - Hui Jin
- Department of Ophthalmology, Chonnam National University Medical School and Hospital, Gwangju 61469, Republic of Korea (J.L.); (H.S.Y.); (H.-I.C.); (J.S.C.); (H.-J.Y.)
| | - Hee Su Yoon
- Department of Ophthalmology, Chonnam National University Medical School and Hospital, Gwangju 61469, Republic of Korea (J.L.); (H.S.Y.); (H.-I.C.); (J.S.C.); (H.-J.Y.)
| | - Hoon-In Choi
- Department of Ophthalmology, Chonnam National University Medical School and Hospital, Gwangju 61469, Republic of Korea (J.L.); (H.S.Y.); (H.-I.C.); (J.S.C.); (H.-J.Y.)
| | - Ji Suk Choi
- Department of Ophthalmology, Chonnam National University Medical School and Hospital, Gwangju 61469, Republic of Korea (J.L.); (H.S.Y.); (H.-I.C.); (J.S.C.); (H.-J.Y.)
| | - Hong Qi
- Department of Ophthalmology, Peking University Third Hospital, Beijing 100191, China;
- Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Beijing 100191, China
| | - Hyeon-Jeong Yoon
- Department of Ophthalmology, Chonnam National University Medical School and Hospital, Gwangju 61469, Republic of Korea (J.L.); (H.S.Y.); (H.-I.C.); (J.S.C.); (H.-J.Y.)
| | - Kyung Chul Yoon
- Department of Ophthalmology, Chonnam National University Medical School and Hospital, Gwangju 61469, Republic of Korea (J.L.); (H.S.Y.); (H.-I.C.); (J.S.C.); (H.-J.Y.)
| |
Collapse
|
8
|
Harrell CR, Volarevic V. Ion Channels as Potential Drug Targets in Dry Eye Disease and Their Clinical Relevance: A Review. Cells 2024; 13:2017. [PMID: 39682765 PMCID: PMC11639998 DOI: 10.3390/cells13232017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 10/17/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
Dry eye disease (DED) is a common multifactorial disorder characterized by a deficiency in the quality and/or quantity of tear fluid. Tear hyperosmolarity, the dysfunction of ion channel proteins, and eye inflammation are primarily responsible for the development and progression of DED. Alterations in the structure and/or function of ion channel receptors (transient receptor potential ankyrin 1 (TRPA1), transient receptor potential melastatin 8 (TRPM8), transient receptor potential vanilloid 1 and 4 (TRPV1 and TRPV4)), and consequent hyperosmolarity of the tears represent the initial step in the development and progression of DED. Hyperosmolarity triggers the activation of ion channel-dependent signaling pathways in corneal epithelial cells and eye-infiltrated immune cells, leading to the activation of transcriptional factors that enhance the expression of genes regulating inflammatory cytokine production, resulting in a potent inflammatory response in the eyes of DED patients. A persistent and untreated detrimental immune response further modifies the structure and function of ion channel proteins, perpetuating tear hyperosmolarity and exacerbating DED symptoms. Accordingly, suppressing immune cell-driven eye inflammation and alleviating tear hyperosmolarity through the modulation of ion channels in DED patients holds promise for developing new therapeutic strategies. Here, we summarize current knowledge about the molecular mechanisms responsible for the inflammation-induced modification of ion channels leading to tear hyperosmolarity and immune cell dysfunction in DED patients. We also emphasize the therapeutic potential of the newly designed immunomodulatory and hypo-osmotic solution d-MAPPS™ Hypo-Osmotic Ophthalmic Solution, which can activate TRPV4 in corneal epithelial cells, stabilize the tear film, enhance natural cytokine communication, and suppress detrimental immune responses, an important novel approach for DED treatment.
Collapse
Affiliation(s)
| | - Vladislav Volarevic
- Center for Research on Harmful Effects of Biological and Chemical Hazards, Departments of Genetics, Microbiology and Immunology, Faculty of Medical Sciences, University of Kragujevac, 69 Svetozar Markovic Street, 34000 Kragujevac, Serbia;
- Faculty of Pharmacy Novi Sad, Heroja Pinkija 4, 21000 Novi Sad, Serbia
| |
Collapse
|
9
|
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
|
10
|
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
|
11
|
Vereertbrugghen A, Pizzano M, Cernutto A, Sabbione F, Keitelman IA, Aguilar DV, Podhorzer A, Fuentes F, Corral-Vázquez C, Guzmán M, Giordano MN, Trevani A, de Paiva CS, Galletti JG. CD4 + T cells drive corneal nerve damage but not epitheliopathy in an acute aqueous-deficient dry eye model. Proc Natl Acad Sci U S A 2024; 121:e2407648121. [PMID: 39560641 PMCID: PMC11621630 DOI: 10.1073/pnas.2407648121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 10/14/2024] [Indexed: 11/20/2024] Open
Abstract
Dry eye disease (DED) is characterized by a dysfunctional tear film in which the corneal epithelium and its abundant nerves are affected by ocular desiccation and inflammation. Although adaptive immunity and specifically CD4+ T cells play a role in DED pathogenesis, the exact contribution of these cells to corneal epithelial and neural damage remains undetermined. To address this, we explored the progression of a surgical DED model in wild-type (WT) and T cell-deficient mice. We observed that adaptive immune-deficient mice developed all aspects of DED comparably to WT mice except for the absence of functional and morphological corneal nerve changes, nerve damage-associated transcriptomic signature in the trigeminal ganglia, and sustained tear cytokine levels. Adoptive transfer of CD4+ T cells from WT DED mice to T cell-deficient mice reproduced corneal nerve damage but not epitheliopathy. Conversely, T cell-deficient mice reconstituted solely with naïve CD4+ T cells developed corneal nerve impairment and epitheliopathy upon DED induction, thus replicating the WT DED phenotype. Collectively, our data show that while corneal neuropathy is driven by CD4+ T cells in DED, corneal epithelial damage develops independently of the adaptive immune response. These findings have implications for T cell-targeting therapies currently in use for DED.
Collapse
Affiliation(s)
- Alexia Vereertbrugghen
- Innate Immunity Laboratory, Institute of Experimental Medicine (National Scientific and Technical Research Council/National Academy of Medicine of Buenos Aires), Buenos Aires1425, Argentina
| | - Manuela Pizzano
- Innate Immunity Laboratory, Institute of Experimental Medicine (National Scientific and Technical Research Council/National Academy of Medicine of Buenos Aires), Buenos Aires1425, Argentina
| | - Agostina Cernutto
- Innate Immunity Laboratory, Institute of Experimental Medicine (National Scientific and Technical Research Council/National Academy of Medicine of Buenos Aires), Buenos Aires1425, Argentina
| | - Florencia Sabbione
- Innate Immunity Laboratory, Institute of Experimental Medicine (National Scientific and Technical Research Council/National Academy of Medicine of Buenos Aires), Buenos Aires1425, Argentina
| | - Irene A. Keitelman
- Innate Immunity Laboratory, Institute of Experimental Medicine (National Scientific and Technical Research Council/National Academy of Medicine of Buenos Aires), Buenos Aires1425, Argentina
| | - Douglas Vera Aguilar
- Innate Immunity Laboratory, Institute of Experimental Medicine (National Scientific and Technical Research Council/National Academy of Medicine of Buenos Aires), Buenos Aires1425, Argentina
| | - Ariel Podhorzer
- Flow Cytometry Unit, Institute of Experimental Medicine (National Scientific and Technical Research Council/National Academy of Medicine of Buenos Aires), Buenos Aires1425, Argentina
| | - Federico Fuentes
- Confocal Microscopy Unit, Institute of Experimental Medicine (National Scientific and Technical Research Council/National Academy of Medicine of Buenos Aires), Buenos Aires1425, Argentina
| | - Celia Corral-Vázquez
- Translational Clinical Research Program, Hospital del Mar Research Institute, Barcelona08003, Spain
| | - Mauricio Guzmán
- Translational Clinical Research Program, Hospital del Mar Research Institute, Barcelona08003, Spain
| | - Mirta N. Giordano
- Innate Immunity Laboratory, Institute of Experimental Medicine (National Scientific and Technical Research Council/National Academy of Medicine of Buenos Aires), Buenos Aires1425, Argentina
| | - Analía Trevani
- Innate Immunity Laboratory, Institute of Experimental Medicine (National Scientific and Technical Research Council/National Academy of Medicine of Buenos Aires), Buenos Aires1425, Argentina
| | | | - Jeremías G. Galletti
- Innate Immunity Laboratory, Institute of Experimental Medicine (National Scientific and Technical Research Council/National Academy of Medicine of Buenos Aires), Buenos Aires1425, Argentina
| |
Collapse
|
12
|
Alam J, Yaman E, de Paiva CS, Li DQ, Villalba Silva GC, Zuo Z, Pflugfelder SC. Changes in conjunctival mononuclear phagocytes and suppressive activity of regulatory macrophages in desiccation induced dry eye. Ocul Surf 2024; 34:348-362. [PMID: 39306240 PMCID: PMC11984642 DOI: 10.1016/j.jtos.2024.09.003] [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: 01/12/2024] [Revised: 09/07/2024] [Accepted: 09/10/2024] [Indexed: 09/27/2024]
Abstract
PURPOSE To evaluate the effects of dry eye on conjunctival immune cell number and transcriptional profiles with attention to mononuclear phagocytes. METHODS Expression profiling was performed by single-cell RNA sequencing on sorted conjunctival immune cells from non-stressed and C57BL/6 mice subjected to desiccating stress (DS). Monocle 3 modeled cell trajectory, scATAC-seq assessed chromatin accessibility and IPA identified canonical pathways. Inflammation and goblet cells were measured after depletion of MRC1+ MΦs with mannosylated clodronate liposomes. RESULTS Mononuclear phagocytes (monocytes, MΦs, DCs) comprised 72 % of immune cells and showed the greatest changes with DS. Distinct DS induced gene expression patterns were seen in phagocytes classified by expression of Ccr2 and [Timd4, Lyve1, Folr2 (TLR)]. Expression of phagocytosis/efferocytosis genes increased in TLF+CCR2- MΦs. Monocytes showed the highest expression of Ace, Cx3cr1, Vegfa, Ifngr1,2, and Stat1 and TLF-CCR2+ cells expressed higher levels of inflammatory mediators (Il1a, Il1b, Il1rn, Nfkb1, Ccl5, MHCII, Cd80, Cxcl10, Icam1). A trajectory from monocyte precursors branched to terminate in regulatory MΦs or in mDCs via transitional MΦ and cDC clusters. Activated pathways in TLF+ cells include phagocytosis, PPAR/RXRα activation, IL-10 signaling, alternate MΦ activation, while inflammatory pathways were suppressed. Depletion of MRC1+ MΦs increased IL-17 and IFN-γ expression and cytokine-expressing T cells, reduced IL-10 and worsened goblet loss. CONCLUSIONS Dryness stimulates distinct gene expression patterns in conjunctival phagocytes, increasing expression of regulatory genes in TLF+ cells regulated in part by RXRα, and inflammatory genes in CCR2+ cells. Regulatory MΦs depletion worsens DS induced inflammation and goblet cell loss.
