Pathogenetic mechanisms of dry eye syndrome in a novel coronavirus infection caused by SARS-CoV-2

Year & Volume - Issue: 
Authors: 
Tatiana N. Safonova, Galina V. Zaitseva
Heading: 
Article type: 
CID: 
e0306
PDF File: 
Abstract: 
The goal of this review was to analyze current knowledge on dry eye disease pathogenesis in a novel coronavirus infection (COVID-19) caused by SARS-CoV-2. Arguments are presented in favor of several possible pathogenic mechanisms of the disease development: inflammation and/or microcirculatory disorders aggravated by exposure to electromagnetic radiation of personal computers and by use of personal protective equipment.
Cite as: 
Safonova TN, Zaitseva GV. Pathogenetic mechanisms of dry eye syndrome in a novel coronavirus infection caused by SARS-CoV-2. Russian Open Medical Journal 2022; 11: e0306.

At the end of 2019, a new severe acute respiratory syndrome- related coronavirus 2 (SARS-CoV-2) has appeared. The clinical spectrum of COVID-19 varies from asymptomatic infection or mild self-limited constitutional symptoms to a hyperinflammatory state (the so-called cytokine storm) with subsequent acute respiratory distress syndrome and lethal outcome [1-3].

SARS-CoV-2 is characterized by high virulence and pronounced tropism of various tissues. This correlates with key mutations in the receptor-binding domain that mediates direct contact with the cellular receptor of angiotensin-converting enzyme-2 (ACE-2) [4]. It is well known that the virus predominantly spreads by airborne transmission route [5, 6]. Common symptoms of infection include hyperthermia, dry cough, shortness of breath, and fatigue. Other clinical signs have been reported, such as gastrointestinal tract dysfunctions [7, 8] and ocular surface disorder - dry eye syndrome (DES) [9, 10].

It should be noted that DES development may even precede the onset of respiratory symptoms [11-13].

Due to the SARS-CoV-2 surface spike glycoprotein [14], the virus in the biological fluid binds with the ACE-2 receptors located on the corneal and conjunctival surface, thereby providing infection with potential site of entry [15-17], which may lead to ocular surface damage and further viral replication [18, 19]. This is evidenced by virus detection in the tear fluid and by polymerase chain reaction (real-time PCR) testing of lower eyelid conjunctival scrapping in patients with positive test results for COVID-19. The meta-analysis conducted by N. Nasiri et al. demonstrated that the organ of vision is involved in the pathological process in 11.03% of patients (95% CI: 5.71-17.72). Most often, patients are presented with dry eye symptoms, and/or foreign body sensation (16%), red eyes (13.3%), lacrimation (12.8%), itching (12.6%), eye pain (9.6%). %), and discharge (8.8%). Approximately one in ten COVID-19 patients develops at least one ocular symptom. Conjunctivitis had the highest prevalence among patients with confirmed COVID-19 (88.8%) [20, 21]. Clinical manifestation of SARS-CoV-associated ocular surface damage includes severe injection, chemosis, follicular conjunctivitis, phlyctena on the bulbar conjunctiva, and mild corneal epithelial edema [22-24].

The mechanism of the damaging effect of the virus on the eye tissue is not fully understood yet. A possible explanation is the direct damaging effect of virions, as well as inflammation activated by the immune system, which triggers compensatory and regenerative processes leading to the cytokine release syndrome (cytokine storm) [25]. The term cytokine storm was first introduced in 1993 to describe graft-versus-host disease and similar processes of rapid cytokine releases associated with autoimmune hemophagocytic lymphohistiocytosis, sepsis, cancer, acute immunotherapeutic responses, and infectious diseases [26]. It is now well established by numerous published studies what causes the inflammatory response cascade. An immune system dysregulation (activation of immune cells via Toll-like receptors - TLR, reduced anti-inflammatory response, etc.) is among major hypotheses on its origin. Activation of immune responses leads to enlarged cytokines levels (IL-6, IL-1P, IL-10, TNF-a, GM-CSF, IP-10, IFN-induced protein-10, IL-17, MCP-3, IL-1RA, etc.), which causes severe inflammatory responses in various body tissues [27-29].

