Next Article in Journal
The Function of the Histamine H4 Receptor in Inflammatory and Inflammation-Associated Diseases of the Gut
Next Article in Special Issue
Ocular Graft-versus-Host Disease in a Chemotherapy-Based Minor-Mismatch Mouse Model Features Corneal (Lymph-) Angiogenesis
Previous Article in Journal
Extinction vs. Abstinence: A Review of the Molecular and Circuit Consequences of Different Post-Cocaine Experiences
Previous Article in Special Issue
MSCs Become Collagen-Type I Producing Cells with Different Phenotype in Allogeneic and Syngeneic Bone Marrow Transplantation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Cascade of Inflammatory, Fibrotic Processes, and Stress-Induced Senescence in Chronic GVHD-Related Dry Eye Disease

1
Department of Ophthalmology, Keio University School of Medicine, Tokyo 160-8582, Japan
2
Division of Cellular Signaling, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo 160-8582, Japan
3
Department of Immunology, School of Medicine, International University of Health and Welfare, Chiba 286-8686, Japan
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(11), 6114; https://doi.org/10.3390/ijms22116114
Submission received: 16 March 2021 / Revised: 30 May 2021 / Accepted: 31 May 2021 / Published: 6 June 2021
(This article belongs to the Special Issue Pathophysiological Mechanisms in Ocular Graft-versus-Host Disease)

Abstract

:
Ocular graft-versus-host disease (GVHD) is a major complication after allogeneic hematopoietic stem cell transplantation. Ocular GVHD affects recipients’ visual function and quality of life. Recent advanced research in this area has gradually attracted attention from a wide range of physicians and ophthalmologists. This review highlights the mechanism of immune processes and the molecular mechanism, including several inflammation cascades, pathogenic fibrosis, and stress-induced senescence related to ocular GVHD, in basic spectrum topics in this area. How the disease develops and what kinds of cells participate in ocular GVHD are discussed. Although the classical immune process is a main pathological pathway in this disease, senescence-associated changes in immune cells and stem cells may also drive this disease. The DNA damage response, p16/p21, and the expression of markers associated with the senescence-associated secretory phenotype (SASP) are seen in ocular tissue in GVHD. Macrophages, T cells, and mesenchymal cells from donors or recipients that increasingly infiltrate the ocular surface serve as the source of increased secretion of IL-6, which is a major SASP driver. Agents capable of reversing the changes, including senolytic reagents or those that can suppress the SASP seen in GVHD, provide new potential targets for the treatment of GVHD. Creating innovative therapies for ocular GVHD is necessary to treat this intractable ocular disease.

1. Introduction

Allogeneic hematopoietic stem cell transplantation (HSCT) is now an established treatment for hematological malignancies and other life-threatening hematological and genetic diseases [1]. The major barrier to successful HSCT is graft-versus-host disease (GVHD), which is one of the most frequent complications after HSCT [2]. The eye is a frequently affected organ after HSCT, and complications mostly present as dry eye disease [3,4,5,6]. More than 50% of recipients develop dry eye disease approximately 6–24 months after HSCT [7,8]. Severe GVHD-related dry eye disease can progress rapidly and lead to blindness, and even in less severe cases, it has a significant impact on patients’ quality of life and quality of vision because humans can obtain 70–80% of their sensory information from vision. Previous acute GVHD, disparity of HLA or MHC or minor histocompatibility antigens, aged donor or recipient, the presence of diabetes, stem cell source of transplantation, female to male transplantation, and Epstein–Barr Virus seropositive donor are reported as risk factors for developing ocular GVHD [6].
The ocular surface consists of a tear film layer, the lacrimal glands, accessory glands, the cornea, conjunctiva including goblet cells [9], the nasolacrimal ducts, the meibomian glands, and possibly glands of Zeis because hair follicles are commonly targeted in skin GVHD [10]; these components maintain the homeostasis of the ocular surface and protect the eye from invading pathogens and other environmental challenges. Ocular surface immunity involves communication between local and systemic inflammatory cells and interstitial stromal cells or epithelial cells by interactions mediated by specific cell surface receptors and soluble factors [11]. The ocular surface becomes an immune-advanced site once homeostasis is dysregulated by a disturbance in the microenvironment on the ocular surface including the goblet cell, which have immunomodulatory function besides mucin production [12]. Goblet cells play important roles for ocular surface health, maintaining dendritic cells in an immature state [13], reducing the number of those cells that are reported to be linked to various inflammation pathways by changing acting antigen-presenting cells [14], and regulating inflammatory cytokines, including IL-13 [15] or interferon-γ [16]. Eye-associated lymphoid tissue (EALT) has been proposed, which is continuous at the ocular surface, including the cornea, conjunctiva, and meibomian gland and its adnexa, including the lacrimal glands (LGs) and lacrimal drainage system [11]. A breakdown of EALT homeostasis may result in dry eye disease [17]. This concept is likely to be associated with immune-mediated dry eye disease, including GVHD, Sjögren’s syndrome, and other conditions of dry eye associated with autoimmune diseases [18]. Specifically, ocular GVHD (oGVHD), representing as dry eye disease, is characterized by chronic inflammation and pathogenic fibrosis on the ocular surface, resembling ocular cicatricial pemphigoid and Stevens–Johnson syndrome [19]. In this review, altered mucosal immunity, pathogenic fibrosis, and stress-induced senescence in oGVHD are discussed. By focusing on the eye region, those findings may be linked to better understanding in other targeted organs in GVHD and other inflammatory fibrotic diseases and age-related diseases and assist the development of novel and innovative therapy in the future.

1.1. Mucosal Barrier of the Ocular Surface

The mucosal immune system includes dendritic cells, macrophages, mast cells, lymphocytes, fibroblasts, and soluble factors interacting with the surface barrier epithelium, including goblet cells and microbiota that collectively contribute to immune surveillance [20]. The conjunctiva has several defense systems that protect the integrity of the surface epithelium and associated mucin layer, including antimicrobial peptides and secreted IgA, which adhere to the mucin layer. The role of IgA has been elucidated in some detail. IgA-producing plasma cells reside in the subepithelial stroma of the lacrimal glands and in the ocular surface mucosal membrane. Secreted IgA dimers bind the IgA receptor on the basement membrane of the basal epithelium and are taken up, transported intracellularly, and thereafter excreted into the tear film by the ocular surface epithelium along with other secretory components [21]. When ocular immune homeostasis is disrupted, the integrity of the ocular surface can break down, resulting in inflammatory mucosal disease and dry eye disease [17]. In ocular GVHD, basement membranes of conjunctival and lacrimal gland epithelia are disrupted through being targeted by T cells [22,23], resulting in an altered balance of production of IgA or the secretion of secretory IgA on the ocular surface.

Tear Film in GVHD-Related Dry Eye Disease

Tear fluid consists of a unique structure containing lipids, water, mucins, a variety of proteins, and electrolytes [24]; it plays a crucial role in visual function, lubrication, protection from invading foreign bodies and pathogens, washing out hazardous materials, and supplying moisture, nutrition, and oxygen on the ocular surface [24]. It is a mysterious fluid found only on the ocular surface. Certain tear cytokines have been proposed as possible biomarkers for ocular GVHD due to elevated interferon-γ, intercellular adhesion molecule-1, interleukin (IL)-6, IL-8, IL-1β, IL-10, IL-17, CXCL-10, tumor necrosis factor (TNF)-α, soluble TNF -α receptor 1, and neutrophil elastase, [25,26,27,28,29,30], MMP-9,VEGF [27], extra DNA [31] and anti-citrullinated protein autoantibodies [32] in the tear film. The level of neutrophil elastase is reported to be correlated with the elevation of myeloperoxidase, MMP8 and MMP9, suggesting the substantial role of neutrophils in tear film in ocular GVHD [33]. Those findings suggest that doxycycline and tissue inhibitors of metalloproteinases (TIMPs) are potentially reducing neutrophils and MMPs and are useful as therapeutic targets in ocular GVHD. It has been suggested that IL-8/CXCL8 and IP-10/CXCL10 have optimal sensitivity and specificity for indicating ocular GVHD [34]. Proteome analyses revealed elevated cytoskeleton protein and nuclear acid binding protein [35]. Decreased levels of IL-7, IP-10, EGF [28], lactoferrin [36], lactotransferrin, transfer and receptor proteins, enzyme modulators, and hydrolases [35] in ocular GVHD patients are reported, suggesting the loss of those regulatory factors for defending pathogens or foreign bodies from the outside world in the ocular surface in patients with cGVHD. Further elucidation of the function and cellular origin producing each soluble mediator is necessary for understanding the underlying mechanism of pathophysiological processes and improving the ocular surface condition in GVHD-related dry eye disease.

