1
|
El Fekih L, Khairallah M, Ben Amor H, Mahmoud A, Chiambaretta F, Messaoud R. Successful management of dry eye disease with a new eye drop formulation combining hyaluronic acid, trehalose, and N-acetyl-aspartyl-glutamic acid (NAAGA). J Fr Ophtalmol 2024; 47:104169. [PMID: 38838456 DOI: 10.1016/j.jfo.2024.104169] [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: 07/28/2023] [Accepted: 10/11/2023] [Indexed: 06/07/2024]
Abstract
PURPOSE To assess the clinical efficacy and safety of T2769, a new preservative-free eye drop combining hyaluronic acid (HA), trehalose and N-acetyl-aspartyl-glutamic acid (NAAGA), in dry eye patients. PATIENTS AND METHODS This was a multicenter, non-comparative, open-label study. After a run-in period with NaCl 0.9% solution, 62 patients with moderate-to-severe dry eye disease (DED) were included and treated with T2769, 3 to 6 times/day for 42 days. The primary efficacy endpoint was the change in global ocular symptomatology assessed on a visual analog scale between Day 1 (D1) and D42. Other efficacy endpoints included ocular surface disease index (OSDI), soothing sensation, individual dry eye symptoms, conjunctival hyperemia, global ocular staining, tear break-up time, Schirmer test, and global efficacy assessed by the investigator. Safety was assessed throughout the study. RESULTS A marked reduction in ocular symptomatology was observed from D1 to D42 with a mean change of -55.9±23.1mm (P<0.001). This was accompanied by a mean change in OSDI score from baseline of -44.6±15.9 (P<0.001) and a substantial soothing sensation in 82.3% of patients. Clinically significant improvements were observed for all ocular symptoms (mainly burning/irritation, stinging/pain, feeling of ocular dryness, foreign body sensation, itching/pruritus). Conjunctival hyperemia significantly decreased in 96.8% of patients (56.5% displayed no conjunctival hyperemia at D42 versus 0% at baseline). All signs and symptoms improved by D14 and further improved at D42. Investigators assessed the ocular efficacy of T2769 as very satisfactory or satisfactory for 91.9% of patients at D42. T2769 was well tolerated, with no ocular adverse events and only a few ocular symptoms upon instillation. CONCLUSION Management of moderate-to-severe DED patients with the new formulation T2769, combining NAAGA to HA and trehalose, led to rapid and significant improvements in dry eye signs and symptoms with good tolerability.
Collapse
Affiliation(s)
- L El Fekih
- Department of Ophthalmology, Internal Security Forces Hospital, La Marsa, Tunisia; Faculty of Medicine, Tunis-El Manar University, Tunis, Tunisia.
| | - M Khairallah
- Fattouma-Bourguiba University Hospital, Monastir, Tunisia
| | - H Ben Amor
- Fattouma-Bourguiba University Hospital, Monastir, Tunisia
| | - A Mahmoud
- Department of Ophthalmology, Tahar Sfar University Hospital, Mahdia, Tunisia
| | - F Chiambaretta
- Department of Ophthalmology, University Hospital Gabriel-Montpied, Clermont-Ferrand, France
| | - R Messaoud
- Department of Ophthalmology, Tahar Sfar University Hospital, Mahdia, Tunisia
| |
Collapse
|
2
|
Valencia-Nieto L, Pinto-Fraga J, Blanco-Vázquez M, Fernández I, López-Miguel A, García-Vázquez C, González-García MJ, Enríquez-de-Salamanca A, Calonge M. Short-Term Efficacy of Ophthalmic Cyclosporine: A 0.1% Cationic Emulsion in Dry Eye Patients Assessed Under Controlled Environment. Ophthalmol Ther 2024; 13:1197-1210. [PMID: 38446281 DOI: 10.1007/s40123-024-00906-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 02/05/2024] [Indexed: 03/07/2024] Open
Abstract
INTRODUCTION To evaluate the short-term efficacy of cyclosporine A (CsA)-0.1% cationic emulsion (CE) in patients with dry eye disease (DED) and mitigation of the inflammatory flares triggered by desiccating stress environments. METHODS A single-center non-randomized clinical trial was performed at a tertiary care setting. Twenty patients with DED treated with CsA 0.1% CE were exposed to a normal controlled environment (NCE) (23 °C, 50% relative humidity) and an adverse controlled environment (ACE) (23 °C, 10% relative humidity, 0.43 m/s localized airflow) during baseline and the 1- and 3-month visits. Patients underwent the following evaluations: conjunctival hyperemia and staining, corneal fluorescein staining (CFS) using the Oxford and Cornea and Contact Lens Research Unit (CCLRU) scale, meibomian gland (MG) secretion quality, Dry Eye Questionnaire-5, Symptom Assessment in Dry Eye (SANDE II), and Change in Dry Eye Symptoms Questionnaire. Multivariate models were adjusted for statistical analysis. RESULTS Nineteen women and one man (mean age, 58.9 ± 12.3 years) completed the study. All symptom questionnaires, CFS, conjunctival hyperemia and staining, and MG secretion quality improved (p ≤ 0.003) with 1 month of treatment; improvements were maintained after 3 months (p ≤ 0.02), except for SANDE II (p ≥ 0.07). The CFS worsening (total CCLRU) after baseline ACE exposure (from 8.6 to 10.1) was higher, although not significant (p = 0.64), compared with 1 month (from 5.4 to 5.8) and 3 months (from 5.0 to 5.9) after treatment. CONCLUSION Topical CsA-0.1% CE improved DED signs and symptoms after 1 month of treatment under controlled environmental conditions. Future studies should confirm the benefit of CsA-0.1% CE in desiccating stress environments. TRIAL REGISTRATION ClinicalTrials.gov identifier, NCT04492878.
Collapse
Affiliation(s)
- Laura Valencia-Nieto
- Instituto de Oftalmobiología Aplicada (IOBA), University of Valladolid, Campus Universitario Miguel Delibes, Paseo de Belén 17, 47011, Valladolid, Spain
| | - José Pinto-Fraga
- Instituto de Oftalmobiología Aplicada (IOBA), University of Valladolid, Campus Universitario Miguel Delibes, Paseo de Belén 17, 47011, Valladolid, Spain
| | - Marta Blanco-Vázquez
- Instituto de Oftalmobiología Aplicada (IOBA), University of Valladolid, Campus Universitario Miguel Delibes, Paseo de Belén 17, 47011, Valladolid, Spain
| | - Itziar Fernández
- Instituto de Oftalmobiología Aplicada (IOBA), University of Valladolid, Campus Universitario Miguel Delibes, Paseo de Belén 17, 47011, Valladolid, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Valladolid, Spain
| | - Alberto López-Miguel
- Instituto de Oftalmobiología Aplicada (IOBA), University of Valladolid, Campus Universitario Miguel Delibes, Paseo de Belén 17, 47011, Valladolid, Spain.
| | - Carmen García-Vázquez
- Instituto de Oftalmobiología Aplicada (IOBA), University of Valladolid, Campus Universitario Miguel Delibes, Paseo de Belén 17, 47011, Valladolid, Spain
| | - María J González-García
- Instituto de Oftalmobiología Aplicada (IOBA), University of Valladolid, Campus Universitario Miguel Delibes, Paseo de Belén 17, 47011, Valladolid, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Valladolid, Spain
| | - Amalia Enríquez-de-Salamanca
- Instituto de Oftalmobiología Aplicada (IOBA), University of Valladolid, Campus Universitario Miguel Delibes, Paseo de Belén 17, 47011, Valladolid, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Valladolid, Spain
| | - Margarita Calonge
- Instituto de Oftalmobiología Aplicada (IOBA), University of Valladolid, Campus Universitario Miguel Delibes, Paseo de Belén 17, 47011, Valladolid, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Valladolid, Spain
| |
Collapse
|
3
|
Zhu X, Li S, Wang M, Yao W, Huang X, Zhao L. Effects of Topical 0.05% Cyclosporine A on Dry Eye Symptoms and Parameters Following Small Incision Lenticule Extraction. J Refract Surg 2024; 40:e229-e238. [PMID: 38593259 DOI: 10.3928/1081597x-20240311-03] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2024]
Abstract
PURPOSE To evaluate the effects of topical 0.05% cyclosporine A on Ocular Surface Disease Index (OSDI) score and ocular surface parameters after small incision lenticule extraction (SMILE) for myopia. METHODS In this study, 151 patients who underwent SMILE were randomized into the control group (71 eyes) and the 0.05% cyclosporine A group (80 eyes). Both groups received standard treatment during the 1 month after SMILE. Over the next 3 months, The control group continued standard therapy (0.3% sodium hyaluronate) and the 0.05% cyclosporine A group received additional 0.05% cyclosporine A. OSDI total and subscale scores, non-invasive tear break-up time (NIBUT), tear lipid layer thickness (LLT), and tear meniscus height (TMH) were assessed preoperatively and postoperatively. RESULTS Compared to baseline, the OSDI scores significantly increased in both groups (P < .001). The 0.05% cyclosporine A group exhibited lower OSDI total scores after administering 0.05% cyclosporine A versus the control group (P = .026). At 1 month of follow-up, NIBUT, LLT, and TMH values significantly decreased in both groups compared to baseline (P < .05). The 0.05% cyclosporine A group exhibited higher NIBUT, LLT, and TMH versus the control group, returning to preoperative values after 2 months. Overall, the OSDI total score and NIBUT values during follow-up were not significantly different between the two groups; however, the LLT and TMH values were significantly different between the two groups (P < .001 and .041, respectively) by repeated measures analysis of variance. CONCLUSIONS Topical 0.05% cyclosporine A was effective in relieving subjective dry eye symptoms and maintaining ocular surface stability in the early postoperative period of SMILE. [J Refract Surg. 2024;40(4):e229-e238.].