Local and systemic cytokine responses are an integral part of the primary response to infection. In this case, the process of antibody-dependent cellular cytotoxicity is triggered, regulating the activity of cellular and humoral immune response and cytokine secretion by NK-cells (natural killer cells), macrophages, granulocytes, etc. [30]. Cytokines, such as interferon-a/S (IFN-a/S) and interleukin-1S (IL-1S), produced by epithelial cells, enable adjacent cell protection via expression of genes encoding IFN, while simultaneously activating immunocompetent NK-cells, which increases lytic potential of NK cells and stimulates the secretion of IFN-y [31]. High levels of activated TNF-a, IL-12, and IL-6 lead to  hyperstimulation of NK cells [32]. With the development of the disease, T-cell responses induce additional production of cytokines against viral cytotoxicity, which results in addition of TLR ligands [33].

With SARS-CoV-2 infection in the blood serum, a high level of cytokines (IL-1P, IL-6, IL-8, IL-10, IFN-y, and TNF-a) was determined [34]. Same cytokines were detected in the tear fluid via multiplex ligation-dependent probe amplification (MLPA) and enzyme-linked immunosorbent assay (ELISA) methods [35]. These findings reinforce the hypothesis on the pathogenetic mechanism of DES development based on the inflammatory process due to the presence of high content of proinflammatory cytokines (IL-1P, IL-6, IL-8, IL-10, IFN-y, and TNF-a) in the tear fluid [36-40]. It was previously observed that SARS-CoV-2 contributes to the progression of DES manifestation. A cytokine storm occurs more often with more severe course of the SARS-CoV-2 disease, leading to the ocular surface damage [41-44]. It was estimated that IL-1P, IL-6, IL-8, IL-10, IFN-y, and TNF-a were also represented and involved in coagulation cascade [45-48].

The presented Figure 1 demonstrates the development of coagulopathy in COVID-19 infection. Activation of mononuclear cells results in a procoagulant response, in which abnormal fibrin deposition and an increase in the incidence of venous thromboembolism occur. SARS-CoV-2 also causes endothelial damage, which is succeeded by release of plasminogen activators, which, in combination with a procoagulant reaction, elevates serum D-dimer, C-reactive protein, ferritin, and procalcitonin levels [49-52]. The simultaneous release of high-molecular-weight multimers of von Willebrand factor leads to thrombosis in the organs and tissues, including blood vessels in the eye [53].

 

Figure 1. Schematic representation of the pathogenesis of COVID-19 coagulopathy caused by the SARS-CoV-2

 

These pathophysiological mechanisms contribute to disseminated intravascular coagulation (DIC), which occurs in over 70% of patients with SARS-CoV-2 infection [54, 55]. Besides hypercoagulation associated with DIC, activation of blood plasma coagulation pathway, intravascular aggregation of platelets and other blood cells, and microcirculation disorders in various organs are present. As a result, a morphological blockade of the bloodstream by fibrin masses and cell aggregates is formed which leads to ischemia and tissue hypoxia [56-58]. The same pathogenetic mechanism underlies DES development in the case of vasculitis associated with systemic diseases [59].

The vessels of the bulbar conjunctiva are the most accessible for visualization to analyze the microcirculatory bloodstream in arterioles, venules, and capillaries located in the conjunctival superficial layers. It is well known that the conjunctiva receives its blood supply from two sources: the superior and inferior arterial arches of the eyelids, which are formed of the lacrimal and anterior ethmoidal arteries, and the anterior ciliary arteries, which originate from the muscular branches. The external carotid artery also partly supplies blood to the bulbar conjunctiva vascular system. There are several vascular layers in the conjunctiva: superficial (subepithelial), middle (subconjunctival), and deep (episcleral). The highest density of capillaries is observed in the eyelids parallel to the conjunctival folds [60]. It is important to analyze the changes in microvasculature due to its significant role in the process of lacrimation: some components of the tear fluid are provided immediately from the microvasculature. Disorder in microvasculature, caused by SARS-CoV-2, may result in qualitative and quantitative disorders of tear fluid composition.

Several methods may provide an objective assessment of the state of the conjunctival microvascular network.