Collapse
Affiliation(s)
- Jehan Alam
- Ocular Surface Center, Department of Ophthalmology, Baylor College of Medicine, Houston, TX, USA
| | - Ebru Yaman
- Ocular Surface Center, Department of Ophthalmology, Baylor College of Medicine, Houston, TX, USA
| | - Cintia S de Paiva
- Ocular Surface Center, Department of Ophthalmology, Baylor College of Medicine, Houston, TX, USA
| | - De-Quan Li
- Ocular Surface Center, Department of Ophthalmology, Baylor College of Medicine, Houston, TX, USA
| | - Gerda Cristal Villalba Silva
- Human Genome Sequencing Center, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Zhen Zuo
- Human Genome Sequencing Center, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Stephen C Pflugfelder
- Ocular Surface Center, Department of Ophthalmology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
13
|
Xie M, Wu Y, Zhang Y, Lu R, Zhai Z, Huang Y, Wang F, Xin C, Rong G, Zhao C, Jiang K, Zhou X, Zhou X, Zhu X, Hong J, Zhang C. Membrane Fusion-Mediated Loading of Therapeutic siRNA into Exosome for Tissue-Specific Application. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2403935. [PMID: 38889294 DOI: 10.1002/adma.202403935] [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: 03/17/2024] [Revised: 05/30/2024] [Indexed: 06/20/2024]
Abstract
Tissue-specific delivery of oligonucleotide therapeutics beyond the liver remains a key challenge in nucleic acid drug development. To address this issue, exploiting exosomes as a novel carrier has emerged as a promising approach for efficient nucleic acid drug delivery. However, current exosome-based delivery systems still face multiple hurdles in their clinical applications. Herein, this work presents a strategy for constructing a hybrid exosome vehicle (HEV) through a DNA zipper-mediated membrane fusion approach for tissue-specific siRNA delivery. As a proof-of-concept, this work successfully fuses a liposome encapsulating anti-NFKBIZ siRNAs with corneal epithelium cell (CEC)-derived exosomes to form a HEV construct for the treatment of dry eye disease (DED). With homing characteristics inherited from exosomes, the siRNA-bearing HEV can target its parent cells and efficiently deliver the siRNA payloads to the cornea. Subsequently, the NFKBIZ gene silencing significantly reduces pro-inflammatory cytokine secretions from the ocular surface, reshapes its inflammatory microenvironment, and ultimately achieves an excellent therapeutic outcome in a DED mouse model. As a versatile platform, this hybrid exosome with targeting capability and designed therapeutic siRNAs may hold great potential in various disease treatments.
Collapse
Affiliation(s)
- Miao Xie
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Yuqing Wu
- Department of Ophthalmology, Eye & ENT Hospital, State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200031, P. R. China
| | - Yilun Zhang
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Ruiyang Lu
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Zimeng Zhai
- Department of Ophthalmology, Eye & ENT Hospital, State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200031, P. R. China
| | - Yangyang Huang
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Fujun Wang
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Changchang Xin
- Department of Ophthalmology, Eye & ENT Hospital, State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200031, P. R. China
| | - Guangyu Rong
- Department of Ophthalmology, Eye & ENT Hospital, State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200031, P. R. China
| | - Chen Zhao
- Department of Ophthalmology, Eye & ENT Hospital, State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200031, P. R. China
| | - Kai Jiang
- Department of Ophthalmology, Eye & ENT Hospital, State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200031, P. R. China
| | - Xujiao Zhou
- Department of Ophthalmology, Eye & ENT Hospital, State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200031, P. R. China
| | - Xingtao Zhou
- Department of Ophthalmology, Eye & ENT Hospital, State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200031, P. R. China
| | - Xinyuan Zhu
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Jiaxu Hong
- Department of Ophthalmology, Eye & ENT Hospital, State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200031, P. R. China
- Department of Ophthalmology, Children's Hospital of Fudan University, Shanghai, 201102, P. R. China
- Key Laboratory of Myopia and Related Eye Diseases, NHC, Shanghai, 200031, P. R. China
- Shanghai Engineering Research Center for Synthetic Immunology, Shanghai, 200032, P. R. China
| | - Chuan Zhang
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| |
Collapse
|
14
|
Tang Y, Qu S, Ning Z, Wu H. Immunopeptides: immunomodulatory strategies and prospects for ocular immunity applications. Front Immunol 2024; 15:1406762. [PMID: 39076973 PMCID: PMC11284077 DOI: 10.3389/fimmu.2024.1406762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 07/01/2024] [Indexed: 07/31/2024] Open
Abstract
Immunopeptides have low toxicity, low immunogenicity and targeting, and broad application prospects in drug delivery and assembly, which are diverse in application strategies and drug combinations. Immunopeptides are particularly important for regulating ocular immune homeostasis, as the eye is an immune-privileged organ. Immunopeptides have advantages in adaptive immunity and innate immunity, treating eye immune-related diseases by regulating T cells, B cells, immune checkpoints, and cytokines. This article summarizes the application strategies of immunopeptides in innate immunity and adaptive immunity, including autoimmunity, infection, vaccine strategies, and tumors. Furthermore, it focuses on the mechanisms of immunopeptides in mediating ocular immunity (autoimmune diseases, inflammatory storms, and tumors). Moreover, it reviews immunopeptides' application strategies and the therapeutic potential of immunopeptides in the eye. We expect the immune peptide to get attention in treating eye diseases and to provide a direction for eye disease immune peptide research.
Collapse
Affiliation(s)
| | | | | | - Hong Wu
- Eye Center of Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
15
|
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
|
16
|
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
|
17
|
Ma K, Zhou H, Zhang W, Liu J, Sha Y, Kong X, Ni Z, Xiang M. Study of IL-17 and Intercellular Adhesion Molecule-1 in Conjunctivochalasis Using Correlation Analysis. Cornea 2024; 43:537-544. [PMID: 38305353 PMCID: PMC10980173 DOI: 10.1097/ico.0000000000003480] [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: 02/15/2023] [Revised: 10/30/2023] [Accepted: 12/12/2023] [Indexed: 02/03/2024]
Abstract
PURPOSE The aim of this study was to observe the expression of interleukin (IL)-17 and intercellular adhesion molecule (ICAM)-1 in conjunctivochalasis (CCH) and to analyze the correlations between cytokines and the severity of CCH. METHODS Serum samples were collected from 22 patients with CCH and 18 normal controls (NCs). The Ocular Surface Disease Index, tear film break-up time, Schirmer I test, and corneal fluorescein staining were used to evaluate the ocular surface signs and symptoms. The concentrations of IL-17, IL-23, and ICAM-1 in serum and cellular supernatants were measured by enzyme-linked immunosorbent assays, and the gene expression levels of cytokines were measured by a quantitative real-time polymerase chain reaction. The relationships between serum concentrations of IL-17, IL-23, and ICAM-1 with clinical ocular surface parameters in CCH were analyzed using the Spearman correlation analysis. RESULTS The concentrations of IL-17 and ICAM-1 in serum and cellular supernatants of CCH were significantly higher than those of NCs (all P < 0.001). The concentrations of IL-23 in serum and cellular supernatants of CCH showed no significant difference from those of NCs ( P > 0.05). The mRNA expression levels of IL-17 and ICAM-1 in conjunctival fibroblasts of CCH were significantly higher than those of NCs (all P < 0.001). The mRNA expression of IL-23 in conjunctival fibroblasts of CCH was higher than that of NCs, without a significant difference ( P > 0.05). Furthermore, the serum concentrations of IL-17 and ICAM-1 were positively correlated with Ocular Surface Disease Index and fluorescein staining (all P < 0.05), and negatively correlated with break-up time and Schirmer I test of CCH (all P < 0.05). CONCLUSIONS The expression levels of IL-17 and ICAM-1 were significantly increased in CCH serum and associated with the disease severity. We postulate that IL-17 and ICAM-1 may play a role in the pathogenesis of CCH. IL-17 and ICAM-1 antagonists may be a potential treatment option for CCH in the future.
Collapse
Affiliation(s)
- Kai Ma
- Department of Ophthalmology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China;
| | - Huanming Zhou
- Department of Ophthalmology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China;
| | - Wei Zhang
- Department of Ophthalmology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China;
| | - Jiang Liu
- Department of Ophthalmology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China;
| | - Yongyi Sha
- Department of Ophthalmology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China;
| | - Xueqing Kong
- Department of Ophthalmology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China;
| | - Zhenhua Ni
- Department of Central Lab, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China; and
| | - Minhong Xiang
- Department of Ophthalmology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China;
- Shanghai Putuo Central School of Clinical Medicine, Anhui Medical University, Hefei, China.
| |
Collapse
|
18
|
Sen M, Eroğul Ö. Retinoic Acid Neutralizes the Effects of Herpes Simplex Virus Type 1-Infected Cell Protein 0 (ICP0) in Retinal Pigment Epithelial Cells. Cureus 2024; 16:e61089. [PMID: 38919217 PMCID: PMC11196970 DOI: 10.7759/cureus.61089] [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: 05/25/2024] [Indexed: 06/27/2024] Open
Abstract
BACKGROUND Herpes simplex virus (HSV) infection of the cornea, uvea, and retina is the leading infectious cause of blindness worldwide. This study examined the effects of retinoic acid (RA) on the protein levels of interleukin (IL)-17A and IL-23 cytokines with known proinflammatory effects and toll-like receptor 3 (TLR3) messenger RNA (mRNA) expression in retinal pigment epithelial (ARPE-19) cells treated with HSV-1-infected cell protein 0 (ICP0). METHODOLOGY We used 3-[4.5-dimethylthiazol-2-yl]-2.5-diphenyl tetrazolium bromide assay to calculate the half maximal inhibitory concentration (IC50) doses of RA and ICP0 in ARPE-19 cells. At the end of 24 hours, protein levels of IL-17A and IL-23 were analyzed using enzyme-linked immunosorbent assay. TLR3 mRNA expression levels were also calculated using reverse transcription-polymerase chain reaction (RT-PCR). RESULTS RA administration decreased IL-17A levels, which were elevated by ICP0. The IL-23 levels were similar between the ICP0-treated and control groups, but the difference was significant between the ICP0-treated group and RA+ICP0 combination. These results showed that RA can significantly increase IL-23 levels in the presence of ICP0. Although ICP0 dramatically increased TLR3 mRNA expression compared with that in the control group, the RA+ICP0 combination returned TLR3 mRNA expression to a level similar to that in the control group (P = 0.419). CONCLUSIONS RA may potentially neutralize HSV-1 ICP0 negative effects in ARPE-19 cells.