Collapse
|
4
|
Abu-Romman A, Scholand KK, Pal-Ghosh S, Yu Z, Kelagere Y, Yazdanpanah G, Kao WWY, Coulson-Thomas VJ, Stepp MA, de Paiva CS. Conditional deletion of CD25 in the corneal epithelium reveals sex differences in barrier disruption. Ocul Surf 2023; 30:57-72. [PMID: 37516317 PMCID: PMC10812880 DOI: 10.1016/j.jtos.2023.07.008] [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: 05/02/2023] [Revised: 07/06/2023] [Accepted: 07/25/2023] [Indexed: 07/31/2023]
Abstract
PURPOSE IL-2 promotes activation, clonal expansion, and deletion of T cells. IL-2 signals through its heterotrimeric receptor (IL-2R) consisting of the CD25, CD122 and CD132 chains. CD25 knockout (KO) mice develop Sjögren Syndrome-like disease. This study investigates whether corneal CD25/IL-2 signaling is critical for ocular health. METHODS Eyes from C57BL/6 mice were collected and prepared for immunostaining or in-situ hybridization. Bulk RNA sequencing was performed on the corneal epithelium from wild-type and CD25KO mice. We generated a conditional corneal-specific deletion of CD25 in the corneal epithelium (CD25Δ/ΔCEpi). Corneal barrier function was evaluated based on the uptake of a fluorescent dye. Mice were subjected to unilateral corneal debridement, followed by epithelial closure over time. RESULTS In C57BL/6 mice, CD25 mRNA was expressed in ocular tissues. Protein expression of CD25, CD122, and CD132 was confirmed in the corneal epithelium. Delayed corneal re-epithelization was seen in female but not male CD25KO mice. There were 771 differentially expressed genes in the corneal epithelium of CD25KO compared to wild-type mice. While barrier function is disrupted in CD25Δ/ΔCEpi mice, re-epithelialization rates are not delayed. CONCLUSIONS All three chains of the IL-2R are expressed in the corneal epithelium. Our results indicate for the first time, deleting CD25 systemically in all tissues in the mouse and deleting CD25 locally in just the corneal epithelium compromises corneal epithelial barrier function, leading to dry eye disease in female mice. Future studies are needed to delineate the pathways used by IL-2 signaling to influence cornea homeostasis.
Collapse
Affiliation(s)
- Anmar Abu-Romman
- Ocular Surface Center, Department of Ophthalmology, Cullen Eye Institute, Baylor College of Medicine, Houston, TX, United States.
| | - Kaitlin K Scholand
- Ocular Surface Center, Department of Ophthalmology, Cullen Eye Institute, Baylor College of Medicine, Houston, TX, United States; Department of Biosciences, Rice University, Houston, TX, United States.
| | - Sonali Pal-Ghosh
- Department of Anatomy and Cell Biology, The George Washington University School of Medicine and Health Sciences, Washington, DC, United States.
| | - Zhiyuan Yu
- Ocular Surface Center, Department of Ophthalmology, Cullen Eye Institute, Baylor College of Medicine, Houston, TX, United States.
| | - Yashaswini Kelagere
- Ocular Surface Center, Department of Ophthalmology, Cullen Eye Institute, Baylor College of Medicine, Houston, TX, United States.
| | - Ghasem Yazdanpanah
- Ocular Surface Center, Department of Ophthalmology, Cullen Eye Institute, Baylor College of Medicine, Houston, TX, United States.
| | - Winston W-Y Kao
- Department of Ophthalmology, University of Cincinnati, Cincinnati, OH, United States.
| | | | - Mary Ann Stepp
- Department of Anatomy and Cell Biology, The George Washington University School of Medicine and Health Sciences, Washington, DC, United States; Department of Ophthalmology, The George Washington University School of Medicine and Health Sciences, Washington, DC, United States.
| | - Cintia S de Paiva
- Ocular Surface Center, Department of Ophthalmology, Cullen Eye Institute, Baylor College of Medicine, Houston, TX, United States; Department of Biosciences, Rice University, Houston, TX, United States.
| |
Collapse
|
5
|
Gordhan HM, Miller ST, Clancy DC, Ina M, McDougal AV, Cutno DK, Brown RV, Lichorowic CL, Sturdivant JM, Vick KA, Williams SS, deLong MA, White JC, Kopczynski CC, Ellis DA. Eyes on Topical Ocular Disposition: The Considered Design of a Lead Janus Kinase (JAK) Inhibitor That Utilizes a Unique Azetidin-3-Amino Bridging Scaffold to Attenuate Off-Target Kinase Activity, While Driving Potency and Aqueous Solubility. J Med Chem 2023. [PMID: 37314941 DOI: 10.1021/acs.jmedchem.3c00519] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
An unmet medical need remains for patients suffering from dry eye disease (DED). A fast-acting, better-tolerated noncorticosteroid anti-inflammatory eye drop could improve patient outcomes and quality of life. Herein, we describe a small-molecule drug discovery effort to identify novel, potent, and water-soluble JAK inhibitors as immunomodulating agents for topical ocular disposition. A focused library of known 3-(4-(2-(arylamino)pyrimidin-4-yl)-1H-pyrazol-1-yl)propanenitriles was evaluated as a molecular starting point. Structure-activity relationships (SARs) revealed a ligand-efficient (LE) JAK inhibitor series, amenable to aqueous solubility. Subsequent in vitro analysis indicated the potential for off-target toxicity. A KINOMEscan selectivity profile of 5 substantiated the likelihood of widespread series affinity across the human kinome. An sp2-to-sp3 drug design strategy was undertaken to attenuate off-target kinase activity while driving JAK-STAT potency and aqueous solubility. Tactics to reduce aromatic character, increase fraction sp3 (Fsp3), and bolster molecular complexity led to the azetidin-3-amino bridging scaffold in 31.
Collapse
Affiliation(s)
- Heeren M Gordhan
- Alcon Research, LLC, Durham, North Carolina 27703, United States
| | - Steven T Miller
- Alcon Research, LLC, Durham, North Carolina 27703, United States
| | - Daphne C Clancy
- Alcon Research, LLC, Durham, North Carolina 27703, United States
| | - Maria Ina
- Alcon Research, LLC, Durham, North Carolina 27703, United States
| | - Alan V McDougal
- Alcon Research, LLC, Durham, North Carolina 27703, United States
| | - D'Quan K Cutno
- Alcon Research, LLC, Durham, North Carolina 27703, United States
| | - Robert V Brown
- Alcon Research, LLC, Durham, North Carolina 27703, United States
| | | | | | - Kyle A Vick
- ID Business Solutions, Ltd., Boston, Massachusetts 02210, United States
| | | | | | - Jeffrey C White
- Baxter Healthcare Corp., Deerfield, Illinois 60015, United States
| | | | - David A Ellis
- Alcon Research, LLC, Durham, North Carolina 27703, United States
| |
Collapse
|
6
|
Monfared N, Murphy PJ. Features and influences on the normal tear evaporation rate. Cont Lens Anterior Eye 2023; 46:101809. [PMID: 36621341 DOI: 10.1016/j.clae.2022.101809] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 12/23/2022] [Accepted: 12/28/2022] [Indexed: 01/09/2023]
Abstract
Tear evaporation is a normal physiological phenomenon that has an important role in regulating blink activity and tear production. An altered tear evaporation rate (TER) is a defining characteristic of evaporative dry eye disease (DED), and the measurement of tear evaporation is a useful clinical test for diagnosis. Reported values for a normal TER cover a broad range, which may be due to the influence of ocular, environmental, and systemic factors. For improved disease diagnosis, a fuller understanding of the normal TER range is essential. This paper reports on a literature review of the current knowledge of these normal influences on TER.
Collapse
Affiliation(s)
- Naeimeh Monfared
- School of Optometry and Vision Science, University of Waterloo, Waterloo, Canada.
| | - Paul J Murphy
- School of Optometry and Vision Science, University of Waterloo, Waterloo, Canada
| |
Collapse
|
7
|
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
|
8
|
Nguyen A, Kolluru A, Beglarian T. Dry eye disease: A review of anti-inflammatory therapies. Taiwan J Ophthalmol 2023; 13:3-12. [DOI: 10.4103/2211-5056.369606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 11/14/2022] [Indexed: 02/15/2023] Open
|
9
|
Effect of a single vectored thermal pulsation treatment of Meibomian gland dysfunction patients under controlled environmental conditions. Sci Rep 2022; 12:16761. [PMID: 36202938 PMCID: PMC9537290 DOI: 10.1038/s41598-022-20994-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 09/21/2022] [Indexed: 11/09/2022] Open
Abstract
To assess the prophylactic effect of LipiFlow treatment in Meibomian gland dysfunction (MGD) patients exposed to an adverse environmental humidity. MGD patients were exposed to normal (23 °C; 50% relative humidity; 30 min) and adverse (23 °C; 10% relative humidity; 2 h) controlled environments consecutively during baseline and follow-up visits (3, 6, and 12 months) after a single LipiFlow treatment. Ocular Surface Disease Index (OSDI), lipid layer thickness (LLT), fluorescein tear break-up time (TBUT), corneal and conjunctival staining, change in dry eye symptoms questionnaire (CDES-Q), and Meibomian gland yielding liquid secretion (MGYLS), were assessed. Linear mixed-effects and cumulative logit mixed models were fitted to assess the effect of the LipiFlow treatment over time and within the controlled environments. Seventeen females and 4 males (59.6 ± 9.4 years) completed the study. LLT and TBUT did not vary significantly (p > 0.05) after LipiFlow treatment. OSDI, corneal and conjunctival staining, and MGYLS scores were improved (p ≤ 0.01) 12 months after treatment. After the adverse exposure, corneal staining increased at all visits (p = 0.01), and there was no significant improvement in CDES-Q scores after LipiFlow treatment (p ≥ 0.07). One LipiFlow treatment improved objective and subjective outcomes in MGD disease for at least one year. Further studies are needed to support that LipiFlow might also help as an adjuvant to avoid acute flares against an adverse environmental humidity.