L. MacKenzie et al., conducted a study of saturation of conjunctival vessels using the principle of retinal oximetry. Their results showed a decrease in hemoglobin saturation with oxygen in the conjunctival and episcleral vessels under conditions of mild hypoxia. In these circumstances, episcleral vessels expand, while conjunctival vessels do not undergo significant changes due to rapid reoxygenation and oxygen diffusion from the environment [61].

Some authors proposed a method for assessing oxygenation in the microcirculatory bloodstream of the anterior eye segment (palpebral and bulbar conjunctiva): diffuse reflectance spectroscopy, utilizing visible light. This method allows determining blood oxygenation from a series of multispectral images obtained with band-pass filters, based on the absorption spectrum of hemoglobin wavelengths, which provides an objective assessment of vascular bloodstream in the ocular anterior segment [62].

Video biomicroscopy of the bulbar conjunctiva allows visualizing all anatomical parts of the microvasculature (arterioles, venules, capillaries, arterio-venous anastomoses) and to assess vascular permeability, interstitial space, and rheological properties of microcirculatory bloodstream [63].

Another method to assess conjunctival blood and lymph flow is the method of laser Doppler flowmetry (LDF) conducted with LASMA MC-1 peripheral blood flow and lymph flow analyzer. LDF is a non-invasive diagnostic method based on optical laser probing of tissues and analysis of the scattered reflected signal induced by moving erythrocytes. The analyzer filters the frequency spectra of the Doppler shift of laser radiation scattered from erythrocytes in the microvasculature, and scatterers from lymph formation in the lymphatic vessels in the speed ranges corresponding to their movement. Laser radiation reflected from static (stationary) tissue components does not change its frequency, while reflected radiation from moving particles (erythrocytes) has a Doppler frequency shift relative to the probing signal. The variable component of the reflected signal is determined by two factors: the concentration of erythrocytes in the test sample and the speed of their movement. This method has high specificity and high sensitivity and allows determining the parameters of microcirculation and lymph flow, such as the coefficient of variation, and neurogenic and myogenic fluctuations [63, 64].

Previously conducted studies recognized the role played by two more factors, which may also contribute to the onset and/or progression of DES in SARS-CoV-2 infection.

One of them is electromagnetic radiation, which is a risk factor for the development of DES due to frequent and prolonged use of personal computers and mobile devices during distance learning or remote work [65]. Blinking frequency decreases to an average of 4-5 times per minute, compared with reference value of up to 15, which leads to excessive evaporation of the tear fluid and precorneal tear film instability. Ultimately, this leads to xerotic changes in the cornea and conjunctiva.

Another triggering factor of DES, associated with SARS-CoV-2, is an extensive use of personal protective equipment (masks, respirators). Improper and prolonged wearing of masks/respirators causes an uneven distribution of exhaled warm air and increase in the evaporation of tear fluid from the ocular surface [66]. This mechanism of DES development is similar to the mechanism underlying constant positive airway pressure (CPAP) therapy, which results in augmented tear evaporation, eye irritation, and squamous metaplasia of the conjunctiva [67].

Collectively, clinical experience to date suggests that COVID-19 infection with SARS-CoV-2 results in ocular surface damage or progression of DES. However, studies of eye damage in patients with SARS-CoV-2 mainly involve the description of the clinical manifestations of the disease. Currently, there are no data on an objective assessment of the ocular surface lesion and its severity and reversibility.

The existing body of research suggests that the mechanism of DES development in SARS-CoV-2 infection is probably associated with the activation of one or two pathways of pathogenesis: inflammation and/or microcirculatory disorders complicated by exposure to electromagnetic radiation from personal computers and the use of personal protective equipment. There are concerns about protective measures during the COVID-19 pandemic and how they could lead to an increase in the number of patients with ocular surface disease. Thus, further studies are needed to determine the components of the pathogenesis of DES in the case of SARS-CoV-2 infection.

