Chloroquine Protects Human Corneal Epithelial Cells from Desiccation Stress Induced Inflammation without Altering the Autophagy Flux

Shivapriya Shivakumar, Trailokyanath Panigrahi, Rohit Shetty, Murali Subramani, Arkasubhra Ghosh, Nallathambi Jeyabalan, Shivapriya Shivakumar, Trailokyanath Panigrahi, Rohit Shetty, Murali Subramani, Arkasubhra Ghosh, Nallathambi Jeyabalan

Abstract

Dry eye disease (DED) is a multifactorial ocular surface disorder affecting millions of individuals worldwide. Inflammation has been associated with dry eye and anti-inflammatory drugs are now being targeted as the alternate therapeutic approach for dry eye condition. In this study, we have explored the anti-inflammatory and autophagy modulating effect of chloroquine (CQ) in human corneal epithelial and human corneal fibroblasts cells exposed to desiccation stress, (an in-vitro model for DED). Gene and protein expression profiling of inflammatory and autophagy related molecular factors were analyzed in HCE-T and primary HCF cells exposed to desiccation stress with and without CQ treatment. HCE-T and HCF cells exposed to desiccation stress exhibited increased levels of activated p65, TNF-α, MCP-1, MMP-9, and IL-6. Further, treatment with CQ decreased the levels of active p65, TNF-α, MCP-1, and MMP-9 in cells underdesiccation stress. Increased levels of LC3B and LAMP1 markers in HCE-T cells exposed to desiccation stress suggest activation of autophagy and the addition of CQ did not alter these levels. Changes in the phosphorylation levels of MAPKinase and mTOR pathway proteins were found in HCE-T cells under desiccation stress with or without CQ treatment. Taken together, the data suggests that HCE-T cells under desiccation stress showed NFκB mediated inflammation, which was rescued through the anti-inflammatory effect of CQ without altering the autophagy flux. Therefore, CQ may be used as an alternate therapeutic management for dry eye condition.

Figures

Figure 1
Figure 1
Morphological and viability analysis of HCE-T, Primary HCF, and HCE cells exposed to desiccation stress. (a) Bright field images of HCE-T cells exposed to desiccation stress, with and without chloroquine (CQ) /cyclosporine (CsA) treatment under the 10X. ((A) Non-desiccated/control HCE-T cells, (B) desiccated HCE-T cells, (C) desiccated cells treated with CQ, and (D) desiccated cells treated with CsA) (Figure 1(a)). (b) Percentage of cell viability of HCE-T cells exposed to desiccation cells with and without (CQ/CsA) treatment (Figure 1(b)). (c) Bright field images of primary HCF cells exposed to desiccation stress, with and without chloroquine (CQ)/cyclosporine (CsA) treatment under the 10X objective. ((A) Non-desiccated/control HCF cells, (B) desiccated HCF cells, (C) HCF cells treated with CQ, (D) desiccated HCF cells treated with CQ, (E) HCF cells treated with CsA, and (F) desiccated cells treated with CsA) (Figure 1(c)). (d) Percentage of cell viability of HCF cells exposed to desiccation cells with and without (CQ/CsA) treatment (Figure 1(d)). (e) Bright field images of primary HCE cells exposed to desiccation stress, with and without chloroquine (CQ)/cyclosporine (CsA) treatment under the 10X objective. ((A) Non-desiccated/control primary HCE cells, (B) desiccated primary HCE cells, (C) primary HCE cells treated with CQ, (D) desiccated cells treated with CQ, (E) primary HCE cells treated with CsA, and (F) desiccated cells treated with CsA (Figure 1(e)). (f) Percentage of cell viability of primary HCE cells exposed to desiccation stress with and without (CQ/ CsA) treatment (Figure 1(f)). Data are the mean ± SD, n = 3; statistical significance was denoted (p < 0.05, ∗∗p < 0.01, ∗∗∗p < 0.001, and ns-nonsignificant as compared to desiccated cells). Note that --- is control, +-- are HCE/HCF cells exposed to desiccation (Des), -+- are HCE/HCF cells treated with chloroquine (CQ), ++- are desiccated HCE/HCF cells treated with chloroquine (CQ), --+ are HCE/HCF cells treated with Cyclosporine (CsA), and -++ are desiccated HCE/HCF cells treated with Cyclosporine (CsA).