Collapse
|
10
|
Gut-derived butyrate suppresses ocular surface inflammation. Sci Rep 2022; 12:4512. [PMID: 35296712 PMCID: PMC8927112 DOI: 10.1038/s41598-022-08442-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 02/23/2022] [Indexed: 12/16/2022] Open
Abstract
Dry eye is a common ocular inflammatory disorder characterized by tear film instability and reduced tear production. There is increasing evidence that homeostasis of the ocular surface is impacted by the intestinal microbiome. We are interested in investigating the potential role of microbially produced small molecules in mediating the interaction between the intestinal microbiota and the ocular surface. One such molecule is butyrate, a short-chain fatty acid (SCFA) produced by certain members of the gut microbiota through fermentation of dietary fiber. Here we show that SCFA transporter SLC5A8 is expressed in vivo in murine conjunctival and corneal epithelium. Pre-treatment of in vitro corneal epithelial cultures or bone marrow-derived dendritic cells (BMDCs) with phenylbutyrate (PBA) reduces lipopolysaccharide-induced pro-inflammatory Tnf expression. Corneal epithelial cultures and BMDCs isolated from Slc5a8 knockout mice are unable to respond to PBA pre-treatment, suggesting that SLC5A8 is required for the protective effect of PBA. The treatment of mice undergoing desiccating stress (DS) with oral tributyrin, a prodrug form of butyrate, reduces inflammation at the ocular surface in vivo, and this effect partially requires SLC5A8. Finally, expression analysis on conjunctival tissue isolated from mice subjected to DS with and without tributyrin treatment revealed that treatment downregulated genes involved in Type I interferon signaling. Together these data support our hypothesis that SCFAs produced in the gut participate in the maintenance of ocular surface homeostasis.
Collapse
|
11
|
Liu Z, Chen D, Chen X, Bian F, Qin W, Gao N, Xiao Y, Li J, Pflugfelder SC, Li DQ. Trehalose Induces Autophagy Against Inflammation by Activating TFEB Signaling Pathway in Human Corneal Epithelial Cells Exposed to Hyperosmotic Stress. Invest Ophthalmol Vis Sci 2021; 61:26. [PMID: 32785678 PMCID: PMC7441355 DOI: 10.1167/iovs.61.10.26] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Purpose Autophagy plays an important role in balancing the inflammatory response to restore homeostasis. The aim of this study was to explore the mechanism by which trehalose suppresses inflammatory cytokines via autophagy activation in primary human corneal epithelial cells (HCECs) exposed to hyperosmotic stress. Methods An in vitro dry eye model was used in which HCECs were cultured in hyperosmolar medium with the addition of sodium chloride (NaCl). Trehalose was applied in different concentrations. The levels of TNF-α, IL-1β, IL-6, and IL-8 were detected using RT-qPCR and ELISA. Cell viability assays, immunofluorescent staining of LC3B, and western blots of Beclin1, Atg5, Atg7, LC3B, and P62 were conducted. The key factors in upstream signaling pathways of autophagy activation were measured: P-Akt, Akt, and transcription factor EB (TFEB). Results Trehalose reduced the proinflammatory mediators TNF-α, IL-1β, IL-6, and IL-8 in primary HCECs at 450 mOsM. This effect was osmolarity dependent, and a level of 1.0% trehalose showed the most suppression. Trehalose promoted autophagosome formation and autophagic flux, as evidenced by increased production of Beclin1, Atg5, and Atg7, as well as higher LC3B I protein turnover to LC3B II, with decreased protein levels of P62/SQSTM1. The addition of 3-methyladenine blocked autophagy activation and increased the release of proinflammatory cytokines. Trehalose further activated TFEB, with translocation from cytoplasm to the nucleus, but diminished Akt activity. Conclusions Our findings demonstrate that trehalose, functioning as an autophagy enhancer, suppresses the inflammatory response by promoting autophagic flux via TFEB activation in primary HCECs exposed to hyperosmotic stress, a process that is beneficial to dry eye.
Collapse
Affiliation(s)
- Zhao Liu
- Ocular Surface Center, Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, United States.,Department of Ophthalmology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Ding Chen
- Ocular Surface Center, Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, United States.,School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xin Chen
- Ocular Surface Center, Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, United States.,School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Fang Bian
- Ocular Surface Center, Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, United States
| | - Wenjuan Qin
- Ocular Surface Center, Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, United States
| | - Ning Gao
- Ocular Surface Center, Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, United States.,Department of Ophthalmology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yangyan Xiao
- Ocular Surface Center, Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, United States
| | - Jinmiao Li
- Ocular Surface Center, Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, United States
| | - Stephen C Pflugfelder
- Ocular Surface Center, Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, United States
| | - De-Quan Li
- Ocular Surface Center, Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, United States
| |
Collapse
|
12
|
Perez VL, Stern ME, Pflugfelder SC. Inflammatory basis for dry eye disease flares. Exp Eye Res 2020; 201:108294. [PMID: 33039458 PMCID: PMC7736538 DOI: 10.1016/j.exer.2020.108294] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 10/02/2020] [Accepted: 10/05/2020] [Indexed: 12/15/2022]
Abstract
Most patients with chronic dry eye disease (DED) have episodic flares, which can be triggered by a variety of activities and environmental stresses. These flares are typically associated with rapid exacerbation of discomfort symptoms, followed by prolonged elevation of inflammation. In an acute flare, ocular surface inflammation begins with a nonspecific innate immune response, in some cases followed by a slower but more specific adaptive immune response. At the ocular surface, epithelial cells are central to the innate immune response, and we discuss their role in DED flares alongside the other core components. Epithelial cells and other cells of the innate response (neutrophils, monocytes, macrophages and dendritic cells) trigger flares in response to increased osmolarity, detected via pattern receptors on their cell surface. Ultimately, downstream signaling pathways activate innate and adaptive immune responses, with consequent inflammation and symptoms. In chronic DED, pathogenic T cells have infiltrated the ocular surface tissues. The established adaptive immune response is likely to lead to flare-ups at lower thresholds of stress, with inflammation maintained over a longer period. Increased understanding of the inflammatory cascades activated during a flare may guide management and improve outcomes.
Collapse
Affiliation(s)
- Victor L Perez
- Duke University School of Medicine, Durham, NC, United States
| | | | | |
Collapse
|
13
|
The cornea in keratoconjunctivitis sicca. Exp Eye Res 2020; 201:108295. [PMID: 33038387 DOI: 10.1016/j.exer.2020.108295] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 10/04/2020] [Accepted: 10/06/2020] [Indexed: 12/22/2022]
Abstract
The lacrimal functional unit (LFU) regulates tear production, composition, distribution and clearance to maintain a stable protective tear layer that is essential for maintaining corneal epithelial health. Dysfunction of the LFU, commonly referred to as dry eye, leads to increased tear osmolarity and levels of inflammatory mediators in tears that cause ocular surface epithelial disease, termed keratoconjunctivitis sicca (KCS). Corneal changes in KCS include glycocalyx loss, barrier disruption, surface irregularity inflammatory cytokine/chemokine production, cornification and apoptosis. These can reduce visual function and the increased shear force on the corneal epithelium can stimulate nociceptors sensitized by inflammation causing irritation and pain that may precede frank clinical signs. Therapy of keratoconjunctivitis sicca should be tailored to improve tear stability, normalize tear composition, improve barrier function and minimize shear forces and damaging inflammation to improve corneal epithelial health.
Collapse
|
14
|
Favre H, Lahoti S, Issa N, Johnson DA, Kheirkhah A. Topical Steroids in Management of Dry Eye Disease. CURRENT OPHTHALMOLOGY REPORTS 2020. [DOI: 10.1007/s40135-020-00249-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
15
|
Usuba FS, Saad CGS, Aikawa NE, Novaes P, Moraes JCB, Santo RM, Carvalho JF, Bonfá E, Alves MR. Improvement of conjunctival cytological grade and tear production in Ankylosing Spondylitis patients under TNF inhibitors: a long-term follow-up. Sci Rep 2020; 10:334. [PMID: 31942038 PMCID: PMC6962203 DOI: 10.1038/s41598-019-57266-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 12/26/2019] [Indexed: 01/12/2023] Open
Abstract
Dry eye disease can compromise the patient’s quality of life. Few studies assessed the ocular surface (OS) in Ankylosing Spondylitis (AS) patients. This study aimed to evaluate the clinical and cytological findings of the OS in patients with AS, classify dry eye disease (DED) severity grade and conjunctival impression cytology (IC), and the effects of TNF inhibitors (TNFi) in a one-year follow-up. A baseline (BL) evaluation included 36 AS patients and 39 healthy controls. They fulfilled the Ocular Surface Index Disease questionnaire and underwent the Schirmer I test, break-up time, vital staining, and conjunctival IC. A DED severity grade, as well as IC rating, was applied. Fourteen of these patients received TNFi and analysis of ocular and systemic AS disease parameters occurred at BL and three months (3 M), and 12 months (12 M) after treatment. The AS patients presented a higher frequency of DED (p = 0.01), a worse score of severity (p = 0.001), and a higher frequency of altered IC (p = 0.007) when compared to controls. The 14 patients under TNFi presented an improvement in all the clinical disease activity parameters throughout the one-year treatment (p < 0.05) even as a concomitant increase in the Schirmer test (p = 0.04), and a significant amelioration in the altered IC to a normal IC (p = 0.006). DED is a frequent and under-diagnosed ocular disease in AS patients. The long-term parallel improvement of disease activity and OS parameters in AS patients receiving TNFi suggests that the OS can be an additional target of systemic inflammation in AS.