References: 
  1. Rodriguez-Morales AJ, Bonilla-Aldana DK, Balbin-Ramon GJ, Rabaan AA, Sah R, Paniz-Mondolfi A, et al. History is repeating itself: Probable zoonotic spillover as the cause of the 2019 novel coronavirus epidemic. Infez Med 2020; 28(1): 3-5. https://pubmed.ncbi.nlm.nih.gov/32009128.
  2. Malik YS, Sircar S, Bhat S, Sharun K, Dhama K, Dadar M, et al. Emerging novel coronavirus (2019-nCoV)-current scenario, evolutionary perspective based on genome analysis and recent developments. Vet Q 2020; 40(1): 68-76. https://doi.org/10.1080/01652176.2020.1727993.
  3. Jiehao C, Jin X, Daojiong L, Zhi Y, Lei X, Zhenghai Q, et al. A case series of children with 2019 novel coronavirus infection: Clinical and epidemiological features. Clin Infect Dis 2020; 71(6): 1547-1551. https://doi.org/10.1093/cid/ciaa198.
  4. Zhou P, Yang XL, Wang XG, Hu B, Zhang L, Zhang W, et al. A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature 2020; 579(7798): 270-273. https://doi.org/10.1038/s41586-020-2012-7.
  5. Ong SWX, Tan YK, Chia PY, Lee TH, Ng OT, Wong MSY, et al. Air, surface environmental, and personal protective equipment contamination by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) from a symptomatic patient. JAMA 2020; 323(16): 1610-1612. https://doi.org/10.1001/jama.2020.3227.
  6. Lu Cw, Liu Xf and Jia Zf. 2019-ncov transmission through the ocular surface must not be ignored. Lancet 2020;395(10224): e39. https://doi.org/10.1016/S0140-6736(20)30313-5.
  7. Gu J, Han B, Wang J. COVID-19: Gastrointestinal manifestations and potential fecal-oral transmission. Gastroenterology 2020; 158(6): 1518-1519. https://doi.org/10.1053/j.gastro.2020.02.054.
  8. Luo S, Zhang X, Xu H. Don't overlook digestive symptoms in patients with 2019 novel coronavirus disease (COVID-19). Clin Gastroenterol Hepatol 2020; 18(7): 1636-1637. https://doi.org/10.1016/j.cgh.2020.03.043.
  9. Guan WJ, Ni ZY, Hu Y, Liang WH, Ou CQ, He JX, et al. Clinical Characteristics of Coronavirus Disease 2019 in China. N Engl J Med 2020; 382(18): 1708-1720. https://doi.org/10.1056/NEJMoa2002032.
  10. Henry BM, Vikse J.N Clinical characteristics of Covid-19 in China. Reply. Engl J Med 2020; 382(19): 1860-1861. https://doi.org/10.1056/NEJMc2005203.
  11. Hong N, Yu W, Xia J, Shen Y, Yap M, Han W. Evaluation of ocular symptoms and tropism of SARS-CoV-2 in patients confirmed with COVID-19. Acta Ophthalmol 2020; 98(5): e649-e655. https://doi.org/10.1111/aos.14445.
  12. Chen L, Deng C, Chen X, Zhang X, Chen B, Yu H, et al. Ocular manifestations and clinical characteristics of 535 cases of COVID-19 in Wuhan, China: A cross-sectional study. Acta Ophthalmol 2020; 98(8): e951-e959. https://doi.org/10.1111/aos.14472.
  13. Cheong KX. Systematic review of ocular involvement of SARS-CoV-2 in coronavirus disease 2019. Curr Ophthalmol Rep 2020; 8(4): 185-194. https://doi.org/10.1007/s40135-020-00257-7.
  14. Hoffmann M, Kleine-Weber H, Pohlmann S. A multibasic cleavage site in the spike protein of SARS-CoV-2. is essential for infection of human lung cells. Mol Cell 2020; 78(4): 779-784 e5. https://doi.org/10.1016/j.molcel.2020.04.022.
  15. Douglas KAA, Douglas VP, Moschos MM. Ocular manifestations of COVID-19 (SARS-CoV-2): A critical review of current literature. In Vivo 2020; 34(3): 1619-1628. https://doi.org/10.21873/invivo.11952.