1.2. Microbiota

The commensal microbiota coevolves with the host in a symbiotic relationship on the ocular surface [37,38]. The host microbiota possesses multiple abilities to activate immunity and contribute to the deterioration of ocular surface tissue related to infections and immune-mediated diseases including GVHD [39,40]. HSCT patients treated with irradiation, immunosuppressive reagents, and/or corticosteroid therapies may lead to immune-compromised conditions in the mucosal immune system. Those factors may lead to altered and pathogenic changes of the microorganisms on the ocular surface. Several reports on the ocular surface show significantly diversified microbiota in GVHD and Stevens–Johnson syndrome using conventional cultures [37] and next-generation sequencing methods [40,41], and they also show characteristic elevation of Lactobacillus and α-diversity in ocular GVHD [41]. In contrast, several studies show dysbiosis in several immune-mediated disorders including Sjogren’s syndrome using 16S ribosomal RNA gene sequencing [39]. There is a close relationship between systemic GVHD and the gut microbiome showing loss of diversity in the intestinal microbiota of HSCT recipients.
In ocular GVHD, the initial site of ocular GVHD might be conjunctival epithelium including goblet cells [12,13,14,15,16], main excretory ducts of conjunctiva, lacrimal glands, and meibomian glands [7,9,22,42,43,44,45,46,47,48,49]. An infiltration of various inflammatory cells in the epithelial region of conjunctiva and main exocrine ducts with goblet cells and capillaries primarily via interactions with T cells, macrophages, and fibroblasts is observed in patients with cGVHD.
An obviously reduced number of goblet cells in ocular GVHD [9,49,50] is related to dysregulated immune function, leading to severe and irreversible damage in ocular GVHD based on the previous reports [12,13,14,15,16,22,48]. The lumen of lacrimal gland and meibomian gland ducts as well as ocular surface including cornea and conjunctiva faces the outside world, and commensal microbiota exist on the mucosal membrane of the ducts. These findings suggest that the interaction of mibrobiota and the mucosal immunity on the ocular surface of the duct lumen is potentially triggering the ocular GVHD. Further elucidation will be needed regarding the relation between the decreased diversity of microbiota in the gastrointestinal tract and elevated diversity of ocular surface in ocular GVHD.

2. Ocular Surface Inflammation

2.1. Early Phase of Inflammatory Response

GVHD is triggered by the interaction of migrating T cells and antigen-presenting cells [51]. Th1 and Th 17 differentiation with decreased regulatory T cells amplifies APC activation, resulting in a cytokine storm. Intensive chemotherapy or irradiation, as a preparative regimen before HSCT, may affect the ocular surface barrier of the conjunctival epithelium and facilitate the leakage of altered microbial metabolites and damage-associated molecules into the subconjunctival stroma and peripheral blood [1]. The disruption of conjunctival mucosal barrier and damage is associated with ocular GVHD, as shown in other mucosal membrane in target organs of GVHD [52]. In ocular GVHD, pseudomembranes of tarsal conjunctiva are frequently observed [53,54,55]. The histopathology of the pseudomembrane is reported as an infiltration of mature T cells from the donor graft to the recipient’s conjunctival epithelia. The pseudomembrane consists of damaged epithelia with a large number of macrophages [53,54,55,56,57,58,59]. Macrophages infiltrate as scavengers to phagocyte the degenerated epithelia as a damage-associated molecular pattern. Plenty of macrophages, CD8+ T cells, CD4+ T cells, and fibroblastic cells are reported to concomitantly infiltrate into the degenerated epithelia, suggesting an overlap syndrome of acute and chronic GVHD in the ocular region [55,56], leading to cytokine storm [51,60], which leads to corneal ulcer [55]. Pathological fibrosis presumably developed by chemokines such as stromal-derived factor-1 stimulates the chemotaxis of the fibroblasts.
Studies have shown that type 2 innate lymphoid cells (ILC2s) recruit myeloid-derived suppressor cells that suppress T cell-mediated GVHD and ILC3s secreting IL-22, enhancing intestinal stem cell function, promoting repair and the release of adenosine and suppressing T cell proliferation [61,62,63,64,65]. It is likely that an altered or reduced number of ILCs play some role under allogeneic circumstances of the ocular surface of dry eye associated with cGVHD. Further studies are required to clarify those issues.
The development of ocular GVHD is a multistep and multifactorial process. (1) A variety of inflammatory cells, including macrophages, dendritic cells, natural killer cells, in innate immunity and T cells, and B cells in adaptive immunity; (2) conjunctival, lacrimal gland, and meibomian gland epithelial cells, including conjunctival goblet cells; and (3) mesenchymal cells all play pivotal roles in the development of ocular GVHD in mice and humans [23,45,49,66,67,68,69,70].
Infiltrated dendritic cells and globular cells increasingly surround the corneal nerves in ocular GVHD recipients compared to non-GVHD recipients [71], while another study showed that those findings are similar in both groups [72]. Confocal microscopic findings can detect morphology and not be able to identify the exact cell type. However, there are two types of cells in the images of corneal confocal microscopy, and dendritic cells might be Langerhans cells and globular cells might be lymphocytes, both of which contribute to the ocular surface inflammation. The Toll-like receptor 2 (TLR2)-nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling pathway in patients with dry eye disease associated with cGVHD has been shown to be significantly activated, suggesting that TLR2 has a significant role in the inflammatory milieu of cGVHD-related dry eye disease [73].
In the conjunctiva, squamous metaplasia, decreased mucin production due to decreased goblet cells [50], decreased secretory vesicles, decreased membrane spanning mucins [49], thinning of conjunctival epithelia, and altered epithelial cell morphology have been reported [23] in patients with cGVHD dry eye disease. Under ultrastructural observation, distorted or branched microvilli of conjunctival epithelia and loss of secretion for membrane-spanning mucin can be observed and may lead to severe dry eye disease in mice [45] and humans [49]. Macrophages and T cells including CD4+ and CD8+ T cells are predominantly infiltrated in the cornea and conjunctiva of animal models of GVHD-related dry eye disease [45,66] and conjunctiva and lacrimal gland of human patients [48,68]. An increase in Th 17 cells and decrease in regulatory T cells by IL-6 production through interactions between T cells and mesenchymal stem cells in the lacrimal gland and conjunctiva may lead to dry eye disease progression in an animal model of GVHD [45,69].
These inflammatory cytokines further amplify pro-inflammatory cytokine and chemokine production and recruit other inflammatory cells, including altered macrophages, mesenchymal cells, including mesenchymal stem cells, fibroblasts, and endothelia or T cells and B cells, which collectively activate the expression of adhesion molecules and conjunctival vascular endothelial molecules [68,69]; these molecules further facilitate the recruitment of inflammatory cells to the ocular surface, thus generating an inflammatory microenvironment [11]. Chronic irritation such as tarsal conjunctival fibrosis or trichiasis with microbial pathogenic change in an immune-compromised host may develop under the influence of one or several triggers, causing chronic inflammation, which can lead to accelerate cGVHD-related dry eye disease (Figure 1).

2.2. Chronic Inflammation

Chronic immune inflammation of the eye includes the acquisition and processing of antigens by ocular antigen-presenting cells, which migrate to the draining lymph nodes through conjunctival afferent lymphatics and veins and then prime naïve CD4+ T cells and induce Th1 and Th17 polarization [74]. Then, primed CD4+ T cells migrate into the conjunctiva and lacrimal gland, where they attach to the activated vascular endothelium and migrate into the targeted microenvironment. Cytokines such as IFN-α and IL-17, which are produced by activated T cells, amplify the immune response by increasing the expression of adhesion molecules including VCAM, VEGF-C, and VEGF-D on conjunctival blood vessels, which leads to ocular surface damage [11,75]. Effector memory Th17 cells contribute to maintain the chronic and relapsing course of dry eye disease [76]. IFN-γ also alters mucins on the ocular surface; these pathogenic changes are implicated in epithelial cell apoptosis, goblet cell loss, and squamous dysplasia [77]. IL-17 increases MMP3 and MMP9 expression and corneal epithelial barrier dysfunction [75]. At the site of corneal perforation, macrophage or CD8+ T cell accumulation and MMP9 elevation are observed in GVHD patients [78,79].
Although these changes do not always occur in all stages of cGVHD-related dry eye disease, immune-mediated inflammation, including the dysregulated activation of innate and adaptive immunity, is a pivotal factor in developing and perpetuating this disease.