Figure 2
Figure 2
Expression of inflammatory related genes and NFκB pathway proteins in HCE-T and HCF cells under desiccation stress. (a) The mRNA expression levels of MCP-1, MMP-9, IL-6, and TNF-α in HCE-T cells under desiccation stress (Figure 2(a)). (b) Immunoblot shows the phosphorylation status of p65 in HCE-T cells exposed to desiccation stress, with and without CQ/CsA treatment. Densitometric analysis of the blots showed the ratios of total p65 and phosphorylated p65 at (Ser536) (Figure 2(b)). (c) The mRNA expression levels of MCP-1, MMP-9, IL-6, and TNF-α in HCF cells under desiccation stress (Figure 2(c)). (d) Immunoblot shows the phosphorylation status of p65 in HCF cells exposed to desiccation with and without CQ/CsA treatment. Densitometric analysis of the blots showed the ratios of total p65 and phosphorylated p65 at (Ser536) (Figure 2(d)). Data are the mean ± SD, n = 3; statistical significance was denoted (p < 0.05, ∗∗p < 0.01, ∗∗∗p < 0.001, and ns- nonsignificant as compared to desiccated cells). Note that --- is control, +-- are HCE-T/HCF cells exposed to desiccation (Des), -+- are HCE-T/HCF cells treated with chloroquine (CQ), ++- are desiccated HCE-T/HCF cells treated with chloroquine (CQ), --+ are HCE-T/HCF cells treated with Cyclosporine (CsA) and -++ are desiccated HCE-T/HCF cells treated with cyclosporine (CsA).
Figure 3
Figure 3
Expression of p65 and IκBα proteins in HCE-T cells under desiccation stress. (a) Immunoblots of nuclear/cytoplasmic fractions shows protein expression levels of p65 (NFκB) and IκBα in HCE-T cells exposed to desiccation stress (treated with or without CQ and CsA). Densitometric analysis of the blots shows the ratios of total p65 to IκBα (Figure 3(a)). (b) GFP-RelA translocation images at 20X magnification of HCE-T cells under desiccation stress treated with/without CQ and CsA (Figure 3(b)). ((A) Non-desiccated/control HCE-T cells, (B) desiccated HCE-T cells, (C) desiccated cells treated with CQ, (D) desiccated HCE-T cells treated with CsA, (E) HCE-T cells treated with TNF-α (10 ng/ml), and (F) HCE-T cells treated with TNF-α (10 ng/ml)+CsA. Data are the mean ± SD values, n = 3, statistical significance was denoted (p < 0.05, ∗∗p < 0.01, ∗∗∗p < 0.001, ns- nonsignificant as compared to levels of desiccated cells). Note that --- is control, +-- HCE-T cells exposed to desiccation (Des), -+- HCE-T cells treated with chloroquine (CQ), ++- desiccated HCE-T cells treated with chloroquine (CQ), --+ HCE-T cells treated with cyclosporine (CsA), and -++ desiccated HCE-T cells treated with cyclosporine (CsA).