  16. Olofsson S, Kumlin U, Dimock K, Arnberg N. Avian influenza and sialic acid receptors: More than meets the eye? Lancet Infect Dis 2005; 5(3): 184-188. https://doi.org/10.1016/j.cell.2020.09.032.
  17. Li Q, Wu J, Nie J, Zhang L, Hao H, Liu S, et al. The impact of mutations in SARS-CoV-2 spike on viral infectivity and antigenicity. Cell 2020; 182(5): 1284-1294. https://doi.org/10.1016/j.cell.2020.07.012.
  18. Belser JA, Rota PA, Tumpey TM. Ocular tropism of respiratory viruses. Microbiol Mol Biol Rev 2013; 77: 144-156. https://doi.org/10.1128/mmbr.00058-12.
  19. Shen J, Wu J, Yang Y, Wang P, Luo T, Guo Y, et al. The paradoxical problem with COVID-19 ocular infection: Moderate clinical manifestation and potential infection risk. Comput Struct Biotechnol J 2021; 19: 1063-1071. https://doi.org/10.1016/j.csbj.2021.01.039.
  20. Nasiri N, Sharifi H, Bazrafshan А, Noori А, Karamouzian M, Sharifi A. Ocular manifestations of COVID-19: A systematic review and meta­analysis. J Ophthalmic Vis Res 2021; 16(1): 103-112. https://doi.org/10.18502/jovr.v16i1.8256.
  21. Inomata T, Kitazawa K, Kuno T, Sung J, Nakamura M, Iwagami M, et al. Clinical and prodromal ocular symptoms in coronavirus disease: A systematic review and meta-analysis. Invest Ophthalmol Vis Sci 2020; 61(10):  29. https://doi.org/10.1167/iovs.61.10.29.
  22. Gupta PC, Kumar MP, Ram J. COVID-19 pandemic from an ophthalmology point of view. Indian J Med Res 2020; 151(5): 411-418. https://doi.org/10.4103/ijmr.IJMR 1369 20.
  23. Torres-Costa S, Lima-Fontes M, Falcao-Reis F, Falcao M. SARS-CoV-2 in ophthalmology: Current evidence and standards for clinical practice. Acta Med Port 2020; 33(9): 593-600. https://doi.org/10.20344/amp.14118.
  24. Maychuk DYu, Atlas SN, Loshkareva AO. Ocular manifestations of coronavirus infection COVID-19 (clinical observation). Vestnik Oftalmologii 2020; 136(4): 118-123. Russian. https://doi.org/10.17116/oftalma2020136041118.
  25. Wang J, Jiang M, Chen X, Montaner LJ. Cytokine storm and leukocyte changes in mild versus severe SARS-CoV-2 infection: Review of 3,939 COVID-19 patients in China and emerging pathogenesis and therapy concepts. J Leukoc Biol 2020; 108(1): 17-41. https://doi.org/10.1002/JLB.3COVR0520-272.
  26. Ferrara JL, Abhyankar S, Gilliland DG. Cytokine storm of graft-versus-host disease: A critical effector role for interleukin-1. Transplant Proc 1993; 25(1 Pt 2): 1216-1267. https://pubmed.ncbi.nlm.nih.gov/8442093.
  27. Chousterman BG, Swirski FK, Weber GF. Cytokine storm and sepsis disease pathogenesis. Semin Immunopathol 2017; 39(5): 517-528. https://doi.org/10.1007/s00281-017-0639-8.
  28. Crayne CB, Albeituni S, Nichols KE, Cron RQ. The immunology of macrophage activation syndrome. Front Immunol 2019; 10: 119. https://doi.org/10.3389/fimmu.2019.00119.
  29. Barry SM, Johnson MA, Janossy G. Cytopathology or immunopathology? The puzzle of cytomegalovirus pneumonitis revisited. Bone Marrow Transplant 2000; 26(6): 591-597. https://doi.org/10.1038/sj.bmt.1702562.
  30. Borrok MJ, Luheshi NM, Beyaz N, Davies GC, Legg JW, Wu H, et al. Enhancement of antibody dependent cell-mediated cytotoxicity by endowing IgG with FcaRI (CD89) binding. MAbs 2015; 7(4): 743-751. https://doi.org/10.1080/19420862.2015.1047570.
