Conjunctival HLA-DR Expression and Its Association With Symptoms and Signs in the DREAM Study

Neeta S Roy, Yi Wei, Yinxi Yu, Gui-Shuang Ying, Eric Kuklinski, Brendan Barry, Maureen G Maguire, Reza Dana, Mary Brightwell-Arnold, Penny A Asbell, for the Dry Eye Assessment and Management (DREAM) Study Group, Neeta S Roy, Yi Wei, Yinxi Yu, Gui-Shuang Ying, Eric Kuklinski, Brendan Barry, Maureen G Maguire, Reza Dana, Mary Brightwell-Arnold, Penny A Asbell, for the Dry Eye Assessment and Management (DREAM) Study Group

Abstract

Purpose: Evaluation of dry eye disease (DED) relies on subjective symptoms and signs. We examined HLA-DR expression (HLA-DR%) in conjunctival cells, a minimally invasive biomarker with objective metrics, as an alternative method.

Methods: Dry Eye Assessment and Management (DREAM) study participants completed the Ocular Surface Disease Index questionnaire. Clinicians evaluated tear volume, tear breakup time, and corneal and conjunctival staining. Conjunctival impression cytology samples (n = 1049) were assessed for HLA-DR% in total cells (TCs), epithelial cells (ECs), and white blood cells (WBCs). Associations (categorized into <5%, 5%-15%, >15%-25%, and >25%) with symptoms and signs were evaluated.

Results: The HLA-DR% varied markedly across samples. Over 40% had <5 HLA-DR% positive cells in TCs and ECs and under 23% in WBCs. Higher HLA-DR% was associated with higher conjunctival staining for ECs (mean score 2.77 for <5% and 3.28 for >25%, linear trend P = 0.009) and TCs (mean score 2.82 for <5% and 3.29 for >25%, linear trend P = 0.04) and in TCs was associated with higher corneal staining (mean score 3.59 for <5% and 4.46 for >25%, linear trend P = 0.03). HLA-DR% in WBCs did not correlated with signs (all P ≥ 0.58), and in TCs, ECs or WBCs were not associated with symptoms (P > 0.06).

Conclusions: The distribution of HLA-DR% in conjunctival cells reflects the heterogeneity of disease in DREAM participants. High percentages of samples with <5% positive cells indicate that HLA-DR% may not be a sensitive marker for DED in all patients.

Translational relevance: High HLA-DR% in ECs in association with high conjunctival staining may identify a subgroup of DED patients prone to epithelial disease and possibly need a different approach from current standards of treatment.

Keywords: HLA-DR; conjunctival cells; correlation; dry eye disease; symptoms and sign.

Figures

Figure 1
Figure 1
Flow cytometer analysis. Dot plots of a representative sample from the study. Total analyzable cells (88.61%) (B) were gated out from a scatter plot of FCS-W (Forward Scatter-Width) versus SSC-A (Side Scatter-Area; A). Sequential gating of EpCAM- or CD45-expressing cells from the total population yielded 90.20% ECs (C) and 1.21% WBCs (F). EpCAM+/HLA-DR+ cells (D) comprise 16.52% of the EC population and CD45+/HLA-DR+ cells (G) comprise 71.07% of the total WBCs.
Figure 2
Figure 2
Distribution of HLA-DR in patient conjunctival cells. (A, B, and C) Show the percentage sample distribution (y axis) of HLA-DR% (x axis) in TCs (TCs), EpCAM-expressing ECs, and CD45-expressing WBCs, respectively, in the 1049 eye samples analyzed. (D) Stacked bars showing the percent distribution of samples in the four levels of HLA-DR% (15%–25%, and >25%).