2.3. Sterile Inflammation

Sterile inflammation, which can be triggered by a wide variety of stimuli, including altered cellular and tissue states or debris, is reported to be associated with GVHD [80]. A body of evidence indicates that extracellular DNA (eDNA), a damage-associated molecular pattern, may be related to cGVHD-related dry eye disease [31,81]. DNase levels are reported to be extremely reduced in the tear film of cGVHD patients [82]. Therefore, eDNA accumulates on the ocular surface, resulting in the severe form of dry eye disease in cGVHD. In addition to macrophage, natural killer cell, T cell, and B cell infiltration, danger signals such as eDNA are reported to be related to the severity and perpetuation of dry eye disease. eDNA is released at the ocular surface by superficial dying conjunctival or corneal epithelial cells, probably resulting in filamentosa on the cornea that leads to severe pain and discomfort in ocular GVHD patients. In patients of cGVHD-related dry eye disease, cell debris in pro-inflammatory state are accumulated in lacrimal gland stroma in humans [83]. With multiple functions in both the innate and adaptive immune systems, autophagy has been demonstrated to take part in the pathogenesis of several immune-related diseases. The study of the pathological mechanism of autophagy may provide a targeted intervention directed against the therapeutic strategy of immune diseases including ocular GVHD.

3. Immune-Mediated Fibrosis

Fibrosis is the extensive deposition of fibrous connective tissue, and it is characterized by the accumulation of collagen and other extracellular matrix (ECM) components [84]. Fibrotic remodeling in various diseases, such as liver cirrhosis, pulmonary fibrosis, systemic sclerosis, and GVHD, can lead to organ dysfunction, causing high morbidity and mortality. Both innate immunity and adaptive immunity are involved in fibrogenesis in ocular GVHD [22,84,85]. Published data support the roles of profibrogenic molecules, including IL-4, IL-6, IL-17, heat shock protein 47, connective tissue growth factor, and transforming growth factor (TGF)-β, in fibroblast activation in ocular cicatricial pemphigoid [86,87,88] that presents similar fibrotic conditions of ocular GVHD. Novel therapeutic options based on inhibiting these pathways will be required.

3.1. Bone Marrow-Derived Cells in Pathogenic Fibrosis

Stromal mesenchymal cells, such as fibroblasts, are classically thought to produce collagens to simply support various organs, but a body of evidence indicates that these mesenchymal cells play some role in immunosurveillance or act as immune cells themselves, functioning as antigen-presenting cells. For example, fibrocytes are reported to act as potent antigen presenting cells [89,90].
CD34+ fibroblasts are reported to reside in the mammary glands, submandibular glands, and thyroid, probably acting in immune surveillance [91,92,93], and they play a pathological role in the lacrimal glands and conjunctiva in cGVHD-related dry eye disease [22,67,69]. Fibroblasts or mesenchymal cells in the stroma may play a role in maintaining the homeostasis of the ocular surface, including the lacrimal gland microenvironment, under healthy conditions, and they act as a regenerative or pathogenic trigger of inflamed tissue [94]. Mesenchymal stromal cells have been shown to be sensors and switches in inflammation [94]. In cGVHD-related dry eye disease, freshly isolated fibroblasts/fibrocytes may be partially derived from the bone marrow and activated to trigger immune-mediated fibrosis in cGVHD by interacting with immune cells, including macrophages, T cells, and B cells [67,69]. The fibrotic process is apparently accelerated, and the resultant excessive fibrosis leads to functional dysfunction in target organs, including the lacrimal glands and conjunctiva [67]. It is likely that the prolonged fibrotic environment in chronic inflammatory lacrimal gland and conjunctiva promotes the recruitment and mobilization of donor-derived fibroblast precursors into inflammatory milieu, which may have unlimited growth potential or may be constantly migrated from circulation in chronic GVHD [67].
In contrast, recent evidence has shown that cultured mesenchymal stem cells (MSCs) are useful for modulating and suppressing GVHD and have been used to treat steroid-refractory cGVHD and regenerate or suppress ocular GVHD in animal models [95,96]. This controversy is due to differences in the use of cultured mesenchymal stem cells or freshly isolated mesenchymal cells, the species of origin, the preparation methods, or the origin of the mesenchymal stem cells used. It may be useful to revisit freshly isolated mesenchymal stem cells whether they are linked to pathogenetic properties in GVHD-related dry eye disease [69]. An evaluation of prospective transplantation of freshly purified bone marrow mesenchymal/stromal cells (BMSCs) and hematopoietic stem cells into BALB/c-RAG2KO mice suggested that transplanted minor antigen-mismatched MHC-compatible BMSCs interact with residual host T cells to induce the autoimmune phenotype observed in fibrosis associated with the sclerodermatous mouse model [69]. While the driver antigen remains to be elucidated, this study suggests that the accidental recognition of self-minor antigens on MHC class II+ bone marrow stromal cells may be involved in pathogenic fibrosis observed in autoimmune diseases, including ocular GVHD [22,67,69].

3.2. Epithelial–Mesenchymal Transition (EMT)

Among the fibrotic processes that could account for cGVHD-related dry eye disease, EMT is a possible candidate for the process because the ocular surface epithelium loses the capability to secrete mucins when the epithelial cells undergo the transition to a mesenchymal phenotype [23]. Several studies have shown that EMT contributes to various fibrotic diseases of the kidneys, lungs, and liver and the ocular surface epithelium [97,98,99] and retinal pigment epithelium [100] in the eye. For example, 40% of fibroblasts in kidney fibrosis arise from epithelial cells via local EMT triggered by inflammatory stress, and 15% of fibroblasts are derived from the bone marrow under inflammatory stress [101]. EMT is characterized by the loss of apical/basal cell polarity and loss of cell-to-cell adhesions, which is followed by the acquisition of a mesenchymal phenotype that promotes migration and invasion capabilities and the expression of mesenchymal markers. EMT is triggered by various stimuli, including irradiation [102]; hypoxia [103]; reactive oxygen species [104]; inflammatory cytokines, such as TGF-β and fibroblast growth factor [105]; disruption of the basal lamina; and exposure of the cytoplasm to the extracellular matrix [106]. These triggers of EMT also cooperate with the pathogenesis of cGVHD after HSCT. Total body irradiation and migrating inflammatory cells generate substantial proinflammatory cytokines [3]. The “cytokine storm” influences T cells in the recipient microenvironment, prompting them to attack host antigens. In addition, reactive oxygen species-mediated lacrimal gland injury has been reported in the bone marrow transplant [107] and other target organs [108] in mice as possible triggers for EMT.
Notably, the rearrangement of cytoskeletal actin filaments is necessary for EMT, but an intact cytoskeleton is required to guide secretary vesicles to the ocular surface and to generate microvilli. Since cGVHD may trigger EMT in the ocular epithelium, the conjunctival microvilli may be abnormal and unable to secrete membrane-spanning mucins [49]. In ocular cGVHD, the production of both gel-forming mucins including MUC5AC and membrane-spanning mucin including MUC1, MUC4, and MUC16 is highly reduced [49]. It is likely that cytotoxic T cells cause the basement membrane of the conjunctival and lacrimal gland epithelium to break down [22,23,49], allowing direct interactions between the cytoskeleton of epithelial cells and stromal extracellular matrix components [106], which may trigger EMT in HSCT recipients.