Figure 4
Figure 4
(a)-(c) Expression of autophagy related genes and proteins in HCE-T under desiccation stress. (a) The mRNA expression levels of LC3A, LC3B, ATG7, and LAMP1 in HCE-T cells exposed to desiccation stress, treated with and without CQ/CsA treatment normalized with β-actin (Figure 4(a)). (b) Immunoblot shows the protein levels of LC3, p62, and LAMP1 HCE-T cells exposed to desiccation stress and treated with and without CQ/CsA treatment (Figure 4(b)). Densitometric analysis of the blots showed the ratios of LAMP1, LC3-II and p62 to GAPDH. (c) Westernblot shows the expression levels of Beclin-1 protein in HCE-T cells exposed to desiccation stress and treated with and without CQ/CsA treatment (Figure 4(c)). Densitometric analysis of the blots showed the ratios of Beclin-1 to GAPDH. Data are the mean ± SD values, n = 3, statistical significance denoted (p < 0.05, ∗∗p < 0.01, ∗∗∗p < 0.001, and ns- nonsignificant as compared to levels of desiccated cells). Note that --- is control, +-- HCE-T cells exposed to desiccation (Des), -+- HCE-T cells treated with chloroquine (CQ), ++- desiccated HCE-T cells treated with Chloroquine (CQ), --+ HCE-T cells treated with cyclosporine (CsA), and -++ desiccated HCE-T cells treated with cyclosporine (CsA).
Figure 5
Figure 5
(a)-(c) Quantification of autophagosomes and lysosomes in desiccated HCE-T cells. (a) Cyto-Id staining for quantification of autophagosome staining in HCE-T cells under desiccation stress, with and without CQ/CsA treatment (Figure 5(a)). (b) LTR- lysotracker red staining to measure lysosome levels staining in HCE-T cells under desiccation stress, with and without CQ/CsA treatment (Figure 5(b)). (c) AO- Lysosomal pH assessed using acridine orange (AO) staining in HCE-T cells under desiccation stress, with and without CQ/CsA treatment (Figure 5(c)). UT- control or undesiccated cells, Des- HCE-T cells exposed to desiccation, CQ- HCE-T cells treated with chloroquine (CQ), CQ+Des- desiccated HCE-T cells treated with chloroquine (CQ), CsA- HCE-T cells treated with cyclosporine (CsA), and CsA+Des- desiccated HCE-T cells treated with cyclosporine (CsA).
Figure 6
Figure 6
Phosphorylation levels of MAP kinase, AKT/p70S6kinase and AMPK proteins in HCE-T cells under desiccation stress. Immunoblot shows phosphorylated ERK1/2, p38, AKT and P70S6kinase, and AMPK (Figure 6). Densitometric analysis of the blots showed the ratios of phosphorylated AKT, p70s6kinase, ERK1/2 and p38 to AKT, p70s6kinase, ERK1/2, and p38. Data are the mean ± SD values, n = 3, statistical significance denoted (p < 0.05, ∗∗p < 0.01, ∗∗∗p < 0.001, and ns- nonsignificant as compared to desiccated cells). Note that --- is control, +-- HCE-T cells exposed to desiccation (Des), -+- HCE-T cells treated with chloroquine (CQ), ++- desiccated HCE-T cells treated with chloroquine (CQ), --+ HCE-T cells treated with cyclosporine (CsA), and -++ desiccated HCE-T cells treated with cyclosporine (CsA).

References

    1. Lemp M. A., Baudouin C., Baum J., et al. The definition and classification of dry eye disease: Report of the definition and classification subcommittee of the International Dry Eye WorkShop (2007) The Ocular Surface. 2007;5(2):75–92. doi: 10.1016/S1542-0124(12)70081-2.
    1. Stern M. E., Gao J., Schwalb T. A., et al. Conjunctival T-cell subpopulations in Sjogren's and non-Sjogren's patients with dry eye. Investigative Ophthalmology & Visual Science. 2002;43(8):2609–2614.
    1. Solomon A., Dursun D., Liu Z., Xie Y., Macri A., Pflugfelder S. C. Pro- and anti-inflammatory forms of interleukin-1 in the tear fluid and conjunctiva of patients with dry-eye disease. Investigative Ophthalmology & Visual Science. 2001;42(10):2283–2292.
    1. Čejková J., Ardan T., Šimonová Z., et al. Nitric oxide synthase induction and cytotoxic nitrogen-related oxidant formation in conjunctival epithelium of dry eye (Sjögren's syndrome) Nitric Oxide: Biology and Chemistry. 2007;17(1):10–17. doi: 10.1016/j.niox.2007.04.006.