  31. Chatenoud L, Ferran C, Reuter A, Legendre C, Gevaert Y, Kreis H, et al. Systemic reaction to the anti-T-cell monoclonal antibody OKT3 in relation to serum levels of tumor necrosis factor and interferon­gamma. N Engl J Med 1989; 320(21): 1420-1421. https://doi.org/10.1056/NEJM198905253202117.
  32. Channappanavar R, Perlman S. Pathogenic human coronavirus infections: causes and consequences of cytokine storm and immunopathology. Semin Immunopathol 2017; 39(5): 529-539. https://doi.org/10.1007/s00281-017-0629-x.
  33. Li T, Xie J, He Y, Fan H, Baril L, Qiu Z, et al. Long-term persistence of robust antibody and cytotoxic T cell responses in recovered patients infected with SARS coronavirus. PLoS One 2006; 1(1): e24. https://doi.org/10.1371/journal.pone.0000024.
  34. Taghiloo S, Soltanshahi M, Aliyali M, Abedi S, Mehravaran H, Ajami A, Asgarian-Omran H. Cytokine profiling in Iranian patients with COVID- 19; association with clinical severity. Iran J Immunol 2021; 18(1): 54­64. https://doi.org/10.22034/iji.2021.87630.1810.
  35. Roda M, Corazza I, Bacchi Reggiani ML, Pellegrini M, Taroni L, Giannaccare G, et al. Dry eye disease and tear cytokine levels - A meta-analysis. Int J Mol Sci 2020; 21(9): 3111. https://doi.org/10.3390/ijms21093111.
  36. Cocho L, Fernandez I, Calonge MC, Martinez V, Gonzalez-Garcia MJ, Caballero L, et al. Biomarkers in ocular chronic graft versus host disease: Tear cytokine- and chemokine-based predictive model. Invest Opthalmol Vis Sci 2016; (57): 746-758. https://doi.org/10.1167/iovs.15-18615.
  37. Gambini G, Savastano MC , Savastano A, De Vico U , Crincoli E, Cozzupoli MG, Culiersi C, Rizzo S. Ocular surface impairment after COVID-19: A cohort study. Cornea 2020; 40(4):477-483. https://doi.org/10.1097/ICO.0000000000002643.
  38. Landsend ECS, Utheim 0A, Pedersen HR, Aass HCD, Lagali N, Dartt DA, et al. The level of inflammatory tear cytokines is elevated in congenital aniridia and associated with meibomian gland dysfunction. Investig Opthalmol Vis Sci 2018; 59(5): 2197-2204. https://doi.org/10.1167/iovs.18-24027.
  39. Mrugacz M, Ostrowska L, Bryl A, Szulc A, Zelazowska-Rutkowska B. Mrugacz G. Pro-inflammatory cytokines associated with clinical severity of dry eye disease of patients with depression. Adv Med Sci 2017; 62(2): 338-344. https://doi.org/10.1016/j.advms.2017.03.003.
  40. Yoon KC, Jeong IY, Park YG, Yang SY. Interleukin-6 and tumor necrosis factor-а levels in tears of patients with dry eye syndrome. Cornea 2007; 26(4): 431-437. https://doi.org/10.1097/ICO.0b013e31803dcda2.
  41. Chen N, Zhou M, Dong X, Qu J, Gong F, Han Y, et al. Epidemiological and clinical characteristics of 99 cases of 2019 novel coronavirus pneumonia in Wuhan, China: A descriptive study. Lancet 2020; 395(10223): 507-513. https://doi.org/10.1016/S0140-6736(20)30211- 7.
  42. Acera A, Rocha G, Vecino E, Lema I, Duran JA. Inflammatory markers in the tears of patients with ocular surface disease. Ophthalmic Res 2008; 40(6): 315-321. https://doi.org/10.1159/000150445.
  43. Bron AJ, de Paiva CS, Chauhan SK, Bonini S, Gabison EE, Jain S, et al. TFOS DEWS II pathophysiology report. Ocul Surf 2017; 15(3): 438-510. https://doi.org/10.1016/jJtos.2017.05.011.