Collapse
Affiliation(s)
- Fany Solange Usuba
- Ophthalmology Department, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil.
| | - Carla Gonçalves Schahin Saad
- Rheumatology Division, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Nadia Emi Aikawa
- Rheumatology Division, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Priscila Novaes
- Ophthalmology Department, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Julio Cesar Bertacini Moraes
- Rheumatology Division, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Ruth Miyuki Santo
- Ophthalmology Department, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Jozelio Freire Carvalho
- Rheumatology Division, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Eloisa Bonfá
- Rheumatology Division, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Milton Ruiz Alves
- Ophthalmology Department, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
16
|
Thia ZZ, Tong L. Update on the role of impression cytology in ocular surface disease. Taiwan J Ophthalmol 2019; 9:141-149. [PMID: 31572650 PMCID: PMC6759557 DOI: 10.4103/tjo.tjo_57_19] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 07/17/2019] [Indexed: 12/11/2022] Open
Abstract
Understanding of the molecular pathology of ocular surface disease (OSD) is poor, and treatment is highly unsatisfactory. To facilitate treatment of OSD, a relatively noninvasive procedure, i.e. impression cytology (IC) has been shown to be useful. Recently, the technologies employed in research studies using IC in OSD have vastly improved, and standardized IC has even been used in clinical trials of dry eye. Here, this review aims to describe the advances of IC in the last 10 years, which serves as an update on the progress in this field since the last major review of IC. OSD that has been recently evaluated include meibomian gland dysfunction, Sjogren's syndrome, Steven–Johnson syndrome, and postmenopausal dry eye. The recent studies (4 longitudinal, 18 cross-sectional analyses) which utilized IC analyzed DNA, RNA, proteins, and ocular surface cells, including memory T-lymphocytes, dendritic cells (DCs), neutrophils, conjunctival epithelial cells, and goblet cells. These studies employed quantification of transcripts associated with inflammation, proteins involved in oxidative stress, enzymes such as matrix metalloproteinases, and cell surface proteins by flow cytometry, such as HLA-DR, cytokine and chemokine receptors, markers for T cell differentiation, and DC activation, in addition to the more traditional morphological evaluation of squamous metaplasia and staining for goblet cells. Some challenges in the clinical use of IC have also been described, including issues related to storage and normalization of data. In summary, advances in IC have permitted a more robust evaluation of the ocular surface and will facilitate progress in the understanding and treatment of OSD.
Collapse
Affiliation(s)
- Zhang-Zhe Thia
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Louis Tong
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Corneal and External Eye Disease Service, Singapore National Eye Center, Singapore.,Singapore Eye Research Institute, Singapore.,Eye-academic Clinical Program, Duke-NUS Medical School, Singapore
| |
Collapse
|
17
|
Fernández I, López-Miguel A, Enríquez-de-Salamanca A, Tesón M, Stern ME, González-García MJ, Calonge M. Response profiles to a controlled adverse desiccating environment based on clinical and tear molecule changes. Ocul Surf 2019; 17:502-515. [DOI: 10.1016/j.jtos.2019.03.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Revised: 03/14/2019] [Accepted: 03/26/2019] [Indexed: 12/18/2022]
|
18
|
Review of Biomarkers in Ocular Matrices: Challenges and Opportunities. Pharm Res 2019; 36:40. [PMID: 30673862 PMCID: PMC6344398 DOI: 10.1007/s11095-019-2569-8] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 01/07/2019] [Indexed: 02/05/2023]
Abstract
Biomarkers provide a powerful and dynamic approach to improve our understanding of the mechanisms underlying ocular diseases with applications in diagnosis, disease modulation or for predicting and monitoring of clinical response to treatment. Defined as measurable indicator of normal or pathological processes, biomarker evaluation has been used extensively in drug development within clinical settings to better comprehend effectiveness of treatment in ocular diseases. Biomarkers in the eye have the advantage of access to multiple ocular matrices via minimally invasive methods. Repeat sampling for biomarker assessment has enabled reproducible objective measures of disease process or biological responses to a drug treatment. This review describes the usage of biomarkers with respect to four commonly sampled ocular matrices in clinic: tears, conjunctiva, aqueous humor and vitreous. Issues that affect the evaluation of biomarkers are discussed along with opportunities to leverage biomarkers such that ultimately, they can be used for customized targeted therapy.
Collapse
|
19
|
Abstract
Toxic keratoconjunctivitis (TK) is an underrecognized complication of ophthalmic drug use and various environmental or occupational exposures. A detailed history and clinical examination are important to identify the offending agent(s). Common drug-related causes of TK include preservatives in ophthalmic medications, topical antimicrobials, and topical anesthetics. Alternatives to benzalkonium chloride as well as preservative-free formulations should be considered in patients requiring long-term topical medication. More advanced cases of TK may require preservative-free topical steroids and/or antibiotics, and occasionally surgical intervention. Early recognition and appropriate management of TK may help prevent permanent ocular and visual damage.
Collapse
|
20
|
|
21
|
Severity, therapeutic, and activity tear biomarkers in dry eye disease: An analysis from a phase III clinical trial. Ocul Surf 2018; 16:368-376. [DOI: 10.1016/j.jtos.2018.05.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 04/27/2018] [Accepted: 05/08/2018] [Indexed: 12/17/2022]
|
22
|
Boboridis KG, Konstas AGP. Evaluating the novel application of cyclosporine 0.1% in ocular surface disease. Expert Opin Pharmacother 2018; 19:1027-1039. [PMID: 29847195 DOI: 10.1080/14656566.2018.1479742] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Ocular surface disease (OSD) is a highly prevalent symptomatic condition caused by dry eye disease (DED), intrinsic, environmental, or iatrogenic causes. It affects patient's visual function and quality of life. Its pathophysiology is centered on tear hyperosmolarity, inflammation, and epithelial damage. Current management is suboptimal and includes artificial tear supplementation and short-term use of topical steroids in severe cases. The recent approval of cyclosporine 0.1% has transformed management strategies of severe DED and moderate-to-severe OSD. Areas covered: This review summarizes existing information on the efficacy, safety, and tolerability of the new cyclosporine 0.1% formulation. Expert opinion: Topical cyclosporine A 0.1% represents a promising, novel medication for the management of DED, Meibomian gland dysfunction, and inflammatory OSD. It is primarily beneficial for those patients requiring topical immunomodulatory therapy. This topical formulation also has the potential to meaningfully improve the management of moderate-to-severe glaucoma therapy-related OSD. Currently there is limited published clinical data concerning the efficacy of topical cyclosporine. There are, however, theoretical advantages when comparing this cyclosporine formulation with other established commercial preparations. Future research is needed to delineate the precise role and value of this medication.
Collapse
Affiliation(s)
- Konstadinos G Boboridis
- a 1st and 3rd University Departments of Ophthalmology , Aristotle University , Thessaloniki , Greece
| | - Anastasios G P Konstas
- a 1st and 3rd University Departments of Ophthalmology , Aristotle University , Thessaloniki , Greece
| |
Collapse
|
23
|
Calonge M, Labetoulle M, Messmer EM, Shah S, Akova YA, Boboridis KG, Merayo-Lloves J, Aragona P, Benítez-Del-Castillo J, Geerling G, Rolando M, Baudouin C. Controlled Adverse Environment Chambers in Dry Eye Research. Curr Eye Res 2018; 43:445-450. [PMID: 29336696 DOI: 10.1080/02713683.2017.1420197] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Dry eye disease (DED) is a common condition with signs and symptoms that vary depending on a wide range of environmental factors to which people are exposed in their daily lives. Factors such as variable temperature, airflow velocity, relative humidity, seasonality, and pollutants can alter the rate of tear film evaporation, improving or exacerbating symptoms of DED. Results from currently available clinical tests do not always correlate well with patient-reported symptoms, and the continually changing environment and variability in DED symptoms present challenges for the design and conduct of clinical trials. Controlled adverse environment chambers allow standardization of temperature, humidity, and airflow and may minimize potential confounding factors in clinical investigations. Their use can promote accurate study of the pathophysiology of DED, discovery of disease biomarkers, and assessment of the effect of various therapeutic approaches on patients' symptoms. Controlled adverse environment chambers have been used to simulate indoor surroundings such as airplane cabins and to test their effects on contact lens wearers. This review summarizes how these chambers may be useful for the development, approval, and differentiation of potential new treatments for DED.