References

    1. Chao W, Belmonte C, Benitez Del Castillo JM, et al. Report of the inaugural meeting of the TFOS i(2) = initiating innovation series: targeting the unmet need for dry eye treatment. Ocul Surf. 2016;14:264–316.
    1. Craig JP, Nichols KK, Akpek EK, et al. TFOS DEWS II definition and classification report. Ocul Surf. 2017;15:276–283.
    1. Wolffsohn JS, Arita R, Chalmers R, et al. TFOS DEWS II diagnostic methodology report. Ocul Surf. 2017;15:539–574.
    1. Zeev MS, Miller DD, Latkany R. Diagnosis of dry eye disease and emerging technologies. Clin Ophthalmol. 2014;8:581–590.
    1. Bartlett JD, Keith MS, Sudharshan L, Snedecor SJ. Associations between signs and symptoms of dry eye disease: a systematic review. Clin Ophthalmol. 2015;9:1719–1730.
    1. Baudouin C, Aragona P, Van Setten G, et al. Diagnosing the severity of dry eye: a clear and practical algorithm. Br J Ophthalmol. 2014;98:1168–1176.
    1. Sullivan BD, Crews LA, Messmer EM, et al. Correlations between commonly used objective signs and symptoms for the diagnosis of dry eye disease: clinical implications. Acta Ophthalmol. 2014;92:161–166.
    1. Vehof J, Sillevis Smitt-Kamminga N, Nibourg SA, Hammond CJ. Predictors of discordance between symptoms and signs in dry eye disease. Ophthalmology. 2017;124:280–286.
    1. Ambroziak AM, Szaflik J, Szaflik JP, Ambroziak M, Witkiewicz J, Skopinski P. Immunomodulation on the ocular surface: a review. Cent Eur J Immunol. 2016;41:195–208.
    1. Bose T, Diedrichs-Mohring M, Wildner G. Dry eye disease and uveitis: a closer look at immune mechanisms in animal models of two ocular autoimmune diseases. Autoimmun Rev. 2016;15:1181–1192.
    1. Pflugfelder SC, Stern M, Zhang S, Shojaei A. LFA-1/ICAM-1 interaction as a therapeutic target in dry eye disease. J Ocul Pharmacol Ther. 2017;33:5–12.
    1. Stevenson W, Chauhan SK, Dana R. Dry eye disease: an immune-mediated ocular surface disorder. Arch Ophthalmol. 2012;130:90–100.
    1. Wei Y, Asbell PA. The core mechanism of dry eye disease is inflammation. Eye Contact Lens. 2014;40:248–256.
    1. Chen W, Cao H, Lin J, Olsen N, Zheng SG. Biomarkers for primary Sjogren's syndrome. Genomics Proteomics Bioinformatics. 2015;13:219–223.
    1. Hagan S. Biomarkers of ocular surface disease using impression cytology. Biomark Med. 2017;11:1135–1147.
    1. Hagan S, Martin E, Enriquez-de-Salamanca A. Tear fluid biomarkers in ocular and systemic disease: potential use for predictive, preventive and personalised medicine. EPMA J. 2016;7:15.
    1. 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.
    1. Abud TB, Amparo F, Saboo US, et al. A clinical trial comparing the safety and efficacy of topical tacrolimus versus methylprednisolone in ocular graft-versus-host disease. Ophthalmology. 2016;123:1449–1457.
    1. Barabino S, Montaldo E, Solignani F, Valente C, Mingari MC, Rolando M. Immune response in the conjunctival epithelium of patients with dry eye. Exp Eye Res. 2010;91:524–529.
    1. Brignole F, Pisella PJ, De Saint Jean M, Goldschild M, Goguel A, Baudouin C. Flow cytometric analysis of inflammatory markers in KCS: 6-month treatment with topical cyclosporin A. Invest Ophthalmol Vis Sci. 2001;42:90–95.
    1. Brignole F, Pisella PJ, Goldschild M, De Saint Jean M, Goguel A, Baudouin C. Flow cytometric analysis of inflammatory markers in conjunctival epithelial cells of patients with dry eyes. Invest Ophthalmol Vis Sci. 2000;41:1356–1363.