Collapse
Affiliation(s)
- Merve Sen
- Department of Medical Services and Techniques, Suhut Vocational School of Health Services, Afyonkarahısar Health Sciences University, Afyonkarahisar, TUR
| | - Özgür Eroğul
- Department of Opthalmology, Faculty of Medicine, Afyonkarahisar Health Science University, Afyonkarahisar, TUR
| |
Collapse
|
19
|
Labetoulle M, Baudouin C, Benitez Del Castillo JM, Rolando M, Rescigno M, Messmer EM, Aragona P. How gut microbiota may impact ocular surface homeostasis and related disorders. Prog Retin Eye Res 2024; 100:101250. [PMID: 38460758 DOI: 10.1016/j.preteyeres.2024.101250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 03/01/2024] [Accepted: 03/04/2024] [Indexed: 03/11/2024]
Abstract
Changes in the bacterial flora in the gut, also described as gut microbiota, are readily acknowledged to be associated with several systemic diseases, especially those with an inflammatory, neuronal, psychological or hormonal factor involved in the pathogenesis and/or the perception of the disease. Maintaining ocular surface homeostasis is also based on all these four factors, and there is accumulating evidence in the literature on the relationship between gut microbiota and ocular surface diseases. The mechanisms involved are mostly interconnected due to the interaction of central and peripheral neuronal networks, inflammatory effectors and the hormonal system. A better understanding of the influence of the gut microbiota on the maintenance of ocular surface homeostasis, and on the onset or persistence of ocular surface disorders could bring new insights and help elucidate the epidemiology and pathology of ocular surface dynamics in health and disease. Revealing the exact nature of these associations could be of paramount importance for developing a holistic approach using highly promising new therapeutic strategies targeting ocular surface diseases.
Collapse
Affiliation(s)
- Marc Labetoulle
- Ophthalmology Départment, Hopital Bicetre, APHP, Université Paris-Saclay, IDMIT Infrastructure, Fontenay-aux-Roses Cedex, France; Hôpital National de la Vision des Quinze, Vingts, IHU ForeSight, Paris Saclay University, Paris, France.
| | - Christophe Baudouin
- Hôpital National de la Vision des Quinze, Vingts, IHU ForeSight, Paris Saclay University, Paris, France
| | - Jose M Benitez Del Castillo
- Departamento de Oftalmología, Hospital Clínico San Carlos, Clínica Rementeria, Instituto Investigaciones Oftalmologicas Ramon Castroviejo, Universidad Complutense, Madrid, Spain
| | - Maurizio Rolando
- Ocular Surface and Dry Eye Center, ISPRE Ophthalmics, Genoa, Italy
| | - Maria Rescigno
- IRCCS Humanitas Research Hospital, via Manzoni 56, Rozzano, 20089, Milan, Italy; Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini, Pieve Emanuele, 20090, MI, Italy
| | | | - Pasquale Aragona
- Department of Biomedical Sciences, Ophthalmology Clinic, University of Messina, Messina, Italy
| |
Collapse
|
20
|
Beatty CJ, Ruiz-Lozano RE, Quiroga-Garza ME, Perez VL, Jester JV, Saban DR. The Yin and Yang of non-immune and immune responses in meibomian gland dysfunction. Ocul Surf 2024; 32:81-90. [PMID: 38224775 PMCID: PMC11881750 DOI: 10.1016/j.jtos.2024.01.004] [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: 09/24/2023] [Revised: 01/04/2024] [Accepted: 01/12/2024] [Indexed: 01/17/2024]
Abstract
Meibomian gland dysfunction (MGD) is a leading cause of dry eye disease and one of the most common ophthalmic conditions encountered in eye clinics worldwide. These holocrine glands are situated in the eyelid, where they produce specialized lipids, or meibum, needed to lubricate the eye surface and slow tear film evaporation - functions which are critical to preserving high-resolution vision. MGD results in tear instability, rapid tear evaporation, changes in local microflora, and dry eye disease, amongst other pathological entities. While studies identifying the mechanisms of MGD have generally focused on gland obstruction, we now know that age is a major risk factor for MGD that is associated with abnormal cell differentiation and renewal. It is also now appreciated that immune-inflammatory disorders, such as certain autoimmune diseases and atopy, may trigger MGD, as demonstrated through a T cell-driven neutrophil response. Here, we independently discuss the underlying roles of gland and immune related factors in MGD, as well as the integration of these two distinct mechanisms into a unified perspective that may aid future studies. From this unique standpoint, we propose a revised model in which glandular dysfunction and immunopathogenic pathways are not primary versus secondary contributors in MGD, but are fluid, interactive, and dynamic, which we likened to the Yin and Yang of MGD.
Collapse
Affiliation(s)
- Cole J Beatty
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, USA; Duke Eye Center, Duke University School of Medicine, Foster Center for Ocular Immunology at Duke Eye Center, Durham, NC, USA
| | - Raul E Ruiz-Lozano
- Duke Eye Center, Duke University School of Medicine, Foster Center for Ocular Immunology at Duke Eye Center, Durham, NC, USA
| | - Manuel E Quiroga-Garza
- Duke Eye Center, Duke University School of Medicine, Foster Center for Ocular Immunology at Duke Eye Center, Durham, NC, USA
| | - Victor L Perez
- Duke Eye Center, Duke University School of Medicine, Foster Center for Ocular Immunology at Duke Eye Center, Durham, NC, USA.
| | - James V Jester
- Department of Ophthalmology and Biomedical Engineering, Gavin Herbert Eye Institute, University of California Irvine, Irvine, CA, USA.
| | - Daniel R Saban
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, USA; Duke Eye Center, Duke University School of Medicine, Foster Center for Ocular Immunology at Duke Eye Center, Durham, NC, USA.
| |
Collapse
|
21
|
Çalık Başaran N, Kırağı D, Tan Ç, Özışık L, Çağdaş Ayvaz ND, Kocabeyoğlu S, Öz ŞG, İrkeç M, Tezcan Fİ. Ocular Changes and Tear Cytokines in Individuals with Low Serum Vitamin D Levels: A Cross-Sectional, Controlled Study. Ocul Immunol Inflamm 2024; 32:287-294. [PMID: 36731535 DOI: 10.1080/09273948.2023.2168698] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/09/2023] [Accepted: 01/10/2023] [Indexed: 02/04/2023]
Abstract
BACKGROUND We investigated the effects of vitamin D on the ocular surface, tear functions, corneal imaging, and tear film cytokine levels. METHODS Fifty-two patients with vitamin D levels were examined in 3 groups according to serum vitamin D levels; 28 in group 1 (<12 ng/ml), 10 in group 2 (12-20 ng/ml), and 14 in group 3 (>20 ng/ml). Ocular surface disease index (OSDI), tear break up time (BUT), lissamine green (LG) staining, Schirmer test, in vivo confocal microscopy (IVCM), and tear collection for cytokine analysis were performed. RESULTS The mean OSDI score was 35.2 ± 23.3, 36.2 ± 17.7, 24.4 ± 18.2 (p = .253), TBUT was 6.7 ± 2.5 sec, 9.3 ± 1.8 sec, 11.1 ± 2.8 sec (p < .001), Schirmer test was 16.7 ± 8.5 mm, 18.7 ± 7.6 mm, and 20.2 ± 7 mm (p = .254), median LG staining grade was 1 (0-3), 1 (0-2), 0 (0-1) (p = .008) in group 1, group 2, and group 3, respectively. Basal epithelial cell density was 4 027 ± 512 cells/mm2, 4 673 ± 451 cells/mm2, 5 067 ± 817 cells/mm2 (p = < 0.001), sub-basal nerve density was 978 ± 204 μm/frame, 1 236 ± 172 μm/frame, 1 425 ± 290 μm/frame (p = <0.001), median number of long nerve fibers was 3 (2-4) nerve/frame, 4 (3-4) nerve/frame, 4 (3-6) nerve/frame (p = .001), and median grade of nerve fiber tortuosity was 2 (0-3), 2.5 (2-3), 3 (2-4) (p < .001) in group 1, group 2, and group 3, respectively. Mean IL-1 β (82.62 ± 15.26, 85.57 ± 17.41, and 66.44 ± 11 ng/ml in group 1, 2 and 3, respectively, p = .002), IL-17 (77.80 ± 24.91, 64.46 ± 25.47, 55.42 ± 12.05 ng/ml in group 1, 2 and 3, respectively, p = .012), and IL-2 (75.7 ± 18.4, 66.13 ± 26.78, and 59.65 ± 16.04 ng/ml in group 1, 2 and 3, respectively, p = .048) levels were significantly lower in group 3, whereas, IL-13 levels were significantly higher in group 3 (16.12 ± 5.24, 19.20 ± 4.90, and 21.6 ± 5.55 ng/ml in groups 1, 2, and 3, respectively, p = .010). CONCLUSIONS Vitamin D deficiency/insufficiency is associated with ocular surface changes shown with significant TBUT, LG staining, and tear film cytokine contents. Besides, significant corneal basal epithelial, sub-basal nerve density, and structural sub-basal nerve changes were associated with lower Vitamin D levels.