Collapse
Affiliation(s)
- Margarita Calonge
- a Instituto Oftalmobiologia Aplicada (IOBA), University of Valladolid , Valladolid , Spain.,b CIBER-BBN , Valladolid , Spain
| | - Marc Labetoulle
- c Department of Ophthalmology , Bicêtre University Hospital , Le Kremlin-Bicêtre , France
| | - Elisabeth M Messmer
- d Department of Ophthalmology , Ludwig Maximilian University , Munich , Germany
| | - Sunil Shah
- e Birmingham and Midland Eye Centre , Birmingham City Hospital , Birmingham , UK
| | - Yonca A Akova
- f Department of Ophthalmology , Bayindir Hospital , Ankara , Turkey
| | - Kostas G Boboridis
- g Oculoplastic and Ocular Surface Disease , Aristotle University of Thessaloniki , Thessaloniki , Greece
| | - Jesús Merayo-Lloves
- h Instituto Universitario Fernández-Vega, University of Oviedo , Oviedo , Spain
| | - Pasquale Aragona
- i Department of Biomedical Sciences, University of Messina , Messina , Italy
| | | | - Gerd Geerling
- k Department of Ophthalmology , University Hospital , Düsseldorf , Germany
| | - Maurizio Rolando
- l Department of Neuroscience, Ophthalmology and Genetics , University of Genoa , Genoa , Italy
| | | |
Collapse
|
24
|
Nättinen J, Jylhä A, Aapola U, Enríquez-de-Salamanca A, Pinto-Fraga J, López-Miguel A, González-García MJ, Stern ME, Calonge M, Zhou L, Nykter M, Uusitalo H, Beuerman R. Topical fluorometholone treatment and desiccating stress change inflammatory protein expression in tears. Ocul Surf 2018; 16:84-92. [DOI: 10.1016/j.jtos.2017.09.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 08/07/2017] [Accepted: 09/11/2017] [Indexed: 10/18/2022]
|
25
|
Abstract
Impression cytology (IC) is a technique which permits the retrieval of the outermost layer of ocular surface cells via the use of various types of filters. It is a minimally invasive method of evaluating human conjunctival epithelial cell morphology in the diagnosis of dry eye disease, a common and distressing disorder associated with ageing, contact lens wear, autoimmune disorders and refractive (LASIK) surgery. IC may also be utilized in the diagnosis of other ocular diseases, such as keratoconus and thyroid orbitopathy. More recently, IC has been utilized for the subsequent investigation of gene and protein expression of conjunctival cells in order to identify novel diagnostic biomarkers and to further our understanding of the mechanisms underlying ocular surface disease. This review will therefore examine the literature concerning the role of IC in identifying cellular markers of eye disease, systemic diseases with ocular involvement and potential novel therapeutic targets.
Collapse
Affiliation(s)
- Suzanne Hagan
- Vision Sciences, Dept. of Life Sciences, Glasgow Caledonian University, G4 0BA, Scotland, UK
| |
Collapse
|
26
|
McMonnies CW. Conjunctival Tear Layer Temperature, Evaporation, Hyperosmolarity, Inflammation, Hyperemia, Tissue Damage, and Symptoms: A Review of an Amplifying Cascade. Curr Eye Res 2017; 42:1574-1584. [DOI: 10.1080/02713683.2017.1377261] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Charles W. McMonnies
- School of Optometry and Vision Science, University of New South Wales, Kensington, Australia
| |
Collapse
|
27
|
Pflugfelder SC, de Paiva CS. The Pathophysiology of Dry Eye Disease: What We Know and Future Directions for Research. Ophthalmology 2017; 124:S4-S13. [PMID: 29055361 PMCID: PMC5657523 DOI: 10.1016/j.ophtha.2017.07.010] [Citation(s) in RCA: 313] [Impact Index Per Article: 39.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 07/10/2017] [Accepted: 07/11/2017] [Indexed: 12/31/2022] Open
Abstract
Clinical and laboratory studies performed over the past few decades have discovered that dry eye is a chronic inflammatory disease that can be initiated by numerous extrinsic or intrinsic factors that promote an unstable and hyperosmolar tear film. These changes in tear composition, in some cases combined with systemic factors, lead to an inflammatory cycle that causes ocular surface epithelial disease and neural stimulation. Acute desiccation activates stress signaling pathways in the ocular surface epithelium and resident immune cells. This triggers production of innate inflammatory mediators that stimulate the production of matrix metalloprotease, inflammatory cell recruitment, and dendritic cell maturation. These mediators, combined with exposure of autoantigens, can lead to an adaptive T cell-mediated response. Cornea barrier disruption develops by protease-mediated lysis of epithelial tight junctions, leading to accelerated cell death; desquamation; an irregular, poorly lubricated cornea surface; and exposure and sensitization of epithelial nociceptors. Conjunctival goblet cell dysfunction and death are promoted by the T helper 1 cytokine interferon gamma. These epithelial changes further destabilize the tear film, amplify inflammation, and create a vicious cycle. Cyclosporine and lifitegrast, the 2 US Food and Drug Administration-approved therapies, inhibit T-cell activation and cytokine production. Although these therapies represent a major advance in dry eye therapy, they are not effective in improving discomfort and corneal epithelial disease in all patients. Preclinical studies have identified other potential therapeutic targets, biomarkers, and strategies to bolster endogenous immunoregulatory pathways. These discoveries will, it is hoped, lead to further advances in diagnostic classification and treatment.
Collapse
Affiliation(s)
| | - Cintia S de Paiva
- Department of Ophthalmology, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
28
|
Rodriguez JD, Lane KJ, Ousler GW, Angjeli E, Smith LM, Abelson MB. Blink: Characteristics, Controls, and Relation to Dry Eyes. Curr Eye Res 2017; 43:52-66. [PMID: 29043838 DOI: 10.1080/02713683.2017.1381270] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Blink is a complex phenomenon that is profoundly affected by diverse endogenous and exogenous stimuli. It has been studied in the context of cognition, emotional, and psychological states, as an indicator of fatigue and sleepiness, particularly in the automobile and transportation industry, in visual tasking, and finally, as it relates to tear film stability and ocular surface health. The fact that it is highly variable and has input from so many sources makes it very difficult to study. In the present review, the behavior of blink in many of these systems is discussed, ultimately returning in each instance to a discussion of how these factors affect blink in the context of dry eyes. Blink is important to ocular surface health and to an individual's optimal functioning and quality of life. Disturbances in blink, as cause or effect, result in a breakdown of tear film stability, optical clarity, and visual function.
Collapse
Affiliation(s)
| | | | | | | | | | - Mark B Abelson
- a Ora, Inc , Andover , MA , USA.,b Department of Ophthalmology , Harvard Medical School , Boston , MA , USA
| |
Collapse
|
29
|
Gokul A, Wang MTM, Craig JP. Tear lipid supplement prophylaxis against dry eye in adverse environments. Cont Lens Anterior Eye 2017; 41:97-100. [PMID: 28943018 DOI: 10.1016/j.clae.2017.09.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Revised: 08/21/2017] [Accepted: 09/08/2017] [Indexed: 11/16/2022]
Abstract
PURPOSE To compare the prophylactic efficacy of single application of lipid and non-lipid containing tear supplements, prior to exposure of symptomatic dry eye subjects to a simulated adverse environment. METHODS Thirty subjects with mild-to-moderate dry eye symptoms participated in the prospective, randomised, double-masked, paired-eye trial. A lipomimetic drop (Systane® Balance) was applied to one eye (randomised), and a non-lipid containing drop (Systane® Ultra) applied simultaneously to the contralateral eye. Subjects were subsequently exposed to a validated simulated adverse environment model created by a standing fan directed towards the eye, at a distance of 1m, for 2.5min. Low contrast glare acuity, lipid layer grade (LLG), non-invasive tear break-up time (NIBUT), temperature variation factor (TVF), and tear meniscus height (TMH) were evaluated at baseline, following eye drop instillation and following simulated adverse environment exposure. RESULTS Both therapies resulted in increased NIBUT (both p<0.001), and prevented its decline below baseline with simulated adverse environment exposure (both p>0.05). However, only the lipomimetic drop increased LLG (p<0.001) and precluded its fall below baseline post-adverse environment exposure (p=0.15). Furthermore, post-instillation and post-exposure LLGs and NIBUT were significantly higher in the lipomimetic group (all p<0.05). No significant changes were observed in glare acuity, TVF and TMH (all p>0.05). More subjects (67%) reported greater ocular comfort in the eye receiving the lipomimetic. CONCLUSIONS Single application of both lipid and non-lipid containing eye drops conferred protective effects against exposure to adverse environmental conditions in subjects with mild-to-moderate dry eye, although the lipomimetic demonstrated superior prophylactic efficacy.
Collapse
Affiliation(s)
- Akilesh Gokul
- Department of Ophthalmology, New Zealand National Eye Centre, The University of Auckland, New Zealand
| | - Michael T M Wang
- Department of Ophthalmology, New Zealand National Eye Centre, The University of Auckland, New Zealand
| | - Jennifer P Craig
- Department of Ophthalmology, New Zealand National Eye Centre, The University of Auckland, New Zealand.
| |
Collapse
|
30
|
Cutolo CA, Barabino S, Bonzano C, Traverso CE. The Use of Topical Corticosteroids for Treatment of Dry Eye Syndrome. Ocul Immunol Inflamm 2017; 27:266-275. [DOI: 10.1080/09273948.2017.1341988] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
| | - Stefano Barabino
- Clinica Oculistica, Di.N.O.G.M.I, University of Genova, Genova, Italy
| | - Chiara Bonzano
- Clinica Oculistica, Di.N.O.G.M.I, University of Genova, Genova, Italy
| | - Carlo Enrico Traverso
- Clinica Oculistica, Di.N.O.G.M.I, University of Genova, Genova, Italy
- U.O.C. Clinica Oculistica, Ospedale Policlinico San Martino, Genova, Italy
| |
Collapse
|
31
|
Sullivan DA, Rocha EM, Aragona P, Clayton JA, Ding J, Golebiowski B, Hampel U, McDermott AM, Schaumberg DA, Srinivasan S, Versura P, Willcox MDP. TFOS DEWS II Sex, Gender, and Hormones Report. Ocul Surf 2017; 15:284-333. [PMID: 28736336 DOI: 10.1016/j.jtos.2017.04.001] [Citation(s) in RCA: 262] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 04/16/2017] [Indexed: 12/21/2022]
Abstract
One of the most compelling features of dry eye disease (DED) is that it occurs more frequently in women than men. In fact, the female sex is a significant risk factor for the development of DED. This sex-related difference in DED prevalence is attributed in large part to the effects of sex steroids (e.g. androgens, estrogens), hypothalamic-pituitary hormones, glucocorticoids, insulin, insulin-like growth factor 1 and thyroid hormones, as well as to the sex chromosome complement, sex-specific autosomal factors and epigenetics (e.g. microRNAs). In addition to sex, gender also appears to be a risk factor for DED. "Gender" and "sex" are words that are often used interchangeably, but they have distinct meanings. "Gender" refers to a person's self-representation as a man or woman, whereas "sex" distinguishes males and females based on their biological characteristics. Both gender and sex affect DED risk, presentation of the disease, immune responses, pain, care-seeking behaviors, service utilization, and myriad other facets of eye health. Overall, sex, gender and hormones play a major role in the regulation of ocular surface and adnexal tissues, and in the difference in DED prevalence between women and men. The purpose of this Subcommittee report is to review and critique the nature of this role, as well as to recommend areas for future research to advance our understanding of the interrelationships between sex, gender, hormones and DED.