    1. Arend W. P., Dayer J.-M. Inhibition of the production and effects of interleukin-1 and tumor necrosis factor α in rheumatoid arthritis. Arthritis & Rheumatism. 1995;38(2):151–160. doi: 10.1002/art.1780380202.
    1. Bresnihan B., Alvaro-Gracia J. M., Cobby M., et al. Treatment of rheumatoid arthritis with recombinant human interleukin-1 receptor antagonist. Arthritis & Rheumatology. 1998;41(12):2196–2204. doi: 10.1002/1529-0131(199812)41:12<2196::AID-ART15>;2-2.
    1. Choy E. H. S., Isenberg D. A., Garrood T., et al. Therapeutic benefit of blocking interleukin-6 activity with an anti-interleukin-6 receptor monoclonal antibody in rheumatoid arthritis: A randomized, double-blind, placebo-controlled, dose-escalation trial. Arthritis & Rheumatology. 2002;46(12):3143–3150. doi: 10.1002/art.10623.
    1. Weber S. M., Chen J.-M., Levitz S. M. Inhibition of mitogen-activated protein kinase signaling by chloroquine. The Journal of Immunology. 2002;168(10):5303–5309. doi: 10.4049/jimmunol.168.10.5303.
    1. Fougeray S., Pallet N. Mechanisms and biological functions of autophagy in diseased and ageing kidneys. Nature Reviews Nephrology. 2015;11(1):34–45. doi: 10.1038/nrneph.2014.201.
    1. Chaabane W., User S. D., El-Gazzah M., et al. Autophagy, apoptosis, mitoptosis and necrosis: interdependence between those pathways and effects on cancer. Archivum Immunologiae et Therapia Experimentalis. 2013;61(1):43–58. doi: 10.1007/s00005-012-0205-y.
    1. Melles R. B., Marmor M. F. Pericentral retinopathy and racial differences in hydroxychloroquine toxicity. Ophthalmology. 2015;122(1):110–116. doi: 10.1016/j.ophtha.2014.07.018.
    1. Al-Bari M. A. Chloroquine analogues in drug discovery: new directions of uses, mechanisms of actions and toxic manifestations from malaria to multifarious diseases. Journal of Antimicrobial Chemotherapy. 2015 doi: 10.1093/jac/dkv018.
    1. Venables P. J. Management of patients presenting with Sjogren's syndrome. Best Practice & Research Clinical Rheumatology. 2006;20(4):791–807. doi: 10.1016/j.berh.2006.05.003.
    1. Kruize A. A., Hene R. J., Kallenberg C. G. M., et al. Hydroxychloroquine treatment for primary Sjogren's syndrome: A two year double blind crossover trial. Annals of the Rheumatic Diseases. 1993;52(5):360–364. doi: 10.1136/ard.52.5.360.
    1. Into T., Horie T., Inomata M., et al. Basal autophagy prevents autoactivation or enhancement of inflammatory signals by targeting monomeric MyD88. Scientific Reports. 2017;7(1) doi: 10.1038/s41598-017-01246-w.
    1. Yoon K.-C., Jeong I.-Y., Park Y.-G., Yang S.-Y. Interleukin-6 and tumor necrosis factor-α levels in tears of patients with dry eye syndrome. Cornea. 2007;26(4):431–437. doi: 10.1097/ICO.0b013e31803dcda2.
    1. Shetty R., Sethu S., Chevour P., et al. Lower Vitamin D Level and Distinct Tear Cytokine Profile Were Observed in Patients with Mild Dry Eye Signs but Exaggerated Symptoms. Translational Vision Science & Technology. 2016;5(6):p. 16. doi: 10.1167/tvst.5.6.16.
    1. Dhamodaran K., Subramani M., Jeyabalan N., et al. Characterization of ex vivo cultured limbal, conjunctival, and oral mucosal cells: a comparative study with implications in transplantation medicine. Molecular Vision. 2015;21:828–845.