Collapse
Affiliation(s)
- Nursel Çalık Başaran
- Medical Faculty, Department of Internal Medicine, Division of General Internal Medicine, Hacettepe University, Ankara, Türkiye
| | - Dila Kırağı
- Medical Faculty, Department of Ophthalmology, Hacettepe University, Ankara, Türkiye
| | - Çağman Tan
- Medical Faculty, Department of Child Health and Diseases, Division of Pediatric Immunology, Hacettepe University, Ankara, Türkiye
| | - Lale Özışık
- Medical Faculty, Department of Internal Medicine, Division of General Internal Medicine, Hacettepe University, Ankara, Türkiye
| | - Nazire Deniz Çağdaş Ayvaz
- Medical Faculty, Department of Child Health and Diseases, Division of Pediatric Immunology, Hacettepe University, Ankara, Türkiye
| | - Sibel Kocabeyoğlu
- Medical Faculty, Department of Ophthalmology, Hacettepe University, Ankara, Türkiye
| | - Şerife Gül Öz
- Medical Faculty, Department of Internal Medicine, Division of General Internal Medicine, Hacettepe University, Ankara, Türkiye
| | - Murat İrkeç
- Medical Faculty, Department of Ophthalmology, Hacettepe University, Ankara, Türkiye
| | - F İlhan Tezcan
- Medical Faculty, Department of Child Health and Diseases, Division of Pediatric Immunology, Hacettepe University, Ankara, Türkiye
| |
Collapse
|
22
|
Yeh SI, Ho TC, Chu TW, Chen SL, Tsao YP. Potential Benefits of Integrin αvβ3 Antagonists in a Mouse Model of Experimental Dry Eye. Cornea 2024; 43:378-386. [PMID: 38015979 DOI: 10.1097/ico.0000000000003427] [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: 03/23/2023] [Accepted: 10/10/2023] [Indexed: 11/30/2023]
Abstract
PURPOSE The purpose of this study was to extensively evaluate the efficacy of integrin αvβ3 antagonists for the treatment of experimental dry eye (EDE). METHODS Vitronectin, an αvβ3 ligand, was used to induce tumor necrosis factor-α gene expression in human THP-1 macrophages. To induce EDE, C57BL/6 mice were housed in a low-humidity controlled environment chamber and injected subcutaneously with scopolamine for 7 days. Subsequently, αvβ3 antagonists, including RGDfD, c(RGDfD), c(RGDiD), c(RGDfK), ATN-161, SB273005, and cilengitide, were administered topically to EDE animals under controlled environment chamber conditions. Corneal epithelial damage in EDE was assessed by fluorescein staining. The density of conjunctival goblet cells and secretion of tears was measured by period acid-Schiff staining and phenol red-impregnated cotton threads, respectively. Inflammation markers, including tumor necrosis factor-α, interleukin (IL)-1β, IL-6, IL-17A, and metalloproteinase (MMP)-9, in the pooled cornea and conjunctiva tissues were examined by real-time polymerase chain reaction. RESULTS The inhibitory effects of αvβ3 antagonists on the vitronectin-induced tumor necrosis factor-α gene expression and integrin-mediated inflammatory signaling were validated in THP-1 macrophages. αvβ3 antagonists ameliorated the impairment of the corneal epithelial barrier with varying therapeutic efficacies, compared with vehicle-treated mice. c(RGDfD) and c(RGDiD) significantly protected against goblet cell loss, tear reduction, and proinflammatory gene expression in EDE. CONCLUSIONS Topical applications of αvβ3 antagonists yield therapeutic benefits in EDE by promoting corneal epithelial defect healing and reducing inflammation. Antagonistic targeting αvβ3 may be a novel promising strategy to treat patients with dry eye disease.
Collapse
Affiliation(s)
- Shu-I Yeh
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
- Department of Ophthalmology, Mackay Memorial Hospital, Taipei, Taiwan
| | - Tsung-Chuan Ho
- Department of Medical Research, Mackay Memorial Hospital, New Taipei City, Taiwan; and
| | - Ting-Wen Chu
- Department of Ophthalmology, Mackay Memorial Hospital, Taipei, Taiwan
| | - Show-Li Chen
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yeou-Ping Tsao
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
- Department of Ophthalmology, Mackay Memorial Hospital, Taipei, Taiwan
- Department of Medical Research, Mackay Memorial Hospital, New Taipei City, Taiwan; and
| |
Collapse
|
23
|
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
|
24
|
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
|
25
|
Lv Z, Li S, Zeng G, Yao K, Han H. Recent progress of nanomedicine in managing dry eye disease. ADVANCES IN OPHTHALMOLOGY PRACTICE AND RESEARCH 2024; 4:23-31. [PMID: 38356795 PMCID: PMC10864857 DOI: 10.1016/j.aopr.2024.01.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/23/2024] [Accepted: 01/25/2024] [Indexed: 02/16/2024]
Abstract
Background Dry eye disease (DED) is a commonly reported ocular complaint that has garnered significant attention in recent research. The global occurrence of DED ranges from 5% to 50%, impacting a substantial proportion of individuals worldwide with increasing frequency. Although topical administration remains the mainstream drug delivery method for ocular diseases, it suffers from drawbacks such as low bioavailability, rapid drug metabolism, and frequent administration requirements. Fortunately, the advancements in nanomedicine offer effective solutions to address the aforementioned issues and provide significant assistance in the treatment of DED. Main text DED is considered a multifactorial disease of the ocular surface and tear film, in which the integrity of tear film function and structure plays a crucial role in maintaining the homeostasis of the ocular surface. The conventional treatment for DED involves the utilization of artificial tear products, cyclosporin, corticosteroids, mucin secretagogues, and nonsteroidal anti-inflammatory drugs. Furthermore, nanomedicine is presently a significant field of study, with numerous clinical trials underway for various nanotherapeutics including nanoemulsions, nanosuspensions, liposomes, and micelles. Notably, some of these innovative nanoformulations have already received FDA approval as novel remedies for DED, and the advancement of nanomedicine is poised to offer enhanced prospects to solve the shortcomings of existing treatments for DED partially. Conclusions This article provides an overview of the latest advancements in nanomedicine for DED treatment, while the field of DED treatment is expected to witness a remarkable breakthrough shortly with the development of nanomedicine, bringing promising prospects for patients worldwide suffering conditions.
Collapse
Affiliation(s)
- Zeen Lv
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, 310009, China
| | - Su Li
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, 310009, China
| | - Guixiang Zeng
- Department of Pediatrics, No. 903 Hospital of PLA Joint Logistic Support Force, Hangzhou, 310013, China
| | - Ke Yao
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, 310009, China
| | - Haijie Han
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, 310009, China
| |
Collapse
|
26
|
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
|
27
|
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
|
28
|
Zhang W, Yin J, Deng Y, Gong Y, Sun X, Chen J. Prostaglandin E2 promotes Th17 differentiation induces corneal epithelial cell apoptosis and participates in the progression of dry eye. Arch Biochem Biophys 2024; 751:109823. [PMID: 37984760 DOI: 10.1016/j.abb.2023.109823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 10/23/2023] [Accepted: 11/16/2023] [Indexed: 11/22/2023]
Abstract
This study is mainly based on T helper type 17 (Th17) cells analysis of the mechanism of prostaglandin E2 (PGE2) promoting the progression of dry eye (DE). Scopolamine and dry environment were used to induce mice DE model. Celecoxib was used to inhibit PGE2. Corneal epithelial cells and CD4+ T cells were used to construct a co-culture system. The osmotic pressure was increased by adding NaCl to simulate DE in vitro. AH6809 and E7046 were used to pre-culture to inhibit EP2/4 in T cells to verify the effect of exogenous PGE2 on Th17 cell differentiation and corneal epithelial cell apoptosis. The function of Th17 cells was analyzed by detecting RORγt and interleukin-17 (IL-17). PGE2 was instilled on the ocular surface to induce DE symptoms of mice. AH6809 and E7046 were used to inhibit EP2/4. The corneal epithelial cell apoptosis was observed by TUNEL. The proportion of Th17 cells in corneal tissue and draining lymph nodes (DLNs) was detected by flow cytometry. In DE mice, the concentration of PGE2 and IL-17 increased in tears, and the proportion of Th17 increased, while inhibition of PGE2 alleviated the symptoms of DE and inhibited Th17 differentiation. Hypertonic environment induces corneal epithelial cells to secrete PGE2. PGE2 promoted the expression of EP2/4 and the differentiation of Th17 cells in vitro. The hypertonic environment promoted PGE2 level and the apoptosis of corneal epithelial cells in the co-culture system. PGE2 alone did not cause corneal epithelial cell apoptosis, while PGE2 promoted apoptosis by promoting Th17. Blocking EP2/4 reduced the induction of Th17 differentiation by PGE2 and the promoted corneal epithelial cell apoptosis. Animal experiments showed that exogenous PGE2 induced DE symptoms. Blocking EP2/4 not only inhibited the proportion of Th17, but also alleviated the apoptosis of corneal epithelial cells caused by PGE2. PGE2 induces aggravation of inflammation by promoting the level of Th17 in the ocular surface, and causes corneal epithelial cell apoptosis, thereby participating in the progression of DE.
Collapse
Affiliation(s)
- Weijia Zhang
- Department of Ophthalmology, Yan 'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, China
| | - Jianwei Yin
- Department of Anesthesiology, Yan 'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, China
| | - Yachun Deng
- Department of Ophthalmology, Yan 'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, China
| | - Yu Gong
- Department of Ophthalmology, Yan 'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, China
| | - Xiaoyu Sun
- Department of Ophthalmology, Yan 'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, China
| | - Jingyao Chen
- Department of Ophthalmology, Yan 'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, China.
| |
Collapse
|
29
|
Calderón-García AÁ, Valencia-Nieto L, Valencia-Sandonis C, López-de la Rosa A, Blanco-Vazquez M, Fernández I, García-Vázquez C, Arroyo-Del Arroyo C, González-García MJ, Enríquez-de-Salamanca A. Gene expression changes in conjunctival cells associated with contact lens wear and discomfort. Ocul Surf 2024; 31:31-42. [PMID: 38128761 DOI: 10.1016/j.jtos.2023.12.004] [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: 09/01/2023] [Revised: 10/27/2023] [Accepted: 12/09/2023] [Indexed: 12/23/2023]
Abstract
PURPOSE This study aimed to analyze the differences in the expression of pain-related genes in conjunctival epithelial cells among symptomatic contact lens (CL) wearers (SCLWs), asymptomatic CL wearers (ACLWs), and non-CL wearers (non-CLWs). METHODS For this study, 60 participants (20 non-CLWs, 40 CLWs) were enrolled. The CLW group comprised 20 ACLWs and 20 SCLWs according to the Contact Lens Dry Eye Questionnaire short form©. Conjunctival cells were collected using impression cytology, and RNA was isolated and used to determine the expression levels of 85 human genes involved in neuropathic and inflammatory pain. The effects of CL wear and discomfort were evaluated using mixed-effects ANOVA with partially nested fixed-effects model. Gene set enrichment analysis was performed to assign biological meaning to sets of differentially expressed genes. RESULTS Six genes (CD200, EDN1, GRIN1, PTGS1, P2RX7, and TNF) were significantly upregulated in CLWs compared to non-CLWs. Eleven genes (ADORA1, BDKRB1, CACNA1B, DBH, GRIN1, GRM1, HTR1A, PDYN, PTGS1, P2RX3, and TNF) were downregulated in SCLWs compared to ACLWs. These genes were mainly related to pain, synaptic transmission and signaling, ion transport, calcium transport and concentration, and cell-cell signaling. CONCLUSIONS CL wear modified the expression of pain- and inflammation-related genes in conjunctival epithelial cells. These changes may be in part, along with other mechanisms, responsible for CL discomfort in SCLWs.