Collapse
Affiliation(s)
- David A Sullivan
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA.
| | - Eduardo M Rocha
- Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Pasquale Aragona
- Department of Biomedical Sciences, Ocular Surface Diseases Unit, University of Messina, Messina, Sicily, Italy
| | - Janine A Clayton
- National Institutes of Health Office of Research on Women's Health, Bethesda, MD, USA
| | - Juan Ding
- Schepens Eye Research Institute, Massachusetts Eye & Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Blanka Golebiowski
- School of Optometry and Vision Science, University of New South Wales, Sydney, Australia
| | - Ulrike Hampel
- Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Alison M McDermott
- The Ocular Surface Institute, College of Optometry, University of Houston, Houston, TX, USA
| | - Debra A Schaumberg
- Harvard School of Public Health, Boston, MA, USA; University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Sruthi Srinivasan
- Centre for Contact Lens Research, School of Optometry, University of Waterloo, Ontario, Canada
| | - Piera Versura
- Department of Specialized, Experimental, and Diagnostic Medicine, University of Bologna, Bologna, Italy
| | - Mark D P Willcox
- School of Optometry and Vision Science, University of New South Wales, Sydney, Australia
| |
Collapse
|
32
|
Bron AJ, de Paiva CS, Chauhan SK, Bonini S, Gabison EE, Jain S, Knop E, Markoulli M, Ogawa Y, Perez V, Uchino Y, Yokoi N, Zoukhri D, Sullivan DA. TFOS DEWS II pathophysiology report. Ocul Surf 2017; 15:438-510. [PMID: 28736340 DOI: 10.1016/j.jtos.2017.05.011] [Citation(s) in RCA: 1113] [Impact Index Per Article: 139.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 05/26/2017] [Indexed: 12/18/2022]
Abstract
The TFOS DEWS II Pathophysiology Subcommittee reviewed the mechanisms involved in the initiation and perpetuation of dry eye disease. Its central mechanism is evaporative water loss leading to hyperosmolar tissue damage. Research in human disease and in animal models has shown that this, either directly or by inducing inflammation, causes a loss of both epithelial and goblet cells. The consequent decrease in surface wettability leads to early tear film breakup and amplifies hyperosmolarity via a Vicious Circle. Pain in dry eye is caused by tear hyperosmolarity, loss of lubrication, inflammatory mediators and neurosensory factors, while visual symptoms arise from tear and ocular surface irregularity. Increased friction targets damage to the lids and ocular surface, resulting in characteristic punctate epithelial keratitis, superior limbic keratoconjunctivitis, filamentary keratitis, lid parallel conjunctival folds, and lid wiper epitheliopathy. Hybrid dry eye disease, with features of both aqueous deficiency and increased evaporation, is common and efforts should be made to determine the relative contribution of each form to the total picture. To this end, practical methods are needed to measure tear evaporation in the clinic, and similarly, methods are needed to measure osmolarity at the tissue level across the ocular surface, to better determine the severity of dry eye. Areas for future research include the role of genetic mechanisms in non-Sjögren syndrome dry eye, the targeting of the terminal duct in meibomian gland disease and the influence of gaze dynamics and the closed eye state on tear stability and ocular surface inflammation.
Collapse
Affiliation(s)
- Anthony J Bron
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK; Vision and Eye Research Unit, Anglia Ruskin University, Cambridge, UK.
| | - Cintia S de Paiva
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX, USA
| | - Sunil K Chauhan
- Schepens Eye Research Institute & Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Stefano Bonini
- Department of Ophthalmology, University Campus Biomedico, Rome, Italy
| | - Eric E Gabison
- Department of Ophthalmology, Fondation Ophtalmologique Rothschild & Hôpital Bichat Claude Bernard, Paris, France
| | - Sandeep Jain
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Erich Knop
- Departments of Cell and Neurobiology and Ocular Surface Center Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Maria Markoulli
- School of Optometry and Vision Science, University of New South Wales, Sydney, Australia
| | - Yoko Ogawa
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Victor Perez
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami, Miami, FL, USA
| | - Yuichi Uchino
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Norihiko Yokoi
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Driss Zoukhri
- Tufts University School of Dental Medicine, Boston, MA, USA
| | - David A Sullivan
- Schepens Eye Research Institute & Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
33
|
Jones L, Downie LE, Korb D, Benitez-del-Castillo JM, Dana R, Deng SX, Dong PN, Geerling G, Hida RY, Liu Y, Seo KY, Tauber J, Wakamatsu TH, Xu J, Wolffsohn JS, Craig JP. TFOS DEWS II Management and Therapy Report. Ocul Surf 2017; 15:575-628. [DOI: 10.1016/j.jtos.2017.05.006] [Citation(s) in RCA: 578] [Impact Index Per Article: 72.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 05/03/2017] [Indexed: 02/06/2023]
|
34
|
Nassiri N, Rodriguez Torres Y, Meyer Z, Beyer MA, Vellaichamy G, Dhaliwal AS, Chungfat N, Hwang FS. Current and emerging therapy of dry eye disease. Part A: pharmacological modalities. EXPERT REVIEW OF OPHTHALMOLOGY 2017. [DOI: 10.1080/17469899.2017.1327350] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Nariman Nassiri
- Kresge Eye Institute - Department of Ophthalmology, School of Medicine, Wayne State University, Detroit, MI, USA
| | - Yasaira Rodriguez Torres
- Kresge Eye Institute - Department of Ophthalmology, School of Medicine, Wayne State University, Detroit, MI, USA
| | - Zachary Meyer
- Kresge Eye Institute - Department of Ophthalmology, School of Medicine, Wayne State University, Detroit, MI, USA
| | - Michael A. Beyer
- Kresge Eye Institute - Department of Ophthalmology, School of Medicine, Wayne State University, Detroit, MI, USA
| | - Gautham Vellaichamy
- Kresge Eye Institute - Department of Ophthalmology, School of Medicine, Wayne State University, Detroit, MI, USA
| | - Amar S. Dhaliwal
- Kresge Eye Institute - Department of Ophthalmology, School of Medicine, Wayne State University, Detroit, MI, USA
| | - Neil Chungfat
- Department of Ophthalmology, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Frank S. Hwang
- Kresge Eye Institute - Department of Ophthalmology, School of Medicine, Wayne State University, Detroit, MI, USA
| |
Collapse
|
35
|
Roy NS, Wei Y, Kuklinski E, Asbell PA. The Growing Need for Validated Biomarkers and Endpoints for Dry Eye Clinical Research. Invest Ophthalmol Vis Sci 2017; 58:BIO1-BIO19. [PMID: 28475698 PMCID: PMC5455411 DOI: 10.1167/iovs.17-21709] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 04/04/2017] [Indexed: 11/30/2022] Open
Abstract
Purpose Biomarkers with minimally invasive and reproducible objective metrics provide the key to future paradigm shifts in understanding of the underlying causes of dry eye disease (DED) and approaches to treatment of DED. We review biomarkers and their validity in providing objective metrics for DED clinical research and patient care. Methods The English-language literature in PubMed primarily over the last decade was surveyed for studies related to identification of biomarkers of DED: (1) inflammation, (2) point-of-care, (3) ocular imaging, and (4) genetics. Relevant studies in each group were individually evaluated for (1) methodological and analytical details, (2) data and concordance with other similar studies, and (3) potential to serve as validated biomarkers with objective metrics. Results Significant work has been done to identify biomarkers for DED clinical trials and for patient care. Interstudy variation among studies dealing with the same biomarker type was high. This could be attributed to biologic variations and/or differences in processing, and data analysis. Correlation with other signs and symptoms of DED was not always clear or present. Conclusions Many of the biomarkers reviewed show the potential to serve as validated and objective metrics for clinical research and patient care in DED. Interstudy variation for a given biomarker emphasizes the need for detailed reporting of study methodology, including information on subject characteristics, quality control, processing, and analysis methods to optimize development of nonsubjective metrics. Biomarker development offers a rich opportunity to significantly move forward clinical research and patient care in DED. Overview DED is an unmet medical need - a chronic pain syndrome associated with variable vision that affects quality of life, is common with advancing age, interferes with the comfortable use of contact lenses, and can diminish results of eye surgeries, such as cataract extraction, LASIK, and glaucoma procedures. It is a worldwide medical challenge with a prevalence rate ranging from 8% to 50%. Many clinicians and researchers across the globe are searching for better answers to understand the mechanisms related to the development and chronicity of DED. Though there have been many clinical trials for DED, few new treatments have emerged over the last decade. Biomarkers may provide the needed breakthrough to propel our understanding of DED to the next level and the potential to realize our goal of truly personalized medicine based on scientific evidence. Clinical trials and research on DED have suffered from the lack of validated biomarkers and less than objective and reproducible endpoints. Current work on biomarkers has provided the groundwork to move forward. This review highlights primarily ocular biomarkers that have been investigated for use in DED, discusses the methodologic outcomes in providing objective metrics for clinical research, and suggests recommendations for further work.