4. Cellular Senescence in Ocular GVHD

Aging is commonly defined as the accumulation of diverse deleterious changes in cells and tissues with advancing age that are responsible for increased risks of disease [109]. In addition, cellular senescence involves genomic instability, telomere loss, oxidative damage, genetic programming, and cell death [109]. The activation of reactive oxygen species [107], renin angiotensin system [110], vascular adhesion protein-1 [111], and endoplasmic reticulum stress [112] together may contribute to stress-induced senescence in ocular GVHD. The pathogenesis of ocular GVHD and stress-induced cellular senescence are potentially associated through the senescence-associated secretory phenotype (SASP). Senescent cells produce cytokines and chemokines in terms of SASP [113], such as IL-6 and CXCL9 in GVHD lacrimal gland in mice [83] (Figure 2).

4.1. Oxidative Stress

Oxidative damage of immune cells may be the earliest sign of immune aging. Recently, macrophages were reported to be promoters of age-related diseases such as macular degeneration [114]. Other studies have shown that oxidative stress in mitochondria can activate ROS, leading to inflammasome activation and the production of pro-inflammatory cytokines [115], resulting in inflammation and age-related findings. In previous reports on immune-mediated dry eye disease in animal models, mitochondrial alterations were found to be central to the disease process [116,117]. The mitochondria contribute to the process of inflammation, and pro-inflammatory soluble factors apparently alter mitochondrial function to amplify their effects. The elevation of mitochondrial oxidative stress results in a vicious inflammatory cycle that exacerbates immune-mediated dry eye disease [118,119]. ROS are elevated in macrophages infiltrated in the lacrimal gland affected by cGVHD in a sclerodermatous mouse model [107]. Oxidative stress markers, including 8-hydroxy-2′-deoxyguanosine (8-OHdG), 4-hydroxy-2-nonenal (4-HNE), and hexonoyl lesion (HEL), and other aging markers, including p16 and p38, were detected in the infiltrating mononuclear cells, mainly the macrophages and endothelium in capillaries in cGVHD and aged mice but not in a syngeneic model or young mice [107].

4.2. Tissue Renin–Angiotensin System (RAS)

The RAS was originally reported to be a blood pressure regulator. Besides systemic RAS, local tissue RAS activation is involved in the innate and adaptive immune systems and fibrosis. Yaguchi, S et al. proved that an angiotensin II (Ang II) type 1 receptor antagonist attenuate lacrimal gland, lung, and liver fibrosis in a murine model of cGVHD [110]. Ang II is reported to be a pro-inflammatory molecule and possible profibrotic molecule that affects progressive damage to the lacrimal glands and ocular surface homeostasis in ocular GVHD. It is reported that the binding of Ang II to the angiotensin type I receptor generates an intracellular free radical that plays a crucial role in tissue damage by accelerating mitochondrial dysfunction [120]. Those findings suggest that tissue RAS is related to stress-induced senescence in ocular GVHD. Blocking Ang II signaling protects against neurodegenerative processes and promotes longevity in an animal model, suggesting targeting Ang II signaling as a therapeutic strategy in inflammatory diseases, including GVHD-related dry eye disease and aging-related changes [121].

4.3. Endoplasmic Reticulum (ER) Stress

Cellular senescence has been reported to be detrimentally involved in ocular cGVHD, and cGVHD can be regarded as an age-associated disease [107]. Telomere shortening in donor hematopoietic stem cells (HSCs) after HSCT [122] suggests that donor immune cells as well as engrafted recipient cells are more senescent in cGVHD under an inflammatory milieu. Previous studies indicate that ER stress plays a role in chronic inflammation and age-related diseases [123,124]. The ER is a cellular organelle essential for proper cell function. When proteins are synthesized in the ER, they need to be folded correctly, and ER chaperones assist in protein folding [125]. However, hypoxia, calcium ion depletion, oxidative injury, viral infections, and inflammatory cytokines prevent the ER from performing its role in normal protein folding [126]. When unfolded and misfolded proteins accumulate in the ER, they cause ER stress, and the following three transmembrane proteins are consequently activated to initiate the unfolded protein response (UPR): inositol requiring (IRE) 1α, PKR-like ER kinase (PERK), and activating transcription factor (ATF) 6α [125]. However, if the UPR is prolonged or unsuccessful, inflammatory and apoptotic pathways are activated [125]. Therefore, an impaired UPR l results in the expression of (1) the pro-inflammatory molecule thioredoxin interaction protein (TXNIP) and transcription factor NF-κB and (2) the apoptotic protein C/EBP homologous protein (CHOP) [112,127].
Mukai S, et al. revealed that the ER stress is elevated in target organs of GVHD including the lacrimal gland and conjunctiva, and ER stress suppressor as well as anti-aging medicine, 4-phenyl butyric acid, can effectively reduce GVHD-related inflammation and fibrosis in targeted organs [112]. Those findings suggest that the pathogenic process of GVHD has aspects of stress-induced senescence and is linked to the underlying mechanism of the pathogenic process of ocular GVHD. Inhibition of the ER stress pathway may be a therapeutic target for cGVHD-related dry eye disease.

4.4. Vascular Adhesion Protein-1

With respect to the aberrant accumulation of inflammatory cells, the overexpression of vascular adhesion protein-1 (VAP-1) has been reported to be implicated in inflammatory diseases [128]. VAP-1 is an endothelial surface glycoprotein that mediates the migration of leukocytes from the bloodstream to tissues [129]. VAP-1 has the following two crucial portions that assist leukocytes in migrating to tissues: a distal adhesion domain and a catalytic amine oxidase region [130]. When immune cells infiltrate sites of inflammation, an amino group in each leukocyte attacks the carbonyl group in VAP-1 [130]. The primary amine is subsequently converted to the corresponding aldehyde, and this catalytic transformation enables inflammatory cells to extravasate into tissues through the blood vessel. Aldehyde is cytotoxic and related to oxidative stress, which plays a role in stress-induced senescence in some way in ocular GVHD. Based on these reports, Mukai S, et al. studied the following: (1) the prevention of immune cell migration to tissues alleviates acute GVHD (aGVHD) and cGVHD, (2) the increased expression of VAP-1 is involved in the pathogenesis of both types of GVHD, and (3) the inhibition of VAP-1 could be an efficacious treatment for the two types of GVHD in a mouse model. The protection of goblet cells through the injection of a VAP-1 inhibitor effectively improves the number of goblet cells in the intestine and conjunctiva in comparison with vehicle-treated GVHD mice.

4.5. Stress-Induced Senescence in Ocular GVHD

Yamane M, et al. [83] showed that the senescent-associated secretory phenotype (SASP) of macrophages, T cells, and fibroblast from lacrimal gland in GVHD mice is elevated and plays an indispensable role for chronic inflammation and fibrosis in ocular GVHD. The authors hypothesized that several stressors arrest the cell cycle, and macrophages exposed to severe damage undergo senescence and produce the SASP factors IL-6 and CXCL9 upon interaction with CD4+osteopontin (OPN)+CD153+ senescent T cells. Then, IL-6, a major driver of the SASP, reinforces macrophage senescence in an autocrine manner. In addition, CXCL9 facilitates the recruitment of neighboring T cells to the microenvironment early after onset [131]. T cell senescence after HSCT was recently reported in an acute GVHD animal model. CD4+OPN+CD153+ senescent T cells may accelerate stress-induced senescence as a late complication of cGVHD. Moreover, IL-6 may induce senescence in a subpopulation of fibroblasts. In addition, OPN, another SASP factor, promotes EMT in cGVHD lacrimal glands. As a result, a subset of senescent/mature fibroblasts synthesize abnormal collagens. Fibroblasts affected by macrophages and T cells also synthesize excessive abnormal collagens and extracellular matrix components [15,70], leading to inflammation and abnormal fibrosis in cGVHD lacrimal glands. Collectively, senescent cells with the SASP or cells with stress-induced senescent features contribute to the pathogenesis of cGVHD in the lacrimal glands, although immunological inflammation is the major underlying mechanism. Inhibiting the SASP induced by senescent cells, using senolytic reagents may be a new clinically translatable strategy for attenuating the effects of cGVHD on the lacrimal glands and other target organs.