Collapse
Affiliation(s)
- Andrés Ángel Calderón-García
- Institute of Applied Ophthalmobiology (IOBA), Universidad de Valladolid, Valladolid, Spain; Department of Theoretical Physics, Atomic and Optics, Universidad de Valladolid, Valladolid, Spain
| | - Laura Valencia-Nieto
- Institute of Applied Ophthalmobiology (IOBA), Universidad de Valladolid, Valladolid, Spain; Department of Theoretical Physics, Atomic and Optics, Universidad de Valladolid, Valladolid, Spain
| | - Cristina Valencia-Sandonis
- Institute of Applied Ophthalmobiology (IOBA), Universidad de Valladolid, Valladolid, Spain; Department of Theoretical Physics, Atomic and Optics, Universidad de Valladolid, Valladolid, Spain
| | - Alberto López-de la Rosa
- Institute of Applied Ophthalmobiology (IOBA), Universidad de Valladolid, Valladolid, Spain; Department of Theoretical Physics, Atomic and Optics, Universidad de Valladolid, Valladolid, Spain
| | - Marta Blanco-Vazquez
- Institute of Applied Ophthalmobiology (IOBA), Universidad de Valladolid, Valladolid, Spain
| | - Itziar Fernández
- Institute of Applied Ophthalmobiology (IOBA), Universidad de Valladolid, Valladolid, Spain; Networking Research Center on Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), Valladolid, Spain; Department of Statistics and Operations Research, Universidad de Valladolid, Valladolid, Spain
| | - Carmen García-Vázquez
- Institute of Applied Ophthalmobiology (IOBA), Universidad de Valladolid, Valladolid, Spain
| | - Cristina Arroyo-Del Arroyo
- Institute of Applied Ophthalmobiology (IOBA), Universidad de Valladolid, Valladolid, Spain; Department of Theoretical Physics, Atomic and Optics, Universidad de Valladolid, Valladolid, Spain
| | - María J González-García
- Institute of Applied Ophthalmobiology (IOBA), Universidad de Valladolid, Valladolid, Spain; Department of Theoretical Physics, Atomic and Optics, Universidad de Valladolid, Valladolid, Spain; Networking Research Center on Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), Valladolid, Spain.
| | - Amalia Enríquez-de-Salamanca
- Institute of Applied Ophthalmobiology (IOBA), Universidad de Valladolid, Valladolid, Spain; Networking Research Center on Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), Valladolid, Spain
| |
Collapse
|
30
|
Rasaruck U, Kasetsuwan N, Kittipibul T, Pongchaikul P, Chatsuwan T. Composition and diversity of meibum microbiota in meibomian gland dysfunction and the correlation with tear cytokine levels. PLoS One 2023; 18:e0296296. [PMID: 38134040 PMCID: PMC10745150 DOI: 10.1371/journal.pone.0296296] [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: 07/17/2023] [Accepted: 12/10/2023] [Indexed: 12/24/2023] Open
Abstract
Meibomian gland dysfunction (MGD) leads to meibum stasis and pathogenic bacteria proliferation. We determined meibum microbiota via next-generation sequencing (NGS) and examined their association with tear cytokine levels in patients with MGD. This cross-sectional study included 44 moderate-severe patients with MGD and 44 healthy controls (HCs). All volunteers underwent assessment with the ocular surface disease index questionnaire, Schirmer without anesthesia, tear break-up time, Oxford grading of ocular surface staining, and lid and meibum features. Sample collection included tears for cytokine detection and meibum for 16S rRNA NGS. No significant differences were observed in the α-diversity of patients with MGD compared with that in HCs. However, Simpson's index showed significantly decreased α-diversity for severe MGD than for moderate MGD (p = 0.045). Principal coordinate analysis showed no significant differences in β-diversity in meibum samples from patients with MGD and HCs. Patients with MGD had significantly higher relative abundances of Bacteroides (8.54% vs. 6.00%, p = 0.015) and Novosphingobium (0.14% vs. 0.004%, p = 0.012) than the HCs. Significantly higher interleukin (IL)-17A was detected in the MGD group than in the HC group, particularly for severe MGD (p = 0.008). Although Bacteroides was more abundant in the MGD group than in the HC group, it was not positively correlated with IL-17A. The relationship between core meibum microbiota and tear cytokine levels remains unclear. However, increased Bacteroides and Novosphingobium abundance may be critical in MGD pathophysiology.
Collapse
Affiliation(s)
- Ubonwan Rasaruck
- Department of Ophthalmology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Ngamjit Kasetsuwan
- Department of Ophthalmology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Excellence Center of Cornea and Limbal Stem Cell Transplantation, Department of Ophthalmology, King Chulalongkorn Memorial Hospital and Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Thanachaporn Kittipibul
- Department of Ophthalmology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Excellence Center of Cornea and Limbal Stem Cell Transplantation, Department of Ophthalmology, King Chulalongkorn Memorial Hospital and Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Pisut Pongchaikul
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital Mahidol University, Samut Prakarn, Thailand
- Integrative Computational Bioscience (ICBS) Center, Mahidol University, Nakorn Pathom, Thailand
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Tanittha Chatsuwan
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Antimicrobial Resistance and Stewardship, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
31
|
Han Y, Guo S, Li Y, Li J, Zhu L, Liu Y, Lv Y, Yu D, Zheng L, Huang C, Li C, Hu J, Liu Z. Berberine ameliorate inflammation and apoptosis via modulating PI3K/AKT/NFκB and MAPK pathway on dry eye. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 121:155081. [PMID: 37748390 DOI: 10.1016/j.phymed.2023.155081] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 06/08/2023] [Accepted: 09/10/2023] [Indexed: 09/27/2023]
Abstract
BACKGROUND Dry eye disease (DED) is a multifactorial disease in ocular surface, and inflammation plays an etiological role. Berberine (BBR) has shown efficacy in treating inflammatory diseases. Yet, there was no adequate information related to the therapeutic effects of BBR for DED. PURPOSE To detect the effects and explore the potential mechanisms of BBR on DED. STUDY DESIGN In vitro, in vivo study and network pharmacology analysis were involved. METHOD The human corneal epithelium cells viability was evaluated with different concentrations of BBR. Dry eye murine model was established by exposing to the desiccating stress, and Ciclosporin (CSA), BBR eye drops or vehicle were topical administration for 7 days. The phenol red cotton tests, Oregon-green-dextran staining and Periodic acid-Schiff staining were performed and evaluated the dry eye after treatment. Inflammation and apoptosis levels of ocular surface were quantified. The potential targets related to berberine and dry eye were collected from databases. The Protein-Protein interaction network analysis and GO & KEGG enrichment analysis were realized by STRING database, Metascape platform and Cytoscape software to find core targets and signaling pathways. The SchrÖdinger software was used to molecular docking and PyMOL software to visualization. Finally, the levels of PI3K/AKT/NFκB and MAPK pathways were detected. RESULT The data revealed BBR could rescue impaired HCE under hyperosmotic conditions. In addition, BBR eye drops could ameliorate dry eye. And BBR eye drops suppressed the inflammatory factors and CD4+T cells infiltration in conjunctiva. Besides, BBR eye drops protected ocular surface by avoiding the severe apoptosis and decreasing the level of MMP-3 and MMP-9. 148 common targets intersection between BBR and dry eye were found via network pharmacology analysis. Core proteins and core pathways were identified through PPI and GO&KEGG enrichment analysis. Molecular docking displayed excellent binding between BBR and those core targets. Finally, in vivo study verified that BBR eye drops had a therapeutic effect in dry eye by inhibiting PI3K/AKT/NFκB and MAPK pathways. CONCLUSION The research provided convincing evidence that BBR could be a candidate drug for dry eye.
Collapse
Affiliation(s)
- Yi Han
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Fujian Engineering and Research Center of Eye Regenerative Medicine, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Eye Institute & Affiliated Xiamen Eye Center, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China; Department of Ophthalmology, the First Affiliated Hospital of University of South China, Postdoctoral mobile station of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Shujia Guo
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Fujian Engineering and Research Center of Eye Regenerative Medicine, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Eye Institute & Affiliated Xiamen Eye Center, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Yunpeng Li
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Fujian Engineering and Research Center of Eye Regenerative Medicine, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Eye Institute & Affiliated Xiamen Eye Center, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Jiani Li
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Fujian Engineering and Research Center of Eye Regenerative Medicine, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Eye Institute & Affiliated Xiamen Eye Center, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Linfangzi Zhu
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Fujian Engineering and Research Center of Eye Regenerative Medicine, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Eye Institute & Affiliated Xiamen Eye Center, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Yuwen Liu
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Fujian Engineering and Research Center of Eye Regenerative Medicine, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Eye Institute & Affiliated Xiamen Eye Center, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Yufei Lv
- Department of Ophthalmology, the First Affiliated Hospital of University of South China, Postdoctoral mobile station of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Dong Yu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Lan Zheng
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Fujian Engineering and Research Center of Eye Regenerative Medicine, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Eye Institute & Affiliated Xiamen Eye Center, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Caihong Huang
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Fujian Engineering and Research Center of Eye Regenerative Medicine, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Eye Institute & Affiliated Xiamen Eye Center, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Cheng Li
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Fujian Engineering and Research Center of Eye Regenerative Medicine, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Eye Institute & Affiliated Xiamen Eye Center, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China; Department of Ophthalmology, the First Affiliated Hospital of University of South China, Postdoctoral mobile station of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| | - Jiaoyue Hu
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Fujian Engineering and Research Center of Eye Regenerative Medicine, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Eye Institute & Affiliated Xiamen Eye Center, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China.
| | - Zuguo Liu
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Fujian Engineering and Research Center of Eye Regenerative Medicine, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Eye Institute & Affiliated Xiamen Eye Center, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China; Department of Ophthalmology, the First Affiliated Hospital of University of South China, Postdoctoral mobile station of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| |
Collapse
|
32
|
Harrell CR, Feulner L, Djonov V, Pavlovic D, Volarevic V. The Molecular Mechanisms Responsible for Tear Hyperosmolarity-Induced Pathological Changes in the Eyes of Dry Eye Disease Patients. Cells 2023; 12:2755. [PMID: 38067183 PMCID: PMC10706334 DOI: 10.3390/cells12232755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 11/18/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Abstract
Dry eye disease (DED) is a multifactorial disorder of the lacrimal system and ocular surface, characterized by a deficiency in the quality and/or quantity of the tear fluid. The multifactorial nature of DED encompasses a number of interconnected underlying pathologies, including loss of homeostasis, instability and hyperosmolarity of the tears, and the induction and propagation of detrimental inflammatory responses in the eyes, which finally results in the development of neurosensory dysfunction and visual disruption. Dryness, grittiness, scratchiness, discomfort, inflammation, burning, watering, ocular fatigue, pain, and decreased functional visual acuity are common symptoms of DED. Eye dysfunction drastically attenuates patients' quality of life. Accordingly, a better understanding of the pathogenic processes that regulate the development and progression of DED is crucially important for the establishment of new and more effective DED-related treatment approaches, which would significantly improve the quality of life of DED patients. Since the process of osmoregulation, which guards the ocular surface epithelia and maintains normal vision, is affected when the osmolarity of the tears is greater than that of the epithelial cells, tear hyperosmolarity (THO) is considered an initial, important step in the development, progression, and aggravation of DED. In order to delineate the role of THO in the pathogenesis of DED, in this review article, we summarize current knowledge related to the molecular mechanisms responsible for the development of THO-induced pathological changes in the eyes of DED patients, and we briefly discuss the therapeutic potential of hypo-osmotic eye drops in DED treatment.