Collapse
Affiliation(s)
- Neeta S. Roy
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, New York, United States
| | - Yi Wei
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, New York, United States
| | - Eric Kuklinski
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, New York, United States
| | - Penny A. Asbell
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, New York, United States
| |
Collapse
|
36
|
Rodriguez-Agirretxe I, Garcia I, Soria J, Suarez TM, Acera A. Custom RT-qPCR-array for glaucoma filtering surgery prognosis. PLoS One 2017; 12:e0174559. [PMID: 28358901 PMCID: PMC5373565 DOI: 10.1371/journal.pone.0174559] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2016] [Accepted: 03/11/2017] [Indexed: 12/27/2022] Open
Abstract
Excessive subconjunctival scarring is the main reason of failure of glaucoma filtration surgery. We analyzed conjunctival and systemic gene expression patterns after non penetrating deep sclerectomy (NPDS). To find expression patterns related to surgical failure and their correlation with the clinical outcomes. This study consisted of two consecutive stages. The first was a prospective analysis of wound-healing gene expression profile of six patients after NPDS. Conjunctival samples and peripheral blood samples were collected before and 15, 90,180, and 360 days after surgery. In the second stage, we conducted a retrospective analysis correlating the late conjunctival gene expression and the outcome of the NPDS for 11 patients. We developed a RT-qPCR Array for 88 key genes associated to wound healing. RT-qPCR Array analysis of conjunctiva samples showed statistically significant differences in 29/88 genes in the early stages after surgery, 20/88 genes between 90 and 180 days after surgery, and only 2/88 genes one year after surgery. In the blood samples, the most important changes occurred in 12/88 genes in the first 15 days after surgery. Correspondence analyses (COA) revealed significant differences between the expression of 20/88 genes in patients with surgical success and failure one year after surgery. Different expression patterns of mediators of the bleb wound healing were identified. Examination of such patterns might be used in surgery prognosis. RT-qPCR Array provides a powerful tool for investigation of differential gene expression wound healing after glaucoma surgery.
Collapse
Affiliation(s)
- Iñaki Rodriguez-Agirretxe
- Instituto Clínico Quirúrgico de Oftalmología, Bilbao, Spain
- Hospital Universitario Donostia, San Sebastian, Spain
| | | | | | | | | |
Collapse
|
37
|
Calonge M, Pinto-Fraga J, González-García MJ, Enríquez-de-Salamanca A, López-de la Rosa A, Fernández I, López-Miguel A. Effects of the External Environment on Dry Eye Disease. Int Ophthalmol Clin 2017; 57:23-40. [PMID: 28282312 DOI: 10.1097/iio.0000000000000168] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
|
38
|
Coursey TG, Tukler Henriksson J, Barbosa FL, de Paiva CS, Pflugfelder SC. Interferon-γ-Induced Unfolded Protein Response in Conjunctival Goblet Cells as a Cause of Mucin Deficiency in Sjögren Syndrome. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:1547-58. [PMID: 27085137 DOI: 10.1016/j.ajpath.2016.02.004] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 01/19/2016] [Accepted: 02/01/2016] [Indexed: 12/21/2022]
Abstract
Goblet cells (GCs) are specialized secretory cells that produce mucins and a variety of other proteins. Significant conjunctival GC loss occurs in both experimental dry eye models and patients with keratoconjunctivitis sicca due to the induction of interferon (IFN)-γ. With the use of a primary murine culture model, we found that GCs are highly sensitive to IFN-γ with significantly reduced proliferation and altered structure with low concentrations. GC cultures treated with IFN-γ have increased gene expression of Muc2 and Muc5AC but do not express these mucin glycoproteins. We hypothesized that IFN-γ induces endoplasmic reticulum stress and the unfolded protein response (UPR) in GCs. Cultures treated with IFN-γ increased expression of UPR-associated genes and proteins. Increased GRP78 and sXBP1 expression was found in experimental dry eye and Sjögren syndrome models and was GC specific. Increased GRP78 was also found in the conjunctiva of patients with Sjögren syndrome at the gene and protein levels. Treatment with dexamethasone inhibited expression of UPR-associated genes and increased mucin production. These results indicate that induction of UPR by IFN-γ is an important cause of GC-associated mucin deficiency observed in aqueous-deficient dry eye. Therapies to block the effects of IFN-γ on the metabolically active endoplasmic reticulum in these cells might enhance synthesis and secretion of the protective GC mucins on the ocular surface.
Collapse
Affiliation(s)
- Terry G Coursey
- Ocular Surface Center, Department of Ophthalmology, Cullen Eye Institute, Baylor College of Medicine, Houston, Texas
| | - Johanna Tukler Henriksson
- Ocular Surface Center, Department of Ophthalmology, Cullen Eye Institute, Baylor College of Medicine, Houston, Texas
| | - Flavia L Barbosa
- Ocular Surface Center, Department of Ophthalmology, Cullen Eye Institute, Baylor College of Medicine, Houston, Texas
| | - Cintia S de Paiva
- Ocular Surface Center, Department of Ophthalmology, Cullen Eye Institute, Baylor College of Medicine, Houston, Texas
| | - Stephen C Pflugfelder
- Ocular Surface Center, Department of Ophthalmology, Cullen Eye Institute, Baylor College of Medicine, Houston, Texas.
| |
Collapse
|
39
|
Pflugfelder SC, De Paiva CS, Moore QL, Volpe EA, Li DQ, Gumus K, Zaheer ML, Corrales RM. Aqueous Tear Deficiency Increases Conjunctival Interferon-γ (IFN-γ) Expression and Goblet Cell Loss. Invest Ophthalmol Vis Sci 2016; 56:7545-50. [PMID: 26618646 DOI: 10.1167/iovs.15-17627] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE To investigate the hypothesis that increased interferon-γ (IFN-γ) expression is associated with conjunctival goblet cell loss in subjects with tear dysfunction. METHODS Goblet cell density (GCD) was measured in impression cytology from the temporal bulbar conjunctiva, and gene expression was measured in cytology samples from the nasal bulbar conjunctiva obtained from 68 subjects, including normal control, meibomian gland disease (MGD), non-Sjögren syndrome (non-SSATD)-, and Sjögren syndrome (SSATD)-associated aqueous tear deficiency. Gene expression was evaluated by real-time PCR. Tear meniscus height (TMH) was measured by optical coherence tomography. Fluorescein and lissamine green dye staining evaluated corneal and conjunctival disease, respectively. Between-group mean differences and correlation coefficients were calculated. RESULTS Compared to control, IFN-γ expression was significantly higher in both ATD groups, and its receptor was higher in SSATD. Expression of IL-13 and its receptor was similar in all groups. Goblet cell density was lower in the SSATD group; expression of MUC5AC mucin was lower and cornified envelope precursor small proline-rich region (SPRR)-2G higher in both ATD groups. Interferon-γ transcript number was inversely correlated with GCD (r = -0.37, P < 0.04) and TMH (r = -0.37, P = 0.02), and directly correlated with lissamine green staining (r = 0.51, P < 0.001) and SPRR-2G expression (r = 0.32, P < 0.05). CONCLUSIONS Interferon-γ expression in the conjunctiva was higher in aqueous deficiency and correlated with goblet cell loss and severity of conjunctival disease. These results support findings of animal and culture studies showing that IFN-γ reduces conjunctival goblet cell number and mucin production.
Collapse
Affiliation(s)
- Stephen C Pflugfelder
- Ocular Surface Center Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, United States
| | - Cintia S De Paiva
- Ocular Surface Center Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, United States
| | - Quianta L Moore
- Ocular Surface Center Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, United States
| | - Eugene A Volpe
- Ocular Surface Center Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, United States
| | - De-Quan Li
- Ocular Surface Center Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, United States
| | - Koray Gumus
- Ocular Surface Center Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, United States 2Department of Ophthalmology, Erciyes University School of Medicine, Kayseri, Turkey
| | - Mahira L Zaheer
- Ocular Surface Center Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, United States
| | - Rosa M Corrales
- Ocular Surface Center Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, United States
| |
Collapse
|
40
|
La Porta Weber S, Becco de Souza R, Gomes JÁP, Hofling-Lima AL. The Use of the Esclera Scleral Contact Lens in the Treatment of Moderate to Severe Dry Eye Disease. Am J Ophthalmol 2016; 163:167-173.e1. [PMID: 26701271 DOI: 10.1016/j.ajo.2015.11.034] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2015] [Revised: 11/25/2015] [Accepted: 11/25/2015] [Indexed: 11/29/2022]
Abstract
PURPOSE To evaluate the efficacy of the Esclera scleral contact lens (SCL) treatment and its impact on clinical testing for moderate to severe dry eye disease (DED). DESIGN Prospective interventional case series. METHODS A total of 41 eyes from 25 patients with moderate to severe DED were evaluated for the Esclera SCL treatment. Best-corrected visual acuity (BCVA), tear osmolarity, the Schirmer I test, tear film breakup time (TBUT), corneal and conjunctival staining, meibomian grading, and Ocular Surface Disease Index and SF-36v2 questionnaires were assessed before and after the SCL treatment. These values were compared to assess the real benefit of using SCL as a treatment for DED. RESULTS Forty-one eyes from 25 patients were fitted with SCL for management of DED. The underlying diseases were Stevens-Johnson syndrome (22 eyes), Sjogren syndrome (11 eyes), graft-vs-host disease (2 eyes), dry eye after keratomileusis in situ (2 eyes), and undifferentiated ocular surface disease (4 eyes). BCVA improved from 0.703 ± 0.55 logMAR with habitual correction to 0.406 ± 0.43 logMAR with SCL (P < .001). There was a significant decrease in tear osmolarity values (338.1 ± 27.1 to 314.25 ± 38.8 mOsm/L, P < .001) and van Bijsterveld scores (3.63 ± 2.33 to 2.63 ± 2.46 grade, P = .015) between the baseline and 12 months after SCL wear. There were also significant improvements in dry eye symptoms and quality of life as assessed by the OSDI and SF-36v2 questionnaires (both with P < .001). CONCLUSIONS The Esclera SCL treatment had a positive impact on tear osmolarity and van Bijsterveld score, as well as an improvement in the patients' BCVA, dry eye symptoms, and quality of life.