5. Treatment

According to the basic mechanism of chronic ocular GVHD, an anti-inflammatory, anti-fibrotic, and anti-aging approach will be required to add or radical treatment in addition to the common approaches including moisture, lubricant, epithelial support, and surgical treatments (Table 1) [3,4,6,45,132,133,134,135].
A specific prophylaxis and/or treatment of ocular GVHD may be utilized including decontamination such as doxycycline [6,143,144,165,166,167]. Pooled human immunoglobulin eye drops have been reported to be effective for ocular GVHD [32]. It can be also used for prophylaxis and/or the treatment of ocular GVHD. At the basic study stage, gentamicin is a possible candidate for the prophylaxis of ocular GVHD [145].
The recommended treatments of ocular GVHD in the inflammatory phase are as follows. Topical preservative-free corticosteroids [143], rebamipide, cyclosporine A [143], and tacrolimus [144,146] for ocular surface inflammation have been shown to be effective. Low-dose topical corticosteroid is shown to have reduced efficacy on ocular GVHD [168]. One of the biological agents, lifitegrast (Xiidra), a topical integrin antagonist, was well-tolerated and led to an improvement in symptoms of KCS in eight (44%) patients [169]. Due to multifactorial aspects of this disease, careful follow up on unexpected events is required [170]. The other biological agent, Anakinra (IL-1Ra antagonist), by suppressing IL-1-mediated inflammation by competitively inhibiting the binding of IL-1α and IL-1β to IL-1 receptor I, may be beneficial as a therapeutic option for patients with dry eye disease [171], and it might have a beneficial effect on ocular GVHD. Tranilast is N-(30, 40-dimethoxycinnamoyl)-anthranilic acid and an analog of a tryptophan metabolite [172]. It has an inhibition of the inflammatory effect by suppressing the recruitment of T cells by the inhibition of CXCL9 expression, an anti-oxidative stress effect through the inhibition of thiorexin-interacting protein (TXNIP) [173], and an anti-fibrotic effect by inhibition of fibroblast activation. Topical tranilast has been reported in a small number of cGVHD-related dry eye patients [174].
As epithelial support, autologous serum has been increased to be used as eye drops for ocular GVHD because it contains the vitamins, several growth factors, and fibronectin that are crucial for ocular surface integrity [134,148,149,175,176]. Cord blood serum [150,151], or platelet lysate eye drops [152,153] also have been reported to show benefits for ocular GVHD patients. Recently, the detection of systemic immunosuppressants in serum has been reported and benefits from using a topical serum eye drop on ocular GVHD [177].
Surgical and other treatments are shown in Table 1 and details are described in cited references.
As a consequence of the ocular condition during systemic therapy, the average response rates of ocular GVHD are 43% for extracorporeal photopheresis [178,179], 31% for rituximab (anti-CD20 antibody), 60% for sirolimus, and 33% for mycophenolate mofetil in corticosteroid-refractory GVHD [6].
It has been shown that IL-6 is elevated early in the course of ocular GVHD in animal models [69,83] and humans [25,26,28,83,180,181]. Systemic tocilizumab [182,183] and sarilumab for rheumatoid arthritis [184] that impact the IL-6 pathway may be beneficial to treat or prevent ocular GVHD. Other biologics including janus kinase (JAK) combined with spleen tyrosine kinase (SYK) have been reported to be well tolerated, suggesting efficacy for ocular GVHD patients as a pilot study [185]. It will be benefit the severe ocular GVHD patients if the biological reagents used both systemic and topical eye drops or ointments as well.
In basic research, ATR type I antagonist [110], VAP-1 inhibitor [111], phenyl butyric acid [112], tranilast [186], heavy chain-hyaluronan/pentraxin 3 (HC-HA/PTX3) [187], ABT-263, a senolytic agent that removes senescent cells, an anti-IL-6 blocking antibody for SASP inhibition [83], and vitamin A-coupled liposomes containing HSP4 siRNA reversed the changes seen in ocular GVHD [162]. Entospletinib (ENTO), small molecule inhibitors of lymphocyte signaling, and a second-generation highly selective SYK inhibitor [147] have been reported to reduce inflammation and its by-product and/or fibrosis in ocular as wells systemic GVHD mice. Pooled human immunoglobulin eye drops have potential as a new candidate in treating ocular GVHD [32]. Creating innovative therapies for ocular GVHD is necessary to treat this intractable ocular disease. Ocular GVHD must be recognized and treated early in the course to prevent the irreversible progression and damage of ocular tissue using these strategies. If the ocular GVHD is found at a later stage, those therapies should be started as early as possible to slow or prevent the further development of this disease.

6. Future Directions

Future therapeutic goals include regeneration of the dysfunctional ocular surface, lacrimal glands and meibomian glands. Broad-based translational research that extends from bench to bedside and makes use of clinical results (bedside to bench) will augment current analyses of the mechanisms underlying dry eye disease and will lead to the development of new anti-inflammatory, anti-fibrotic, and anti-senescent approaches or other specific interventions based on these mechanisms.

7. Patents

Y.O. has a patent in Japan (Patent No. 4966019; Name; Topical application and oral intake of tranilast for the treatment of chronic GVHD-related dry eye disease) and Y.O and K.T. have patent application number JP 2017-018643 published as JPA2017-178922, application number JP2018-510646 published as WO2017/175808, and application number JP 2019-004730 published as JPA2020-111548. Y.K. reports no conflicts of interest related to this study.

Author Contributions

Y.O. Conceptualization, methodology, software, validation, formal analysis, investigation, resources, data curation, writing—original draft preparation, writing; Y.O. Review and editing, visualization, supervision, project administration: Y.O., K.T., Y.K. and Funding acquisition: Y.O. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by grants from the Japanese Ministry of Education, Science, Sports, Culture and Technology (26462668, 18K09421, and 21K09572)A grant from AMO Japan KK, 2021. to Y.O.

Institutional Review Board Statement

For the study at Keio University, All experimental procedures were in accordance with the Institutional Guidelines on Animal Experimentation at Keio University and were approved by the Keio University Institutional Animal Care and Use Committee (# 09152). For human study at Keio University, Institutional Ethical Committee approved this retrospective study (#20170350).

Acknowledgments

The author thanks Shigeto Shimmura, Masataka Kuwana, Masataka Kawai, Saori Yaguchi, Mio Yamane, Masaki Fukui, He Jingliang, Yang Fan, Eisuke Shimizu, Shinri Sato, Kazuki Asai and Robert Rusch for continuous support of this work.

Conflicts of Interest

The authors have a patent and is applying the patents as described above.