  44. Wei Y, Gadaria-Rathod N, Epstein S, Asbell P. Tear cytokine profile as a noninvasive biomarker of inflammation for ocular surface diseases: Standard operating procedures. Invest Ophthalmol Vis Sci 2013; 54(13): 8327-8336. https://doi.org/10.1167/iovs.13-12132.
  45. Iba T, Levy JH, Levi M, Connors JM, Thachil J. Coagulopathy of coronavirus disease 2019. Crit Care Med 2020; 48(9): 1358-1364. https://doi.org/10.1097/CCM.0000000000004458.
  46. Magro C, Mulvey JJ, Berlin D, Nuovo G, Salvatore S, Harp J, et al. Complement associated microvascular injury and thrombosis in the pathogenesis of severe COVID19 infection: A report of five cases. Transl Res 2020; 220: 1-13. https://doi.org/10.1016/j.trsl.2020.04.007.
  47. Conti P, Ronconi G, Caraffa A, Gallenga CE, Ross R, Frydas I, et al. Induction of proinflammatory cytokines (IL-1 and IL-6) and lung inflammation by coronavirus-19 (COVI-19 or SARS-CoV-2): Anti­inflammatory strategies. J Biol Regul Homeost Agents 2020; 34(2): 327­331. https://doi.org/10.23812/conti-e.
  48. Mehta P, McAuley DF, Brown M, Sanchez E, Tattersall RS, Manson JJ; HLH Across Speciality Collaboration, UK. COVID-19. Consider cytokine storm syndromes and immunosuppression. Lancet 2020; 395(10229): 1033-1034. https://doi.org/10.1016/S0140-6736(20)30628-0.
  49. Zhang L, Yan X, Fan Q, Liu H, Liu X, Liu Z, et al. D-dimer levels on admission to predict in-hospital mortality in patients with Covid-19. J Thromb Haemost 2020; 18(6): 1324-1329. https://doi.org/10.1111/jth.14859.
  50. Amgalan A, Othman M. Exploring possible mechanisms for COVID19 induced thrombocytopenia: Unanswered questions. J Thromb Haemost 2020; 18(6): 1514-1516. https://doi.org/10.1111/jth.14832.
  51. Ji HL, Zhao R, Matalon S, Matthay MA. Elevated plasmin(ogen) as a common risk factor for COVID-19 susceptibility. Physiol Rev 2020; 100: 1065-1075. https://doi.org/10.1152/physrev.00013.2020.
  52. Shorr AF, Thomas SJ, Alkins SA, Fitzpatrick TM, Ling GS. D-dimer correlates with proinflammatory cytokine levels and outcomes in critically ill patients. Chest 2002; 121(4): 1262-1268. https://doi.org/10.1378/chest.121.4.1262.
  53. Levi M, Thachil J. Coronavirus Disease 2019 Coagulopathy: Disseminated intravascular coagulation and thrombotic microangiopathy - Either, neither, or both. Semin Thromb Hemost 2020; 46(7): 781-784. https://doi.org/10.1055/s-0040-1712156.
  54. Al-Samkari H, Karp Leaf RS, Dzik WH, Carlson JCT, Fogerty AE, Waheed A, et al. COVID-19 and coagulation: Bleeding and thrombotic manifestations of SARS-CoV-2 infection. Blood 2020; 136(4): 489-500. https://doi.org/10.1182/blood.2020006520.
  55. Toh CH, Hoots WK; SSC on Disseminated Intravascular Coagulation of the ISTH. The scoring system of the Scientific and Standardisation Committee on Disseminated Intravascular Coagulation of the International Society on Thrombosis and Haemostasis: A 5-year overview. J Thromb Haemost 2007; 5(3): 604-606. https://doi.org/10.1111/j.1538-7836.2007.02313.x.
  56. Touaoussa A, El Youssi H, El Hassani I, Hanouf D, El Bergui I, Zoulati G, et al. Disseminated intravascular coagulation: clinical and biological diagnosis. Ann Biol Clin (Paris) 2015; 73(6): 657-663. French. https://doi.org/10.1684/abc.2015.1100.
  57. Ho LW, Kam PC, Thong CL. Disseminated intravascular coagulation. Curr Anesth Crit Care 2005; 16(3): 151-161. https:ZZdoi.org/10.1111Zj.1469-0691.2004.01044.