    1. Brignole-Baudouin F, Baudouin C, Aragona P, et al. A multicentre, double-masked, randomized, controlled trial assessing the effect of oral supplementation of omega-3 and omega-6 fatty acids on a conjunctival inflammatory marker in dry eye patients. Acta Ophthalmol. 2011;89:e591–e597.
    1. Brignole-Baudouin F, Riancho L, Ismail D, Deniaud M, Amrane M, Baudouin C. Correlation between the inflammatory marker HLA-DR and signs and symptoms in moderate to severe dry eye disease. Invest Ophthalmol Vis Sci. 2017;58:2438–2448.
    1. Buggage R, Amrane M, Ismail D, Lemp M, Baudouin C, Baudouin F. The correlation between ocular surface inflammation and corneal fluorescein staining (CFS) in patients with moderate to severe dry eye disease (DED) participating in a randomized clinical trial. Acta Ophthalmologica. 2011;89:4172.
    1. Fernandez KB, Epstein SP, Raynor GS, et al. Modulation of HLA-DR in dry eye patients following 30 days of treatment with a lubricant eyedrop solution. Clin Ophthalmol. 2015;9:1137–1145.
    1. Leonardi A, Van Setten G, Amrane M, et al. Efficacy and safety of 0.1% cyclosporine A cationic emulsion in the treatment of severe dry eye disease: a multicenter randomized trial. Eur J Ophthalmol. 2016;26:287–296.
    1. Liu X, Wang S, Kao AA, Long Q. The effect of topical pranoprofen 0.1% on the clinical evaluation and conjunctival HLA-DR expression in dry eyes. Cornea. 2012;31:1235–1239.
    1. Mrugacz M, Zak J, Bakunowicz-Lazarczyk A, Wysocka J, Minarowska A. Flow cytometric analysis of HLA-DR antigen in conjunctival epithelial cells of patients with cystic fibrosis. Eye (Lond) 2007;21:1062–1066.
    1. Pflugfelder SC, Bian F, Gumus K, Farley W, Stern ME, De Paiva CS. Severity of Sjogren's syndrome keratoconjunctivitis sicca increases with increased percentage of conjunctival antigen-presenting cells. Int J Mol Sci. 2018;19:E2760.
    1. Pisella PJ, Brignole F, Debbasch C, et al. Flow cytometric analysis of conjunctival epithelium in ocular rosacea and keratoconjunctivitis sicca. Ophthalmology. 2000;107:1841–1849.
    1. Rolando M, Barabino S, Mingari C, Moretti S, Giuffrida S, Calabria G. Distribution of conjunctival HLA-DR expression and the pathogenesis of damage in early dry eyes. Cornea. 2005;24:951–954.
    1. Sanchez MA, Torralbo-Jimenez P, Giron N, et al. Comparative analysis of carmellose 0.5% versus hyaluronate 0.15% in dry eye: a flow cytometric study. Cornea. 2010;29:167–171.
    1. Trinh L, Brignole-Baudouin F, Raphael M, et al. Th1 and Th2 responses on the ocular surface in uveitis identified by CCR4 and CCR5 conjunctival expression. Am J Ophthalmol. 2007;144:580–585.
    1. Versura P, Profazio V, Schiavi C, Campos EC. Hyperosmolar stress upregulates HLA-DR expression in human conjunctival epithelium in dry eye patients and in vitro models. Invest Ophthalmol Vis Sci. 2011;52:5488–5496.
    1. Roche PA, Furuta K. The ins and outs of MHC class II-mediated antigen processing and presentation. Nat Rev Immunol. 2015;15:203–216.
    1. De Saint Jean M, Brignole F, Feldmann G, Goguel A, Baudouin C. Interferon-gamma induces apoptosis and expression of inflammation-related proteins in Chang conjunctival cells. Invest Ophthalmol Vis Sci. 1999;40:2199–2212.
    1. Kambayashi T, Laufer TM. Atypical MHC class II-expressing antigen-presenting cells: can anything replace a dendritic cell? Nat Rev Immunol. 2014;14:719–730.