Collapse
Affiliation(s)
- Sarah La Porta Weber
- Department of Ophthalmology, Ocular Surface Advanced Center, Federal University of Sao Paulo, Sao Paulo, Brazil.
| | - Rodrigo Becco de Souza
- Department of Ophthalmology, Ocular Surface Advanced Center, Federal University of Sao Paulo, Sao Paulo, Brazil
| | - José Álvaro Pereira Gomes
- Department of Ophthalmology, Ocular Surface Advanced Center, Federal University of Sao Paulo, Sao Paulo, Brazil
| | - Ana Luisa Hofling-Lima
- Department of Ophthalmology, Ocular Surface Advanced Center, Federal University of Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
41
|
Abstract
PURPOSE OF REVIEW Sjögren's syndrome affects exocrine glands leading to a dry mouth and dry eyes. Dry eye manifestations can precede the diagnosis of Sjögren's syndrome by many years. Innumerous spontaneous and inducible Sjögren's syndrome models have been used to study the pathogenesis of Sjögren's syndrome. This review focuses on recent human data, ocular and extraglandular manifestations of animal models, what is known, what is still unknown and how we need to look, and their correlation correspondence to human disease. RECENT FINDINGS Hallmarks of dry eye in Sjögren's syndrome include increased corneal staining, goblet cell loss and low tear volume. Confocal microscopy and impression cytology are able to clarify new markers of the ocular disease. Extraglandular manifestations should be an alert more severe complications in the eye. Some models have strong sex and exocrine gland predilection, whereas aging generally worsens the disease phenotype. Although most models do not display a significant increase in corneal staining or tear secretion impairment, conjunctival infiltration and decrease in goblet cells are frequently seen. SUMMARY We have seen great advances in the role of inflammation in ocular, oral and extra-glandular manifestations of Sjögren's syndrome. Several mechanisms and mediators of Sjögren's syndrome have been elucidated in animal model studies.
Collapse
|
42
|
Clinical and Molecular Inflammatory Response in Sjögren Syndrome-Associated Dry Eye Patients Under Desiccating Stress. Am J Ophthalmol 2016; 161:133-41.e1-2. [PMID: 26456254 DOI: 10.1016/j.ajo.2015.09.039] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 09/30/2015] [Accepted: 09/30/2015] [Indexed: 11/20/2022]
Abstract
PURPOSE To evaluate the response of the lacrimal function unit in Sjögren syndrome (SS)-associated dry eye patients exposed to 2 simulated daily life environmental conditions. DESIGN Prospective crossover pilot study. METHODS Fourteen female SS dry eye patients were exposed for 2 hours to a controlled normal condition (23 C, 45% relative humidity, and air flow 0.10 m/s) and a controlled adverse condition that simulates desiccating stress (23 C, 5% relative humidity, and air flow 0.10 m/s). The following dry eye tests were performed before and after the exposure: tear osmolarity, phenol red thread test, conjunctival hyperemia, fluorescein tear break-up time, corneal fluorescein staining, conjunctival lissamine green staining, and Schirmer test. Levels of 16 molecules were analyzed in tears by multiplex immunobead analysis. RESULTS Clinical evaluation showed lacrimal functional unit impairment after the desiccating stress: significantly increased tear osmolarity (315.7 ± 3.0 vs 327.7 ± 5.1 mOsm/L, P = .03), conjunctival hyperemia (1.3 ± 0.1 vs 1.6 ± 0.1, P = .05), and corneal staining in temporal (3.5 ± 0.5 vs 4.7 ± 0.4, P = .01) and nasal (3.6 ± 0.5 vs 4.5 ± 0.5, P = .04) areas. Tear concentrations increased for interleukin-1 receptor antagonist (16 557.1 ± 4047.8 vs 31 895.3 ± 5916.5 pg/mL, P = .01), interleukin-6 (63.8 ± 20.2 vs 111.5 ± 29.6 pg/mL, P = .02), interleukin-8 (2196.1 ± 737.9 vs 3753.2 ± 1106.0 pg/mL, P = .03), and matrix metalloproteinase-9 (101 515.6 ± 37 088.4 vs 145 867.1 ± 41 651.5 pg/mL, P = .03). After the simulated normal condition, only a significant increase in nasal corneal staining (2.9 ± 0.5 vs 3.6 ± 0.5, P = .03) was observed. CONCLUSIONS Even a short exposure to a desiccating environment can produce a significant deterioration of the lacrimal function unit in female SS dry eye patients. The often unnoticed exposure to these conditions during daily life may increase inflammatory activity rapidly, triggering an ocular surface deterioration.
Collapse
|
43
|
Pinto-Fraga J, López-Miguel A, González-García MJ, Fernández I, López-de-la-Rosa A, Enríquez-de-Salamanca A, Stern ME, Calonge M. Topical Fluorometholone Protects the Ocular Surface of Dry Eye Patients from Desiccating Stress: A Randomized Controlled Clinical Trial. Ophthalmology 2015; 123:141-53. [PMID: 26520171 DOI: 10.1016/j.ophtha.2015.09.029] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 09/18/2015] [Accepted: 09/18/2015] [Indexed: 12/18/2022] Open
Abstract
PURPOSE To assess the efficacy of topical 0.1% fluorometholone in dry eye disease (DED) patients for ameliorating the worsening of the ocular surface when exposed to adverse environments. DESIGN Single-center, double-masked, randomized, vehicle-controlled clinical trial. PARTICIPANTS Forty-one patients showing moderate to severe DED. METHODS Patients randomly received 1 drop 4 times daily of either topical 0.1% fluorometholone (FML group) or topical polyvinyl alcohol (PA group) for 22 days. Corneal and conjunctival staining, conjunctival hyperemia, tear film breakup time (TBUT), tear osmolarity, and the Symptom Assessment in Dry Eye (SANDE) questionnaire scores were determined at baseline. Variables were reassessed on day 21 before and after undergoing a 2-hour controlled adverse environment exposure and again on day 22. MAIN OUTCOMES MEASURES Percentage of patients showing an increase 1 point or more in corneal staining and a reduction of 2 points or more (0-10 scale) in SANDE score, after the controlled adverse environment exposure and 24 hours later. RESULTS After 21 days of treatment, the FML group showed greater improvements in corneal and conjunctival staining, hyperemia, and TBUT than the PA group (P≤0.03). After the adverse exposure, the percentage of patients having a 1-grade or more increase in corneal staining was significantly (P = 0.03) higher in the PA group (63.1% vs. 23.8%, respectively). Additionally, the FML group showed no significant changes in corneal staining (mean, 0.86; 95% confidence interval [CI], 0.47-1.25; vs. mean, 1.05; 95% CI, 0.59-1.51, for visit 2 and 3, respectively), conjunctival staining (mean, 0.95; 95% CI, 0.54-1.37 vs. mean, 1.19; 95% CI, 0.75-1.63), and hyperemia (mean, 0.71; 95% CI, 0.41-1.02 vs. 1.14; 95% CI, 0.71-1.58) after the exposure, whereas for the PA group, there was significant worsening (P≤0.009) in these variables (corneal staining: mean, 1.95; 95% CI, 1.57-2.33 vs. mean, 2.58; 95% CI, 2.17-2.98; conjunctival staining: mean, 1.68; 95% CI, 1.29-2.08 vs. mean, 2.47; 95% CI, 2.07-2.88; hyperemia: mean, 1.95; 95% CI, 1.63-2.26 vs. mean, 2.84; 95% CI, 2.62-3.07). CONCLUSIONS Three-week topical 0.1% fluorometholone therapy is effective not only in reducing ocular surface signs in DED patients, but also especially in preventing exacerbation caused by exposure to a desiccating stress.
Collapse
Affiliation(s)
- José Pinto-Fraga
- Instituto Universitario de Oftalmobiología Aplicada, University of Valladolid, Valladolid, Spain; Biomedical Research Networking Center on Bioengineering, Biomaterials and Nanomedicine, Zaragoza, Spain
| | - Alberto López-Miguel
- Instituto Universitario de Oftalmobiología Aplicada, University of Valladolid, Valladolid, Spain; VISIÓN I+D, SL, Valladolid, Spain
| | - María J González-García
- Instituto Universitario de Oftalmobiología Aplicada, University of Valladolid, Valladolid, Spain; Biomedical Research Networking Center on Bioengineering, Biomaterials and Nanomedicine, Zaragoza, Spain
| | - Itziar Fernández
- Instituto Universitario de Oftalmobiología Aplicada, University of Valladolid, Valladolid, Spain; Biomedical Research Networking Center on Bioengineering, Biomaterials and Nanomedicine, Zaragoza, Spain
| | - Alberto López-de-la-Rosa
- Instituto Universitario de Oftalmobiología Aplicada, University of Valladolid, Valladolid, Spain
| | - Amalia Enríquez-de-Salamanca
- Instituto Universitario de Oftalmobiología Aplicada, University of Valladolid, Valladolid, Spain; Biomedical Research Networking Center on Bioengineering, Biomaterials and Nanomedicine, Zaragoza, Spain
| | | | - Margarita Calonge
- Instituto Universitario de Oftalmobiología Aplicada, University of Valladolid, Valladolid, Spain; Biomedical Research Networking Center on Bioengineering, Biomaterials and Nanomedicine, Zaragoza, Spain.
| |
Collapse
|