References

  1. Ferrara, J.L.; E Levine, J.; Reddy, P.; Holler, E. Graft-versus-host disease. Lancet 2009, 373, 1550–1561. [Google Scholar] [CrossRef]
  2. Jagasia, M.H. National Institutes of Health Consensus Development Project on Criteria for Clinical Trials in Chronic Graft-versus-Host Disease: I. The 2014 Diagnosis and Staging Working Group report. Biol. Blood Marrow Transplant. J. Am. Soc. Blood Marrow Transplant. 2015, 21, 389–401.e1. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Shikari, H.; Antin, J.H.; Dana, R. Ocular Graft-versus-Host Disease: A Review. Surv. Ophthalmol. 2013, 58, 233–251. [Google Scholar] [CrossRef]
  4. Giannaccare, G.; Pellegrini, M.; Bernabei, F.; Scorcia, V.; Campos, E. Ocular surface system alterations in ocular graft-versus-host disease: All the pieces of the complex puzzle. Graefe’s Arch. Clin. Exp. Ophthalmol. 2019, 257, 1341–1351. [Google Scholar] [CrossRef]
  5. Munir, S.Z.; Aylward, J. A Review of Ocular Graft-Versus-Host Disease. Optom. Vis. Sci. 2017, 94, 545–555. [Google Scholar] [CrossRef] [PubMed]
  6. Inamoto, Y.; Valdés-Sanz, N.; Ogawa, Y.; Alves, M.; Berchicci, L.; Galvin, J.; Greinix, H.; Hale, G.A.; Horn, B.; Kelly, D.; et al. Ocular graft-versus-host disease after hematopoietic cell transplantation: Expert review from the Late Effects and Quality of Life Working Committee of the CIBMTR and Transplant Complications Working Party of the EBMT. Bone Marrow Transplant. 2019, 54, 662–673. [Google Scholar] [CrossRef]
  7. Ogawa, Y.; Okamoto, S.; Wakui, M.; Watanabe, R.; Yamada, M.; Yoshino, M.; Ono, M.; Yang, H.-Y.; Mashima, Y.; Oguchi, Y.; et al. Dry eye after haematopoietic stem cell transplantation. Br. J. Ophthalmol. 1999, 83, 1125–1130. [Google Scholar] [CrossRef] [Green Version]
  8. Uchino, M.; Ogawa, Y.; Uchino, Y.; Mori, T.; Okamoto, S.; Tsubota, K. Comparison of stem cell sources in the severity of dry eye after allogeneic haematopoietic stem cell transplantation. Br. J. Ophthalmol. 2012, 96, 34–37. [Google Scholar] [CrossRef] [PubMed]
  9. Wang, Y.; Ogawa, Y.; Dogru, M.; Tatematsu, Y.; Uchino, M.; Kamoi, M.; Okada, N.; Okamoto, S.; Tsubota, K. Baseline profiles of ocular surface and tear dynamics after allogeneic hematopoietic stem cell transplantation in patients with or without chronic GVHD-related dry eye. Bone Marrow Transplant. 2010, 45, 1077–1083. [Google Scholar] [CrossRef]
  10. Villarreal, C.D.V.; Alanis, J.C.S.; Pérez, J.C.J.; Candiani, J.O. Cutaneous graft-versus-host disease after hematopoietic stem cell transplant—A review. An. Bras. Dermatol. 2016, 91, 336–343. [Google Scholar] [CrossRef]
  11. McDermott, A.M.; Perez, V.; Huang, A.J.; Pflugfelder, S.C.; Stern, M.E.; Baudouin, C.; Beuerman, R.W.; Burns, A.R.; Calder, V.L.; Calonge, M.; et al. Pathways of corneal and ocular surface inflammation: A perspective from the cullen symposium. Ocul. Surf. 2005, 3, S131–S138. [Google Scholar] [CrossRef]
  12. Barnett, B.P.; Afshari, N.A. Dupilumab-Associated Mucin Deficiency (DAMD). Transl. Vis. Sci. Technol. 2020, 9, 29. [Google Scholar] [CrossRef] [Green Version]
  13. Contreras-Ruiz, L.; Ghosh-Mitra, A.; Shatos, M.A.; Dartt, D.A.; Masli, S. Modulation of Conjunctival Goblet Cell Function by Inflammatory Cytokines. Mediat. Inflamm. 2013, 2013, 1–11. [Google Scholar] [CrossRef] [Green Version]
  14. Contreras-Ruiz, L.; Masli, S. Immunomodulatory Cross-Talk between Conjunctival Goblet Cells and Dendritic Cells. PLoS ONE 2015, 10, e0120284. [Google Scholar] [CrossRef]
  15. De Paiva, C.S.; Raince, J.K.; McCellan, A.; Shanmugam, K.P.; Pangelinan, S.B.; A Volpe, E.; Corrales, R.M.; Farley, W.J.; Corry, D.B.; Li, D.-Q.; et al. Homeostatic control of conjunctival mucosal goblet cells by NKT-derived IL-13. Mucosal Immunol. 2011, 4, 397–408. [Google Scholar] [CrossRef] [PubMed]
  16. Zhang, X.; De Paiva, C.S.; Su, Z.; Volpe, E.A.; Li, D.-Q.; Pflugfelder, S.C. Topical interferon-gamma neutralization prevents conjunctival goblet cell loss in experimental murine dry eye. Exp. Eye Res. 2014, 118, 117–124. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Knop, N.; Knop, E. Regulation of the inflammatory component in chronic dry eye disease by the eye-associated lymphoid tissue (EALT). Dev. Ophthal. 2010, 45, 23–39. [Google Scholar] [CrossRef]
  18. Steven, P.; Schwab, S.; Kiesewetter, A.; Saban, D.R.; Stern, M.E.; Gehlsen, U. Disease-Specific Expression of Conjunctiva Associated Lymphoid Tissue (CALT) in Mouse Models of Dry Eye Disease and Ocular Allergy. Int. J. Mol. Sci. 2020, 21, 7514. [Google Scholar] [CrossRef]
  19. Bron, A.J.; de Paiva, C.S.; Chauhan, S.K.; Bonini, S.; Gabison, E.E.; Jain, S.; Knop, E.; Markoulli, M.; Ogawa, Y.; Perez, V.; et al. TFOS DEWS II pathophysiology report. Ocul. Surf. 2017, 15, 438–510. [Google Scholar] [CrossRef] [PubMed]
  20. Ueta, M.; Kinoshita, S. Innate immunity of the ocular surface. Brain Res. Bull. 2010, 81, 219–228. [Google Scholar] [CrossRef]
  21. Narayanan, S.; Redfern, R.L.; Miller, W.L.; Nichols, K.K.; McDermott, A.M. Dry Eye Disease and Microbial Keratitis: Is There a Connection? Ocul. Surf. 2013, 11, 75–92. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Ogawa, Y.; Yamazaki, K.; Kuwana, M.; Mashima, Y.; Nakamura, Y.; Ishida, S.; Toda, I.; Oguchi, Y.; Tsubota, K.; Okamoto, S.; et al. A significant role of stromal fibroblasts in rapidly progressive dry eye in patients with chronic GVHD. Investig. Ophthalmol. Vis. Sci. 2001, 42, 111–119. [Google Scholar]
  23. Ogawa, Y.; Shimmura, S.; Kawakita, T.; Yoshida, S.; Kawakami, Y.; Tsubota, K. Epithelial Mesenchymal Transition in Human Ocular Chronic Graft-Versus-Host Disease. Am. J. Pathol. 2009, 175, 2372–2381. [Google Scholar] [CrossRef] [Green Version]
  24. Willcox, M.D.; Argüeso, P.; Georgiev, G.; Holopainen, J.M.; Laurie, G.W.; Millar, T.J.; Papas, E.B.; Rolland, J.P.; Schmidt, T.A.; Stahl, U.; et al. TFOS DEWS II Tear Film Report. Ocul. Surf. 2017, 15, 366–403. [Google Scholar] [CrossRef] [Green Version]
  25. Riemens, A.; Stoyanova, E.; Rothova, A.; Kuiper, J. Cytokines in tear fluid of patients with ocular graft-versus-host disease after allogeneic stem cell transplantation. Mol. Vis. 2012, 18, 797–802. [Google Scholar]
  26. Jung, J.W.; Han, S.J.; Song, M.K.; Kim, T.-I.; Kim, E.K.; Min, Y.H.; Cheong, J.-W.; Seo, K.Y. Tear Cytokines as Biomarkers for Chronic Graft-versus-Host Disease. Biol. Blood Marrow Transplant. 2015, 21, 2079–2085. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Byun, Y.-S.; Yoo, Y.-S.; Kang, M.-J.; Whang, W.-J.; Na, K.-S.; Mok, J.-W.; Joo, C.-K. Marked infiltration of neutrophils at the upper palpebral conjunctiva in patients with chronic graft-versus-host disease. Ocul. Surf. 2019, 17, 295–302. [Google Scholar] [CrossRef] [PubMed]