    1. Baudouin C, Brignole F, Becquet F, Pisella PJ, Goguel A. Flow cytometry in impression cytology specimens. A new method for evaluation of conjunctival inflammation. Invest Ophthalmol Vis Sci. 1997;38:1458–1464.
    1. Epstein SP, Gadaria-Rathod N, Wei Y, Maguire MG, Asbell PA. HLA-DR expression as a biomarker of inflammation for multicenter clinical trials of ocular surface disease. Exp Eye Res. 2013;111:95–104.
    1. Berns B, Demolis P, Scheulen M. How can biomarkers become surrogate endpoints? Eur J Cancer. 2007;5:37–40.
    1. Katz R. Biomarkers and surrogate markers: an FDA perspective. NeuroRx. 2004;1:189–195.
    1. Dry Eye Assessment and Management Study Research Group, Asbell PA, Maguire MG, et al. n-3 fatty acid supplementation for the treatment of dry eye disease. N Engl J Med. 2018;378:1681–1690.
    1. Schiffman RM, Christianson MD, Jacobsen G, Hirsch JD, Reis BL. Reliability and validity of the Ocular Surface Disease Index. Arch Ophthalmol. 2000;118:615–621.
    1. Asbell PA, Maguire MG, Peskin E, et al. Dry Eye Assessment and Management (DREAM(c)) Study: Study design and baseline characteristics. Contemp Clin Trials. 2018;71:70–79.
    1. Lemp MA. Report of the National Eye Institute/Industry workshop on clinical trials in dry eyes. CLAO J. 1995;21:221–232.
    1. Huang JF, Yafawi R, Zhang M, et al. Immunomodulatory effect of the topical ophthalmic Janus kinase inhibitor tofacitinib (CP-690, 550) in patients with dry eye disease. Ophthalmology. 2012;119:e43–e50.
    1. Yafawi R, Ko M, Sace FP, John-Baptiste A. Limitations of an ocular surface inflammatory biomarker in impression cytology specimens. Cutan Ocul Toxicol. 2013;32:46–53.
    1. Danova M, Torchio M, Comolli G, Sbrana A, Antonuzzo A, Mazzini G. The role of automated cytometry in the new era of cancer immunotherapy. Mol Clin Oncol. 2018;9:355–361.
    1. Richards AJ, Staats J, Enzor J, et al. Setting objective thresholds for rare event detection in flow cytometry. J Immunol Methods. 2014;409:54–61.
    1. Finak G, Langweiler M, Jaimes M, et al. Standardizing flow cytometry immunophenotyping analysis from the Human ImmunoPhenotyping Consortium. Sci Rep. 2016;6:20686.
    1. Kalina T, Brdickova N, Glier H, et al. Frequent issues and lessons learned from EuroFlow QA. J Immunol Methods. 2018. pp. S0022–1759.–30140.
    1. Ferrari G, Rabiolo A, Bignami F, et al. Quantifying ocular surface inflammation and correlating it with inflammatory cell infiltration in vivo: a novel method. Invest Ophthalmol Vis Sci. 2015;56:7067–7075.
    1. Li N, Deng XG, He MF. Comparison of the Schirmer I test with and without topical anesthesia for diagnosing dry eye. Int J Ophthalmol. 2012;5:478–481.
    1. Uchiyama E, Aronowicz JD, Butovich IA, McCulley JP. Pattern of vital staining and its correlation with aqueous tear deficiency and meibomian gland dropout. Eye Contact Lens. 2007;33:177–179.
    1. Hernandez-Molina G, Leal-Alegre G, Michel-Peregrina M. The meaning of anti-Ro and anti-La antibodies in primary Sjogren's syndrome. Autoimmun Rev. 2011;10:123–125.
    1. Hernandez-Molina G, Vargas-Alarcon G, Rodriguez-Perez JM, Martinez-Rodriguez N, Lima G, Sanchez-Guerrero J. High-resolution HLA analysis of primary and secondary Sjogren's syndrome: a common immunogenetic background in Mexican patients. Rheumatol Int. 2015;35:643–649.
    1. Loiseau P, Lepage V, Djelal F, et al. HLA class I and class II are both associated with the genetic predisposition to primary Sjogren syndrome. Hum Immunol. 2001;62:725–731.

Source: PubMed

3
Subskrybuj