Collapse
Affiliation(s)
- Carl Randall Harrell
- Regenerative Processing Plant, LLC, 34176 US Highway 19 N, Palm Harbor, FL 34684, USA;
| | - Lisa Feulner
- Advanced Eye Care & Aesthetics, 104 Plumtree Rd Suite 107, Bel Air, MD 21015, USA;
| | - Valentin Djonov
- Institute of Anatomy, University of Bern, Baltzerstrasse 2, 3012 Bern, Switzerland;
| | - Dragica Pavlovic
- Departments of Genetics and Microbiology and Immunology, Center for Harm Reduction of Biological and Chemical Hazards, Faculty of Medical Sciences, University of Kragujevac, 69 Svetozar Markovic Street, 34000 Kragujevac, Serbia;
| | - Vladislav Volarevic
- Departments of Genetics and Microbiology and Immunology, Center for Harm Reduction of Biological and Chemical Hazards, Faculty of Medical Sciences, University of Kragujevac, 69 Svetozar Markovic Street, 34000 Kragujevac, Serbia;
| |
Collapse
|
33
|
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
|
34
|
Yang C, Li Y, Liu Y, Xu Z, Li W, Cao W, Jin K, Liu Y. Protection of Barrier Function in Cultured Human Corneal Epithelial Cells by Semaphorin 4D. Curr Eye Res 2023; 48:894-903. [PMID: 37395011 DOI: 10.1080/02713683.2023.2232572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 06/23/2023] [Indexed: 07/04/2023]
Abstract
PURPOSE Corneal epithelial barrier function is important to maintain corneal homeostasis and is impaired by inflammation. We aimed to investigate the localization of semaphorin 4D (Sema4D) in the cornea, and its effects on the barrier function of cultured corneal epithelial cells. METHODS The expressions of semaphorin4 D and its receptor in the murine cornea were examined by immunoblot, immunofluorescent staining and confocal microscopy observations. Human corneal epithelial (HCE) cells stimulated by TNF-α or IL-1β were cultured with or without Sema4D. Cell viability was examined by CCK8, cell migration was evaluated by scratch wound assay, and barrier function was assessed by transepithelial electrical resistance (TEER) and Dextran-FITC permeability assay. The expression of tight junction proteins in HCE cells was examined by immunoblot, immunofluorescent staining and qRT-PCR. RESULTS We demonstrated that the protein of Sema4D and its receptor, plexin-B1, was expressed in murine cornea. Sema4D induced an increase in the TEER and a decrease in the permeability of HCE cells. It also induced the expression of tight junction protein ZO-1, occludin and claudin-1 in HCE cells. Furthermore, under stimulation of TNF-α or IL-1β, Sema4D treatment could inhibit the decreased TEER and the elevated permeability of HCE cells. CONCLUSIONS Sema4D is located distinctly in corneal epithelial cells and promoted their barrier function by increasing the expression of tight junction proteins. Sema4D may act as a preventive for maintaining corneal epithelial barrier function during ocular inflammation.
Collapse
Affiliation(s)
- Chengcheng Yang
- Department of Ophthalmology, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, P.R. China
| | - Yunzepeng Li
- Department of Ophthalmology, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, P.R. China
| | - Ye Liu
- Department of Pathology, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, P.R. China
| | - Zhenghua Xu
- Department of Ophthalmology, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, P.R. China
| | - Wei Li
- Department of Pathology, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, P.R. China
| | - Wanwei Cao
- Department of Pathology, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, P.R. China
| | - Kai Jin
- Department of Ophthalmology, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, P.R. China
| | - Yang Liu
- Department of Ophthalmology, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, P.R. China
| |
Collapse
|
35
|
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
|
36
|
Li X, Chen K, Wang Z, Li J, Wang X, Xie C, Tong J, Shen Y. The mTOR signalling in corneal diseases: A recent update. Biochem Pharmacol 2023; 213:115620. [PMID: 37217140 DOI: 10.1016/j.bcp.2023.115620] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/16/2023] [Accepted: 05/16/2023] [Indexed: 05/24/2023]
Abstract
Corneal diseases affect 4.2 million people worldwide and are a leading cause of vision impairment and blindness. Current treatments for corneal diseases, such as antibiotics, steroids, and surgical interventions, have numerous disadvantages and challenges. Thus, there is an urgent need for more effective therapies. Although the pathogenesis of corneal diseases is not fully understood, it is known that injury caused by various stresses and postinjury healing, such as epithelial renewal, inflammation, stromal fibrosis, and neovascularization, are highly involved. Mammalian target of rapamycin (mTOR) is a key regulator of cell growth, metabolism, and the immune response. Recent studies have revealed that activation of mTOR signalling extensively contributes to the pathogenesis of various corneal diseases, and inhibition of mTOR with rapamycin achieves promising outcomes, supporting the potential of mTOR as a therapeutic target. In this review, we detail the function of mTOR in corneal diseases and how these characteristics contribute to disease treatment using mTOR-targeted drugs.
Collapse
Affiliation(s)
- Xiang Li
- Department of Ophthalmology, the First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Kuangqi Chen
- Department of Ophthalmology, the First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Zixi Wang
- Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiayuan Li
- Department of Ophthalmology, the First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Xiawei Wang
- Department of Ophthalmology, the First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Chen Xie
- Department of Ophthalmology, the First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang Province, China.
| | - Jianping Tong
- Department of Ophthalmology, the First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang Province, China.
| | - Ye Shen
- Department of Ophthalmology, the First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang Province, China.
| |
Collapse
|
37
|
Faustine G, Prijanti AR, Wibowo H. Effect of Lutein on Ocular Goblet Cell, IFN-γ, and IL-17 Concentration in Dry Eye-Induced Mice Model. Cureus 2023; 15:e42009. [PMID: 37593298 PMCID: PMC10430303 DOI: 10.7759/cureus.42009] [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: 07/17/2023] [Indexed: 08/19/2023] Open
Abstract
Introduction Dry eye disease affects a substantial number of individuals globally and significantly impacts their quality of life and productivity. Understanding the underlying mechanisms and managing dry eye disease poses substantial challenges. Recent research has highlighted the involvement of various inflammatory mediators in the pathogenesis of dry eye disease, including the cytokines interferon (IFN)-γ and interleukin (IL)-17. Activation of stress signaling pathways and residential immune cells on the ocular epithelial surface ignites epithelial changes, destabilizes tear film, amplifies inflammation and creates an endless loop. Lutein is a β-carotenoid antioxidant which has been proven to be beneficial in many ocular diseases due to its protective and anti-inflammatory effect induced by various stimulators. Lutein also acts as a direct and indirect antioxidant agent, suppressing oxidative stress and mitigating oxidative damage. The purpose of this research is to investigate the potential therapeutic effects of lutein in a mouse model of dry eye, aiming to elucidate its impact on ocular manifestation, goblet cells count, IFN-γ and IL-17 level. Methods Desiccating stress was induced in C57BL/6 mice. In a separate group, lutein was administered subcutaneously on a daily basis throughout the experimental period. Clinical manifestations of dry eye, including ocular surface changes, were documented photographically. Goblet cell concentration was assessed through Periodic Acid-Schiff (PAS) staining, and the levels of IFN-γ and IL-17 were measured using enzyme-linked immunosorbent assay (ELISA). Data obtained from these assessments were compared between the experimental groups to determine the potential effects of lutein on dry eye pathology and cytokine levels. Results Significant differences were observed in clinical observations and goblet cell concentrations among the groups; however, no statistically significant differences were found in the levels of IFN-γ and IL-17 between the groups. The untreated group exhibited significantly higher opacities and irregularities compared to the lutein-treated group, whereas the mean goblet cell count was highest in the lutein-treated group. Conclusion Lutein administration improves clinical observations and goblet cell concentrations in a mouse model of dry eye. The treated group exhibited improved ocular surface integrity, but no significant differences in the tested cytokine levels were observed. These findings suggest that lutein supplementation could be a promising therapeutic option for managing dry eye disease. Further research is needed to understand the underlying mechanisms and long-term effects of lutein in dry eye management.
Collapse
Affiliation(s)
| | - Ani R Prijanti
- Biochemistry and Molecular Biology, Universitas Indonesia, Jakarta, IDN
| | - Heri Wibowo
- Parasitology, Universitas Indonesia, Jakarta, IDN
| |
Collapse
|
38
|
Zhuang D, Misra SL, Mugisho OO, Rupenthal ID, Craig JP. NLRP3 Inflammasome as a Potential Therapeutic Target in Dry Eye Disease. Int J Mol Sci 2023; 24:10866. [PMID: 37446038 DOI: 10.3390/ijms241310866] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 06/19/2023] [Accepted: 06/22/2023] [Indexed: 07/15/2023] Open
Abstract
Dry eye disease (DED) is a multifactorial ocular surface disorder arising from numerous interrelated underlying pathologies that trigger a self-perpetuating cycle of instability, hyperosmolarity, and ocular surface damage. Associated ocular discomfort and visual disturbance contribute negatively to quality of life. Ocular surface inflammation has been increasingly recognised as playing a key role in the pathophysiology of chronic DED. Current readily available anti-inflammatory agents successfully relieve symptoms, but often without addressing the underlying pathophysiological mechanism. The NOD-like receptor protein-3 (NLRP3) inflammasome pathway has recently been implicated as a key driver of ocular surface inflammation, as reported in pre-clinical and clinical studies of DED. This review discusses the intimate relationship between DED and inflammation, highlights the involvement of the inflammasome in the development of DED, describes existing anti-inflammatory therapies and their limitations, and evaluates the potential of the inflammasome in the context of the existing anti-inflammatory therapeutic landscape as a therapeutic target for effective treatment of the disease.