  28. Hu, B.; Qiu, Y.; Hong, J. Tear cytokine levels in the diagnosis and severity assessment of ocular chronic graft-versus-host disease(GVHD). Ocul. Surf. 2020, 18, 298–304. [Google Scholar] [CrossRef] [PubMed]
  29. Steven, P.; Scherer, D.; Krösser, S.; Beckert, M.; Cursiefen, C.; Kaercher, T. Semifluorinated Alkane Eye Drops for Treatment of Dry Eye Disease--A Prospective, Multicenter Noninterventional Study. J. Ocul. Pharmacol. Ther. 2015, 31, 498–503. [Google Scholar] [CrossRef] [Green Version]
  30. Sakimoto, T.; Ohnishi, T.; Ishimori, A. Significance of Ectodomain Shedding of TNF Receptor 1 in Ocular Surface. Investig. Ophthalmol. Vis. Sci. 2014, 55, 2419–2423. [Google Scholar] [CrossRef] [Green Version]
  31. Sonawane, S.; Khanolkar, V.; Namavari, A.; Chaudhary, S.; Gandhi, S.; Tibrewal, S.; Jassim, S.H.; Shaheen, B.; Hallak, J.; Horner, J.H.; et al. Ocular Surface Extracellular DNA and Nuclease Activity Imbalance: A New Paradigm for Inflammation in Dry Eye Disease. Investig. Ophthalmol. Vis. Sci. 2012, 53, 8253–8263. [Google Scholar] [CrossRef] [PubMed]
  32. Kwon, J.; Surenkhuu, B.; Raju, I.; Atassi, N.; Mun, J.; Chen, Y.-F.; Sarwar, M.A.; Rosenblatt, M.; Pradeep, A.; An, S.; et al. Pathological consequences of anti-citrullinated protein antibodies in tear fluid and therapeutic potential of pooled human immune globulin-eye drops in dry eye disease. Ocul. Surf. 2020, 18, 80–97. [Google Scholar] [CrossRef]
  33. Arafat, S.N.; Robert, M.-C.; Abud, T.; Spurr-Michaud, S.; Amparo, F.; Dohlman, C.H.; Dana, R.; Gipson, I.K. Elevated Neutrophil Elastase in Tears of Ocular Graft-Versus-Host Disease Patients. Am. J. Ophthalmol. 2017, 176, 46–52. [Google Scholar] [CrossRef]
  34. Cocho, L.; Fernandez, I.; Calonge, M.; Martínez, V.; González-García, M.J.; Caballero, L.; López-Corral, L.; García-Vázquez, C.; Vazquez, L.; Stern, M.E.; et al. Biomarkers in Ocular Chronic Graft Versus Host Disease: Tear Cytokine- and Chemokine-Based Predictive Model. Investig. Ophthalmol. Vis. Sci. 2016, 57, 746. [Google Scholar] [CrossRef] [Green Version]
  35. Gerber-Hollbach, N.; Plattner, K.; O’Leary, O.E.; Jenoe, P.; Moes, S.; Drexler, B.; Schoetzau, A.; Halter, J.P.; Goldblum, D. Tear Film Proteomics Reveal Important Differences Between Patients With and Without Ocular GvHD After Allogeneic Hematopoietic Cell Transplantation. Investig. Ophthalmol. Vis. Sci. 2018, 59, 3521–3530. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Sonobe, H.; Ogawa, Y.; Yamada, K.; Shimizu, E.; Uchino, Y.; Kamoi, M.; Saijo, Y.; Yamane, M.; Citterio, D.; Suzuki, K.; et al. A novel and innovative paper-based analytical device for assessing tear lactoferrin of dry eye patients. Ocul. Surf. 2019, 17, 160–166. [Google Scholar] [CrossRef]
  37. Shimizu, E.; Ogawa, Y.; Saijo, Y.; Yamane, M.; Uchino, M.; Kamoi, M.; Fukui, M.; Yang, F.; He, J.; Mukai, S.; et al. Commensal microflora in human conjunctiva; characteristics of microflora in the patients with chronic ocular graft-versus-host disease. Ocul. Surf. 2019, 17, 265–271. [Google Scholar] [CrossRef] [PubMed]
  38. Miller, D.; Iovieno, A. The role of microbial flora on the ocular surface. Curr. Opin. Allergy Clin. Immunol. 2009, 9, 466–470. [Google Scholar] [CrossRef] [PubMed]
  39. De Paiva, C.S.; Jones, D.B.; Stern, M.E.; Bian, F.; Moore, Q.L.; Corbiere, S.; Pflugfelder, S.C. Altered Mucosal Microbiome Diversity and Disease Severity in Sjögren Syndrome. Sci. Rep. 2016, 6, 23561. [Google Scholar] [CrossRef]
  40. Kittipibul, T.; Puangsricharern, V.; Chatsuwan, T. Comparison of the ocular microbiome between chronic Stevens-Johnson syndrome patients and healthy subjects. Sci. Rep. 2020, 10, 1–9. [Google Scholar] [CrossRef] [Green Version]
  41. Zilliox, M.J.; Gange, W.S.; Kuffel, G.; Mores, C.R.; Joyce, C.; de Bustros, P.; Bouchard, C.S. Assessing the ocular surface microbiome in severe ocular surface diseases. Ocul. Surf. 2020, 18, 706–712. [Google Scholar] [CrossRef]
  42. Ban, Y.; Ogawa, Y.; Ibrahim, O.M.; Tatematsu, Y.; Kamoi, M.; Uchino, M.; Yaguchi, S.; Dogru, M.; Tsubota, K. Morphologic evaluation of meibomian glands in chronic graft-versus-host disease using in vivo laser confocal microscopy. Mol. Vis. 2011, 17, 2533–2543. [Google Scholar] [PubMed]
  43. Dikmetas, O.; Kocabeyoglu, S.; Mocan, M.C. The Association between Meibomian Gland Atrophy and Corneal Subbasal Nerve Loss in Patients with Chronic Ocular Graft-versus-host Disease. Curr. Eye Res. 2021, 46, 1–6. [Google Scholar] [CrossRef] [PubMed]
  44. Engel, L.A.; Wittig, S.; Bock, F.; Sauerbier, L.; Scheid, C.; Holtick, U.; Chemnitz, J.-M.; Hallek, M.; Cursiefen, C.; Steven, P. Meibography and meibomian gland measurements in ocular graft-versus-host disease. Bone Marrow Transplant. 2015, 50, 961–967. [Google Scholar] [CrossRef] [Green Version]
  45. He, J.; Yamane, M.; Shibata, S.; Fukui, M.; Shimizu, E.; Yano, T.; Mukai, S.; Kawakami, Y.; Li, S.; Tsubota, K.; et al. Ocular Surface and Tear Film Characteristics in a Sclerodermatous Chronic Graft-Versus-Host Disease Mouse Model. Cornea 2018, 37, 486–494. [Google Scholar] [CrossRef] [PubMed]
  46. Hwang, H.S.; Ha, M.; Kim, H.-S.; Na, K.-S. Longitudinal analysis of meibomian gland dropout in patients with ocular graft-versus-host disease. Ocul. Surf. 2019, 17, 464–469. [Google Scholar] [CrossRef] [PubMed]
  47. Kim, S.; Yoo, Y.-S.; Kim, H.S.; Joo, C.-K.; Na, K.-S. Changes of meibomian glands in the early stage of post hematopoietic stem cell transplantation. Exp. Eye Res. 2017, 163, 85–90. [Google Scholar] [CrossRef]
  48. Ogawa, Y.; Kuwana, M.; Yamazaki, K.; Mashima, Y.; Yamada, M.; Mori, T.; Okamoto, S.; Oguchi, Y.; Kawakami, Y. Periductal Area as the Primary Site for T-Cell Activation in Lacrimal Gland Chronic Graft-Versus-Host Disease. Investig. Ophthalmol. Vis. Sci. 2003, 44, 1888–1896. [Google Scholar] [CrossRef] [Green Version]
  49. Tatematsu, Y.; Ogawa, Y.; Shimmura, S.; Dogru, M.; Yaguchi, S.; Nagai, T.; Yamazaki, K.; Kameyama, K.; Okamoto, S.; Kawakami, Y.; et al. Mucosal microvilli in dry eye patients with chronic GVHD. Bone Marrow Transplant. 2012, 47, 416–425. [Google Scholar] [CrossRef] [PubMed]
  50. Wang, Y.; Ogawa, Y.; Dogru, M.; Kawai, M.; Tatematsu, Y.; Uchino, M.; Okada, N.; Igarashi, A.; Kujira, A.; Fujishima, H.; et al. Ocular surface and tear functions after topical cyclosporine treatment in dry eye patients with chronic graft-versus-host disease. Bone Marrow Transplant. 2008, 41, 293–302. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Koyama, M.; Hill, G.R. The primacy of gastrointestinal tract antigen-presenting cells in lethal graft-versus-host disease. Blood 2019, 134, 2139–2148. [Google Scholar] [CrossRef]