  58. Bick RL. Disseminated intravascular coagulation: a review of etiology, pathophysiology, diagnosis, and management: Guidelines for care. Clin Appl Thromb Hemost 2002; 8(1): 1-31. https://doi.org/10.1177/107602960200800103.
  59. Menezo V, Lightman S. The eye in systemic vasculitis. Clin Med (Lond) 2004; 4(3): 250-254. https://doi.org/10.7861/clinmedicine.4-3-250.
  60. Eroshevsky TI. Eye Diseases. Moscow: Medicine, 1977; 246 p. Russian. https://ru1lib.org/book/3235844/4b12e7
  61. MacKenzie LE, Choudhary TR, McNaught AI, Harvey AR. In vivo oximetry of human bulbar conjunctival and episcleral microvasculature using snapshot multispectral imaging. Exp Eye Res 2016; 149: 48-58. https://doi.org/10.1016/j.exer.2016.06.008.
  62. Shmireva VF, Petrov SYu, Antonov AA, Stratonnikov AA, Savel'eva TA, Shevchik SA, et al. The study of the metabolism of the tissues in the anterior segment of the eye in relation to hemoglobin oxygenation in venous system at primary open-angle glaucoma. Glaukoma 2008; (3): 3-10. Russian. https://www.elibrary.ru/item.asp?id=11652548.
  63. Sirotin BZ, Zhmerenetsky KV. Microcirculation in Cardiovascular Diseases. Khabarovsk: Publishing House of the Far Eastern State Medical University. 2009;150 p. Russian. https://textarchive.ru/c-1648553-pall.html.
  64. Safonova TN, Kintyukhina NP, Sidorov VV, Gladkova OV, Reyn ES. Microcirculatory blood and lymph flow examination in eyelid skin by laser Doppler flowmetry. The Russian Annals of Ophthalmology 2017; 133(3): 16-21. Russian. https://doi.org/10.17116/oftalma2017133316-21.
  65. Takhchidi KP, Mitronina ML, Potapova LS, Sidorov VV. Investigation of microcirculation status of anterior segment of eyes by method of the laser doppler flowmetry in children with different refraction types. Fyodorov Journal of Ophthalmic Surgery 2011; (4): 49-53. Russian. https://www.elibrary.ru/item.asp?id=18903089.
  66. Uchino M, Yokoi N, Uchino Y, Dogru M, Kawashima M, Komuro A, et al. Prevalence of dry eye disease and its risk factors in visual display terminal users: The Osaka study. Am J Ophthalmol 2013; 156(4): 759­766. https://doi.org/10.1016/j.ajo.2013.05.040.
  67. Kuroyedov AV, Zavadski PCh, Brezhnev AYu, Gorodnichii VV, Gazizova IR, Seleznev AV, et al. Impact of personal protective equipment of respiratory and visual organs on development and progression of dry eye syndrome. Ophthalmology in Russia 2020; 17(3): 519-526. Russian. https://doi.org/10.18008/1816-5095-2020-3-519-526.
  68. Hayirci E, Yagci A, Palamar M, Basoglu OK, Veral A. The effect of continous positive airway pressure treatment for obstructive sleep apnea syndrome on the ocular surface. Cornea 2012; 31(6): 604-608. https://doi.org/10.1097/ICO.0b013e31824a2040.
About the Authors: 

Tatiana N. Safonova - MD, PhD, Lead Researcher, Department of Lacrimal Apparatus Pathology, Research Institute of Eye Diseases, Moscow, Russia. https://orcid.org/0000-0002-4601-0904. 
Galina V. Zaitseva - MD, PhD, Junior Researcher, Department of Lacrimal Apparatus Pathology, Research Institute of Eye Diseases, Moscow, Russia. https://orcid.org/0000-0001-8575-3076.

Received 8 April 2021, Revised 9 November 2021, Accepted 24 May 2022 
© 2021, Russian Open Medical Journal 
Correspondence to Galina V. Zaitseva. Phone: +79688707721. E-mail: privezentseva.galya@mail.ru.

DOI: 
10.15275/rusomj.2022.0306