Collapse
Affiliation(s)
- Dian Zhuang
- Department of Ophthalmology, New Zealand National Eye Centre, Faculty of Medical and Health Sciences, The University of Auckland, Auckland 1142, New Zealand
| | - Stuti L Misra
- Department of Ophthalmology, New Zealand National Eye Centre, Faculty of Medical and Health Sciences, The University of Auckland, Auckland 1142, New Zealand
| | - Odunayo O Mugisho
- Buchanan Ocular Therapeutics Unit, Department of Ophthalmology, New Zealand National Eye Centre, Faculty of Medical and Health Sciences, The University of Auckland, Auckland 1142, New Zealand
| | - Ilva D Rupenthal
- Buchanan Ocular Therapeutics Unit, Department of Ophthalmology, New Zealand National Eye Centre, Faculty of Medical and Health Sciences, The University of Auckland, Auckland 1142, New Zealand
| | - Jennifer P Craig
- Department of Ophthalmology, New Zealand National Eye Centre, Faculty of Medical and Health Sciences, The University of Auckland, Auckland 1142, New Zealand
| |
Collapse
|
39
|
Chen C, Zeng P, Yao X, Huang Z, Ling Y, Huang Y, Hou L, Li H, Zhu D, Ma W. Gut microbiota combined with fecal metabolomics reveals the effects of FuFang Runzaoling on the microbial and metabolic profiles in NOD mouse model of Sjögren's syndrome. BMC Complement Med Ther 2023; 23:195. [PMID: 37312184 DOI: 10.1186/s12906-023-04017-5] [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: 01/16/2023] [Accepted: 05/29/2023] [Indexed: 06/15/2023] Open
Abstract
OBJECTIVE Sjögren's syndrome (SS) is an inflammatory autoimmune disease characterized by high levels of chronic lymphocyte infiltration. Differences and dysfunction in the gut microbiota and metabolites may be closely related to the pathogenesis of SS. The purpose of this study was to reveal the relationship between the gut microbiota and metabolome in NOD mice as a model of SS and the role of FuFang Runzaoling (FRZ), which is a clinically effective in treating SS. METHODS NOD mice were gavaged with FRZ for 10 weeks. The ingested volume of drinking water, submandibular gland index, pathologic changes of the submandibular glands, and serum cytokines interleukin (IL)-6, IL-10, IL-17 A, and tumor necrosis factor-alpha (TNF-α) were determined. The roles of FRZ on gut microbiota and fecal metabolites were explored by 16 S rRNA gene sequencing and liquid chromatography-mass spectrometry (LC-MC), respectively. The correlation between them was determined by Pearson correlation analysis. RESULTS Compared with the model group, the drinking water volume of NOD mice treated with FRZ increased and the submandibular gland index decreased. FRZ effectively ameliorated lymphocyte infiltration in the small submandibular glands in mice. Serum levels of IL-6, TNF-α, and IL-17 A decreased, and IL-10 increased. The Firmicutes/Bacteroidetes ratio in the FRZ treatment group was higher. FRZ significantly downregulated the relative abundance of the family Bacteroidaceae and genus Bacteroides, and significantly upregulated the relative abundance of genus Lachnospiraceae_UCG-001. Orthogonal projections to latent structures discriminant analysis (OPLS-DA) revealed the significant change in fecal metabolites after FRZ treatment. Based on criteria of OPLS-DA variable influence on projection > 1, P < 0.05, and fragmentation score > 50, a total of 109 metabolites in the FRZ-H group were differentially regulated (47 downregulated and 62 upregulated) compared to their expressions in the model group. Kyoto Encyclopedia of Genes and Genomes pathway analysis revealed enriched metabolic of sphingolipid metabolism, retrograde endocannabinoid signaling, GABAergic synapse, necroptosis, arginine biosynthesis, and metabolism of histidine, alanine, aspartate, and glutamate. Correlation analysis between gut microbiota and fecal metabolites suggested that the enriched bacteria were related to many key metabolites. CONCLUSIONS Taken together, we found FRZ could reduce the inflammatory responses in NOD mice by regulating the gut microbiota, fecal metabolites, and their correlation to emerge a therapeutic effect on mice with SS. This will lay the foundation for the further studies and applications of FRZ, and the use of gut microbiotas as drug targets to treat SS.
Collapse
Affiliation(s)
- Changming Chen
- Department of Rheumatology and Immunology, the Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Ping Zeng
- Department of Rheumatology and Immunology, the Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Xueming Yao
- Department of Rheumatology and Immunology, the Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Zhaowei Huang
- Department of Rheumatology and Immunology, the Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Yi Ling
- Department of Rheumatology and Immunology, the Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Ying Huang
- Department of Rheumatology and Immunology, the Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Lei Hou
- Department of Rheumatology and Immunology, the Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Hufan Li
- Department of Rheumatology and Immunology, the Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Dan Zhu
- Department of Rheumatology and Immunology, the Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Wukai Ma
- Department of Rheumatology and Immunology, the Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China.
| |
Collapse
|
40
|
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
|
41
|
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
|
42
|
Mitrović J, Hrkač S, Tečer J, Golob M, Ljilja Posavec A, Kolar Mitrović H, Grgurević L. Pathogenesis of Extraarticular Manifestations in Rheumatoid Arthritis-A Comprehensive Review. Biomedicines 2023; 11:biomedicines11051262. [PMID: 37238933 DOI: 10.3390/biomedicines11051262] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/16/2023] [Accepted: 04/18/2023] [Indexed: 05/28/2023] Open
Abstract
Rheumatoid arthritis (RA) is among the most prevalent and debilitating autoimmune inflammatory chronic diseases. Although it is primarily characterized by destructive peripheral arthritis, it is a systemic disease, and RA-related extraarticular manifestations (EAMs) can affect almost every organ, exhibit a multitude of clinical presentations, and can even be asymptomatic. Importantly, EAMs largely contribute to the quality of life and mortality of RA patients, particularly substantially increased risk of cardiovascular disease (CVD) which is the leading cause of death in RA patients. In spite of known risk factors related to EAM development, a more in-depth understanding of its pathophysiology is lacking. Improved knowledge of EAMs and their comparison to the pathogenesis of arthritis in RA could lead to a better understanding of RA inflammation overall and its initial phases. Taking into account that RA is a disorder that has many faces and that each person experiences it and responds to treatments differently, gaining a better understanding of the connections between the joint and extra-joint manifestations could help to create new treatments and improve the overall approach to the patient.
Collapse
Affiliation(s)
- Joško Mitrović
- Division of Clinical Immunology, Rheumatology and Allergology, Department of Internal Medicine, Dubrava University Hospital, School of Medicine and Faculty of Pharmacy and Biochemistry, University of Zagreb, Avenija Gojka Šuška 6, 10000 Zagreb, Croatia
| | - Stela Hrkač
- Division of Clinical Immunology, Rheumatology and Allergology, Department of Internal Medicine, Dubrava University Hospital, School of Medicine and Faculty of Pharmacy and Biochemistry, University of Zagreb, Avenija Gojka Šuška 6, 10000 Zagreb, Croatia
| | - Josip Tečer
- Division of Clinical Immunology, Rheumatology and Allergology, Department of Internal Medicine, Dubrava University Hospital, School of Medicine and Faculty of Pharmacy and Biochemistry, University of Zagreb, Avenija Gojka Šuška 6, 10000 Zagreb, Croatia
| | - Majda Golob
- Division of Clinical Immunology, Rheumatology and Allergology, Department of Internal Medicine, Dubrava University Hospital, School of Medicine and Faculty of Pharmacy and Biochemistry, University of Zagreb, Avenija Gojka Šuška 6, 10000 Zagreb, Croatia
| | - Anja Ljilja Posavec
- Polyclinic for the Respiratory Tract Diseases, Prilaz Baruna Filipovića 11, 10000 Zagreb, Croatia
| | - Helena Kolar Mitrović
- Department of Rheumatology and Rehabilitation, Zagreb University Hospital Center, University of Zagreb School of Medicine, Kišpatićeva 12, 10000 Zagreb, Croatia
| | - Lovorka Grgurević
- Center for Translational and Clinical Research, Department of Proteomics, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
- Department of Anatomy, "Drago Perovic", School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| |
Collapse
|
43
|
Shan H, Liu W, Li Y, Pang K. The Autoimmune Rheumatic Disease Related Dry Eye and Its Association with Retinopathy. Biomolecules 2023; 13:724. [PMID: 37238594 PMCID: PMC10216215 DOI: 10.3390/biom13050724] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 04/19/2023] [Accepted: 04/20/2023] [Indexed: 05/28/2023] Open
Abstract
Dry eye disease is a chronic disease of the ocular surface characterized by abnormal tear film composition, tear film instability, and ocular surface inflammation, affecting 5% to 50% of the population worldwide. Autoimmune rheumatic diseases (ARDs) are systemic disorders with multi-organ involvement, including the eye, and play a significant role in dry eye. To date, most studies have focused on Sjögren's syndrome (one of the ARDs) since it manifests as two of the most common symptoms-dry eyes and a dry mouth-and attracts physicians to explore the relationship between dry eye and ARDs. Many patients complained of dry eye related symptoms before they were diagnosed with ARDs, and ocular surface malaise is a sensitive indicator of the severity of ARDs. In addition, ARD related dry eye is also associated with some retinal diseases directly or indirectly, which are described in this review. This review also summarizes the incidence, epidemiological characteristics, pathogenesis, and accompanying ocular lesions of ARD's related dry eye, emphasizing the potential role of dry eye in recognition and monitoring among ARDs patients.