    1. Wu J., Du Y., Mann M. M., Funderburgh J. L., Wagner W. R. Corneal stromal stem cells versus corneal fibroblasts in generating structurally appropriate corneal stromal tissue. Experimental Eye Research. 2014;120:71–81. doi: 10.1016/j.exer.2014.01.005.
    1. Klionsky D. J., Abdelmohsen K., Abe A., Abedin M. J., Abeliovich H., Acevedo Arozena A., et al. Guidelines for the use and interpretation of assays for monitoring autophagy (3rd edition)," Autophagy. Autophagy. (3rd) 2016;12:1–222.
    1. Hovakimyan M., Ramoth T., Löbler M., et al. Evaluation of protective effects of trehalose on desiccation of epithelial cells in three dimensional reconstructed human corneal epithelium. Current Eye Research. 2012;37(11):982–989. doi: 10.3109/02713683.2012.700754.
    1. Matsuo T. Trehalose protects corneal epithelial cells from death by drying. British Journal of Ophthalmology. 2001;85(5):610–612. doi: 10.1136/bjo.85.5.610.
    1. Geerling G., MacLennan S., Hartwig D. Autologous serum eye drops for ocular surface disorders. British Journal of Ophthalmology. 2004;88(11):1467–1474. doi: 10.1136/bjo.2004.044347.
    1. Giebel J., Woenckhaus C., Fabian M., Tost F. Age-related differential expression of apoptosis-related genes in conjunctival epithelial cells. Acta Ophthalmologica Scandinavica. 2005;83(4):471–476. doi: 10.1111/j.1600-0420.2005.00472.x.
    1. Tost F. [Practical conjunctival cytology] Der Ophthalmologe. 1999;96(4):276–289. doi: 10.1007/s003470050407.
    1. Massingale M. L., Li X., Vallabhajosyula M., Chen D., Wei Y., Asbell P. A. Analysis of inflammatory cytokines in the tears of dry eye patients. Cornea. 2009;28(9):1023–1027. doi: 10.1097/ICO.0b013e3181a16578.
    1. Enriquez-de-Salamanca A., Castellanos E., Stern M. E., et al. Tear cytokine and chemokine analysis and clinical correlations in evaporative-type dry eye disease. Molecular Vision. 2010;16:862–873.
    1. Pflugfelder S. C., Jones D., Ji Z., Afonso A., Monroy D. Altered cytokine balance in the tear fluid and conjunctiva of patients with Sjogren's syndrome keratoconjunctivitis sicca. Current Eye Research. 1999;19(3):201–211. doi: 10.1076/ceyr.19.3.201.5309.
    1. Chotikavanich S., de Paiva C. S., Li D. Q., et al. Production and activity of matrix metalloproteinase-9 on the ocular surface increase in dysfunctional tear syndrome. Investigative Ophthalmology & Visual Science. 2009;50(7):3203–3209. doi: 10.1167/iovs.08-2476.
    1. Acera A., Rocha G., Vecino E., Lema I., Durán J. A. Inflammatory markers in the tears of patients with ocular surface disease. Ophthalmic Research. 2008;40(6):315–321. doi: 10.1159/000150445.
    1. Corrales R. M., Stern M. E., De Paiva C. S., Welch J., Li D.-Q., Pflugfelder S. C. Desiccating stress stimulates expression of matrix metalloproteinases by the corneal epithelium. Investigative Ophthalmology & Visual Science. 2006;47(8):3293–3302. doi: 10.1167/iovs.05-1382.
    1. Higuchi A., Kawakita T., Tsubota K. IL-6 induction in desiccated corneal epithelium in vitro and in vivo. Molecular Vision. 2011;17:2400–2406.
    1. Baldwin Jr. A. S. The NF-κB and IκB proteins: new discoveries and insights. Annual Review of Immunology. 1996;14:649–683. doi: 10.1146/annurev.immunol.14.1.649.
    1. Qian M., Fang X., Wang X. Autophagy and inflammation. linical and Translational Medicine. 2017;6(1) doi: 10.1186/s40169-017-0154-5.
    1. Ratnakumar S., Hesketh A., Gkargkas K., et al. Phenomic and transcriptomic analyses reveal that autophagy plays a major role in desiccation tolerance in Saccharomyces cerevisiae. Molecular BioSystems. 2011;7(1):139–149. doi: 10.1039/c0mb00114g.
    1. Shetty R., Sharma A., Pahuja N., et al. Oxidative stress induces dysregulated autophagy in corneal epithelium of keratoconus patients. PLoS ONE. 2017;12(9)
    1. Xing S., Zhang Y., Li J., et al. Beclin 1 knockdown inhibits autophagic activation and prevents the secondary neurodegenerative damage in the ipsilateral thalamus following focal cerebral infarction. Autophagy. 2012;8(1):63–76. doi: 10.4161/auto.8.1.18217.
    1. Ertel W., Morrison M. H., Ayala A., Chaudry I. H. Chloroquine attenuates hemorrhagic shock-induced suppression of Kupffer cell antigen presentation and major histocompatibility complex class II antigen expression through blockade of tumor necrosis factor and prostaglandin release. Blood. 1991;78(7):1781–1788.
    1. Picot S., Peyron F., Donadille A., Vuillez J.-P., Barbe G., Ambroise-Thomas P. Chloroquine-induced inhibition of the production of TNF, but not of IL-6, is affected by disruption of iron metabolism. The Journal of Immunology. 1993;80(1):127–133.
    1. van den B. E., Borne B. A., de Rooij H. H., le Cessie S., Verweij C. L. Chloroquine and hydroxychloroquine equally affect tumor necrosis factor-alpha, interleukin 6, and interferon-gamma production by peripheral blood mononuclear cells. The Journal of Rheumatology. 1997;24(1):55–60.
    1. Karres I., Kremer J.-P., Dietl I., Steckholzer U., Jochum M., Ertel W. Chloroquine inhibits proinflammatory cytokine release into human whole blood. American Journal of Physiology-Regulatory, Integrative and Comparative Physiology. 1998;274(4):R1058–R1064. doi: 10.1152/ajpregu.1998.274.4.R1058.
    1. Weber S. M., Levitz S. M. Chloroquine interferes with lipopolysaccharide-induced TNF-α gene expression by a nonlysosomotropic mechanism. The Journal of Immunology. 2000;165(3):1534–1540. doi: 10.4049/jimmunol.165.3.1534.
    1. Chang L., Karin M. Mammalian MAP kinase signalling cascades. Nature. 2001;410(6824):37–40. doi: 10.1038/35065000.
    1. Cowan K. J., Storey K. B. Mitogen-activated protein kinases: new signaling pathways functioning in cellular responses to environmental stress. Journal of Experimental Biology. 2003;206(7):1107–1115. doi: 10.1242/jeb.00220.
    1. Huang Z., Tunnacliffe A. Response of human cells to desiccation: Comparison with hyperosmotic strees response. The Journal of Physiology. 2004;558(1):181–191. doi: 10.1113/jphysiol.2004.065540.
    1. Bennett B. L., Sasaki D. T., Murray B. W., et al. SP600125, an anthrapyrazolone inhibitor of Jun N-terminal kinase. Proceedings of the National Acadamy of Sciences of the United States of America. 2001;98(24):13681–13686. doi: 10.1073/pnas.251194298.
    1. Strozyk E., Kulms D. The role of AKT/mTOR pathway in stress response to UV-irradiation: Implication in skin carcinogenesis by regulation of apoptosis, autophagy and senescence. International Journal of Molecular Sciences. 2013;14(8):15260–15285. doi: 10.3390/ijms140815260.
    1. Park J., Kwon D., Choi C., Oh J.-W., Benveniste E. N. Chloroquine induces activation of nuclear factor-κB and subsequent expression of pro-inflammatory cytokines by human astroglial cells. Journal of Neurochemistry. 2003;84(6):1266–1274. doi: 10.1046/j.1471-4159.2003.01623.x.

Source: PubMed

3
Abonner