Collapse
Affiliation(s)
| | | | | | - Kunpeng Pang
- Department of Ophthalmology, Qilu Hospital of Shandong University, Jinan 250012, China
| |
Collapse
|
44
|
Galor A, Britten-Jones AC, Feng Y, Ferrari G, Goldblum D, Gupta PK, Merayo-Lloves J, Na KS, Naroo SA, Nichols KK, Rocha EM, Tong L, Wang MTM, Craig JP. TFOS Lifestyle: Impact of lifestyle challenges on the ocular surface. Ocul Surf 2023; 28:262-303. [PMID: 37054911 DOI: 10.1016/j.jtos.2023.04.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 04/07/2023] [Indexed: 04/15/2023]
Abstract
Many factors in the domains of mental, physical, and social health have been associated with various ocular surface diseases, with most of the focus centered on aspects of dry eye disease (DED). Regarding mental health factors, several cross-sectional studies have noted associations between depression and anxiety, and medications used to treat these disorders, and DED symptoms. Sleep disorders (both involving quality and quantity of sleep) have also been associated with DED symptoms. Under the domain of physical health, several factors have been linked to meibomian gland abnormalities, including obesity and face mask wear. Cross-sectional studies have also linked chronic pain conditions, specifically migraine, chronic pain syndrome and fibromyalgia, to DED, principally focusing on DED symptoms. A systematic review and meta-analysis reviewed available data and concluded that various chronic pain conditions increased the risk of DED (variably defined), with odds ratios ranging from 1.60 to 2.16. However, heterogeneity was noted, highlighting the need for additional studies examining the impact of chronic pain on DED signs and subtype (evaporative versus aqueous deficient). With respect to societal factors, tobacco use has been most closely linked to tear instability, cocaine to decreased corneal sensitivity, and alcohol to tear film disturbances and DED symptoms.
Collapse
Affiliation(s)
- Anat Galor
- Bascom Palmer Eye Institute, University of Miami, Miami, FL, USA; Surgical Services, Miami Veterans Administration, Miami, FL, USA.
| | - Alexis Ceecee Britten-Jones
- Department of Optometry and Vision Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Victoria, Australia
| | - Yun Feng
- Department of Ophthalmology, Peking University Eye Center, Peking University Third Hospital, Beijing, China
| | - Giulio Ferrari
- Cornea and Ocular Surface Unit, Eye Repair Lab, San Raffaele Scientific Institute, Milan, Italy
| | - David Goldblum
- Pallas-Kliniken, Olten, Bern, Zurich, Switzerland; University of Basel, Basel, Switzerland
| | - Preeya K Gupta
- Triangle Eye Consultants, Raleigh, NC, USA; Department of Ophthalmology, Tulane University, New Orleans, LA, USA
| | - Jesus Merayo-Lloves
- Instituto Universitario Fernandez-Vega, Universidad de Oviedo, Principality of Asturias, Spain
| | - Kyung-Sun Na
- Department of Ophthalmology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Shehzad A Naroo
- College of Health and Life Sciences, Aston University, Birmingham, UK
| | - Kelly K Nichols
- School of Optometry, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Eduardo M Rocha
- Department of Ophthalmology, Othorynolaringology and Head & Neck Surgery, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Louis Tong
- Cornea and External Eye Disease Service, Singapore National Eye Center, Ocular Surface Research Group, Singapore Eye Research Institute, Eye Academic Clinical Program, Duke-National University of Singapore, Singapore
| | - Michael T M Wang
- Department of Ophthalmology, New Zealand National Eye Centre, The University of Auckland, Auckland, New Zealand
| | - Jennifer P Craig
- Department of Ophthalmology, New Zealand National Eye Centre, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
45
|
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
|
46
|
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
|
47
|
Ma K, Liu J, Sha Y, Zhang W, Ni Z, Kong X, Xiang M. Qi Jing Mingmu decoction inhibits the p38 signaling pathway in conjunctivochalasis fibroblasts by down-regulation of Th17 cell differentiation. JOURNAL OF ETHNOPHARMACOLOGY 2023; 301:115812. [PMID: 36223843 DOI: 10.1016/j.jep.2022.115812] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 09/25/2022] [Accepted: 10/04/2022] [Indexed: 06/16/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Qi Jing Mingmu (QJMM) decoction is a traditional Chinese medicine that has been widely used for the clinical treatment of conjunctivochalasis (CCH). It is an effective treatment to relieve ocular symptoms including improving tear film and promoting tear secretion. However, its effects and molecular mechanisms need to be elucidated. AIM OF THE STUDY To determine whether QJMM decoction affected T helper 17 (Th17) cell differentiation of CCH patients. MATERIALS AND METHODS Blood samples and conjunctival tissues were collected from CCH patients and normal controls. The fibroblasts were separately induced, and CD4+ T cells were incubated with increasing concentrations of QJMM decoction and co-cultured with CCH fibroblasts. Th17 cell numbers were then analyzed using flow cytometry. Serum levels of interleukin 17 (IL-17) and IL-22 were detected using enzyme-linked immunosorbent assays. The expressions of signal proteins and genes were detected using western blotting and quantitative real-time PCR. RESULTS Compared with normal controls, Th17 cell numbers and serum levels of IL-17 and IL-22 were elevated in patients with CCH. QJMM decoction down-regulated the expressions of IL-17, IL-22, and STAT3 of CD4+T cells from CCH patients, suggesting that QJMM decoction impeded Th17 cell differentiation. QJMM decoction-treated CD4+ T cells inhibited the expression of p38 in CCH fibroblasts. CONCLUSION QJMM decoction inhibited Th17 cell differentiation of CD4+T cells from CCH patients, and QJMM decoction-treated CD4+T cells down-regulated the p38 signal pathway in CCH fibroblasts. Our study showed that Th17 cells may be good candidates for clinical treatment of CCH.
Collapse
Affiliation(s)
- Kai Ma
- Department of Ophthalmology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China.
| | - Jiang Liu
- Department of Ophthalmology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China.
| | - Yongyi Sha
- Department of Ophthalmology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China.
| | - Wei Zhang
- Department of Ophthalmology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China.
| | - Zhenhua Ni
- Department of Central Lab, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China.
| | - Xueqing Kong
- Department of Ophthalmology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China.
| | - Minhong Xiang
- Department of Ophthalmology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China; Shanghai Putuo Central School of Clinical Medicine, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
48
|
Sharma S, Tyagi K, Dang S. Use of nanotechnology in dry eye syndrome. NANOTECHNOLOGY IN OPHTHALMOLOGY 2023:227-246. [DOI: 10.1016/b978-0-443-15264-1.00010-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
49
|
Sekhon AS, He B, Iovieno A, Yeung SN. Pathophysiology of Corneal Endothelial Cell Loss in Dry Eye Disease and Other Inflammatory Ocular Disorders. Ocul Immunol Inflamm 2023; 31:21-31. [PMID: 34678119 DOI: 10.1080/09273948.2021.1980808] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
PURPOSE Dry eye disease (DED) and other inflammatory ocular disorders have been reported to be associated with decreased corneal endothelial cell density (CECD), however the mechanism of underlying endothelial cell loss remains unknown. METHODS We conducted a comprehensive literature search of English-written publications on dry eye disease, corneal endothelial cell loss, Sjögren's syndrome, and Graft Vs Host Disease (GVHD), to review the effects of DED and other inflammatory ocular surface conditions on CECD. RESULTS A total of 78 studies were included in our study. Loss of corneal neurotrophic support, cytotoxic stress, and a heightened immune response, all of which may occur secondarily to a common causative agent such as inflammation, are major contributors to reduced CECD. CONCLUSION More studies are needed to determine how the interrelated pathways of altered corneal nerve function and upregulated expression of inflammatory activity influence corneal endothelial cell loss.
Collapse
Affiliation(s)
- Amardeep S Sekhon
- Department of Anesthesiology, Pharmacology and Therapeutics, Faculty of Medicine, University of British Columbia, Vancouver, Canada
| | - Bonnie He
- Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Alfonso Iovieno
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, Canada
| | - Sonia N Yeung
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, Canada
| |
Collapse
|
50
|
Saccu G, Menchise V, Gai C, Bertolin M, Ferrari S, Giordano C, Manco M, Dastrù W, Tolosano E, Bussolati B, Calautti E, Camussi G, Altruda F, Fagoonee S. Bone Marrow Mesenchymal Stromal/Stem Cell-Derived Extracellular Vesicles Promote Corneal Wound Repair by Regulating Inflammation and Angiogenesis. Cells 2022; 11:3892. [PMID: 36497151 PMCID: PMC9736484 DOI: 10.3390/cells11233892] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 11/22/2022] [Accepted: 11/28/2022] [Indexed: 12/05/2022] Open
Abstract
Severe corneal damage leads to complete vision loss, thereby affecting life quality and impinging heavily on the healthcare system. Current clinical approaches to manage corneal wounds suffer from severe drawbacks, thus requiring the development of alternative strategies. Of late, mesenchymal stromal/stem cell (MSC)-derived extracellular vesicles (EVs) have become a promising tool in the ophthalmic field. In the present study, we topically delivered bone-marrow-derived MSC-EVs (BMSC-EVs), embedded in methylcellulose, in a murine model of alkali-burn-induced corneal damage in order to evaluate their role in corneal repair through histological and molecular analyses, with the support of magnetic resonance imaging. Our data show that BMSC-EVs, used for the first time in this specific formulation on the damaged cornea, modulate cell death, inflammation and angiogenetic programs in the injured tissue, thus leading to a faster recovery of corneal damage. These results were confirmed on cadaveric donor-derived human corneal epithelial cells in vitro. Thus, BMSC-EVs modulate corneal repair dynamics and are promising as a new cell-free approach for intervening on burn wounds, especially in the avascularized region of the eye.
Collapse
Affiliation(s)
- Gabriele Saccu
- Molecular Biotechnology Center “Guido Tarone”, Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Turin, Italy
| | - Valeria Menchise
- Institute of Biostructure and Bioimaging, National Research Council (CNR), Molecular Biotechnology Center “Guido Tarone”, 10126 Turin, Italy
| | - Chiara Gai
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy
| | | | | | - Cristina Giordano
- Ophthalmology Veterinary Practice, C.so Galileo Ferraris 121, 10126 Turin, Italy
| | - Marta Manco
- Molecular Biotechnology Center “Guido Tarone”, Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Turin, Italy
| | - Walter Dastrù
- Molecular Biotechnology Center “Guido Tarone”, Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Turin, Italy
| | - Emanuela Tolosano
- Molecular Biotechnology Center “Guido Tarone”, Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Turin, Italy
| | - Benedetta Bussolati
- Molecular Biotechnology Center “Guido Tarone”, Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Turin, Italy
| | - Enzo Calautti
- Molecular Biotechnology Center “Guido Tarone”, Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Turin, Italy
| | - Giovanni Camussi
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy
| | - Fiorella Altruda
- Molecular Biotechnology Center “Guido Tarone”, Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Turin, Italy
| | - Sharmila Fagoonee
- Institute of Biostructure and Bioimaging, National Research Council (CNR), Molecular Biotechnology Center “Guido Tarone”, 10126 Turin, Italy
| |
Collapse
|