  52. Zeiser, R.; Blazar, B.R. Acute Graft-versus-Host Disease—Biologic Process, Prevention, and Therapy. N. Engl. J. Med. 2017, 377, 2167–2179. [Google Scholar] [CrossRef]
  53. Jabs, D.A.; Wingard, J.; Green, W.R.; Farmer, E.R.; Vogelsang, G.; Saral, R. The eye in bone marrow transplantation. III. Conjunctival graft-vs-host disease. Arch. Ophthalmol. 1989, 107, 1343–1348. [Google Scholar] [CrossRef]
  54. Saito, T. Ocular manifestation of acute graft-versus-host disease after allogeneic peripheral blood stem cell transplantation. Int. J. Hematol. 2002, 75, 332–334. [Google Scholar] [CrossRef]
  55. Uchino, M.; Ogawa, Y.; Kawai, M.; Shimada, H.; Kameyama, K.; Okamoto, S.; Tsubota, K. Ocular complications in a child with acute graft-versus-host disease following cord blood stem cell transplantation: Therapeutic challenges. Acta Ophthalmol. Scand 2006, 84, 545–548. [Google Scholar] [CrossRef] [PubMed]
  56. Hayashi, S.; Shimizu, E.; Uchino, M.; Yazu, H.; Aketa, N.; Tsubota, K.; Ogawa, Y. The Overlap Syndrome: A Case Report of Chronic Graft-Versus-Host Disease After the Development of a Pseudomembrane. Cornea 2020. online ahead of print. [Google Scholar] [CrossRef] [PubMed]
  57. Allan, E.J.; Flowers, M.E.D.; Lin, M.P.; E Bensinger, R.; Martin, P.J.; Wu, M.C. Visual Acuity and Anterior Segment Findings in Chronic Graft-Versus-Host Disease. Cornea 2011, 30, 1392–1397. [Google Scholar] [CrossRef] [PubMed]
  58. Hirst, L.W.; Jabs, D.A.; Tutschka, P.J.; Green, W.R.; Santos, G.W. The eye in bone marrow transplantation. I. Clinical study. Arch. Ophthalmol. 1983, 101, 580–584. [Google Scholar] [CrossRef]
  59. Kerty, E.; Vigander, K.; Flage, T.; Brinch, L. Ocular findings in allogeneic stem cell transplantation without total body irradiation. Ophthalmology 1999, 106, 1334–1338. [Google Scholar] [CrossRef]
  60. Kim, S.K. Ocular Graft vs. Host Disease. Ocul. Surf. 2005, 3, S177–S179. [Google Scholar] [CrossRef]
  61. Quatrini, L.; Tumino, N.; Moretta, F.; Besi, F.; Vacca, P.; Moretta, L. Helper Innate Lymphoid Cells in Allogenic Hematopoietic Stem Cell Transplantation and Graft Versus Host Disease. Front. Immunol. 2020, 11, 582098. [Google Scholar] [CrossRef] [PubMed]
  62. Shao, L.; Pan, S.; Zhang, Q.-P.; Jamal, M.; Chen, L.-H.; Yin, Q.; Wu, Y.-J.; Xiong, J.; Xiao, R.-J.; Kwong, Y.-L.; et al. An Essential Role of Innate Lymphoid Cells in the Pathophysiology of Graft-vs.-Host Disease. Front. Immunol. 2019, 10. [Google Scholar] [CrossRef] [PubMed]
  63. Vacca, P.; Montaldo, E.; Croxatto, D.; Moretta, F.; Bertaina, A.; Vitale, C.; Locatelli, F.; Mingari, M.C.; Moretta, L. NK Cells and Other Innate Lymphoid Cells in Hematopoietic Stem Cell Transplantation. Front. Immunol. 2016, 7, 188. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Konya, V.; Mjosberg, J. Innate lymphoid cells in graft-versus-host disease. Am. J. Transplant. 2015, 15, 2795–2801. [Google Scholar] [CrossRef] [PubMed]
  65. Munneke, J.M.; Björklund, A.T.; Mjösberg, J.M.; Garming-Legert, K.; Bernink, J.H.; Blom, B.; Huisman, C.; Van Oers, M.H.J.; Spits, H.; Malmberg, K.-J.; et al. Activated innate lymphoid cells are associated with a reduced susceptibility to graft-versus-host disease. Blood 2014, 124, 812–821. [Google Scholar] [CrossRef]
  66. Herretes, S.; Ross, D.B.; Duffort, S.; Barreras, H.; Yaohong, T.; Saeed, A.M.; Murillo, J.C.; Komanduri, K.V.; Levy, R.B.; Perez, V.L. Recruitment of Donor T Cells to the Eyes During Ocular GVHD in Recipients of MHC-Matched Allogeneic Hematopoietic Stem Cell Transplants. Investig. Ophthalmol. Vis. Sci. 2015, 56, 2348–2357. [Google Scholar] [CrossRef] [Green Version]
  67. Ogawa, Y.; Kodama, H.; Kameyama, K.; Yamazaki, K.; Yasuoka, H.; Okamoto, S.; Inoko, H.; Kawakami, Y.; Kuwana, M. Donor Fibroblast Chimerism in the Pathogenic Fibrotic Lesion of Human Chronic Graft-Versus-Host Disease. Investig. Ophthalmol. Vis. Sci. 2005, 46, 4519–4527. [Google Scholar] [CrossRef] [Green Version]
  68. Rojas, B.; Cuhna, R.; Zafirakis, P.; Ramirez, J.M.; Lizan-garciı, M.; Zhao, T.; Foster, C.S. Cell populations and adhesion molecules expression in conjunctiva before and after bone marrow transplantation. Exp. Eye Res. 2005, 81, 313–325. [Google Scholar] [CrossRef] [PubMed]
  69. Ogawa, Y.; Morikawa, S.; Okano, H.; Mabuchi, Y.; Suzuki, S.; Yaguchi, T.; Sato, Y.; Mukai, S.; Yaguchi, S.; Inaba, T.; et al. MHC-compatible bone marrow stromal/stem cells trigger fibrosis by activating host T cells in a scleroderma mouse model. eLife 2016, 5, e09394. [Google Scholar] [CrossRef] [PubMed]
  70. Perez, V.L.; Barsam, A.; Duffort, S.; Urbieta, M.; Barreras, H.; Lightbourn, C.; Komanduri, K.V.; Levy, R.B. Novel Scoring Criteria for the Evaluation of Ocular Graft-versus-Host Disease in a Preclinical Allogeneic Hematopoietic Stem Cell Transplantation Animal Model. Biol. Blood Marrow Transplant. 2016, 22, 1765–1772. [Google Scholar] [CrossRef] [Green Version]
  71. He, J.; Ogawa, Y.; Mukai, S.; Saijo-Ban, Y.; Kamoi, M.; Uchino, M.; Yamane, M.; Ozawa, N.; Fukui, M.; Mori, T.; et al. In Vivo Confocal Microscopy Evaluation of Ocular Surface with Graft-Versus-Host Disease-Related Dry Eye Disease. Sci. Rep. 2017, 7, 1–10. [Google Scholar] [CrossRef]
  72. Kheirkhah, A.; Qazi, Y.; Arnoldner, M.A.; Suri, K.; Dana, R. In Vivo Confocal Microscopy in Dry Eye Disease Associated With Chronic Graft-Versus-Host Disease. Investig. Ophthalmol. Vis. Sci. 2016, 57, 4686–4691. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. He, C.; Lai, P.; Weng, J.; Lin, S.; Wu, K.; Du, X.; Liu, X. Toll-like receptor 2-mediated NF-κB inflammatory responses in dry eye associated with cGVHD. Mol. Vis. 2011, 17, 2605–2611. [Google Scholar] [PubMed]
  74. Bettelli, E.; Oukka, M.; Kuchroo, V.K. T(H)-17 cells in the circle of immunity and autoimmunity. Nat. Immunol. 2007, 8, 345–350. [Google Scholar] [CrossRef]
  75. Stevenson, W.; Chauhan, S.K.; Dana, R. Dry eye disease: An immune-mediated ocular surface disorder. Arch. Ophthalmol. 2012, 130, 90–100. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Chen, Y.; Chauhan, S.K.; Lee, H.S.; Saban, D.R.; Dana, R. Chronic dry eye disease is principally mediated by effector memory Th17 cells. Mucosal Immunol. 2014, 7, 38–45. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Stern, M.E.; Schaumburg, C.S.; Pflugfelder, S.C. Dry Eye as a Mucosal Autoimmune Disease. Int. Rev. Immunol. 2013, 32, 19–41. [Google Scholar] [CrossRef]
  78. Inagaki, E.; Ogawa, Y.; Matsumoto, Y.; Kawakita, T.; Shimmura, S.; Tsubota, K. Four cases of corneal perforation in patients with chronic graft-versus-host disease. Mol. Vis. 2011, 17, 598–606. [Google Scholar] [PubMed]
  79. Suzuki, M.; Usui, T.; Kinoshita, N.; Yamagami, S.; Amano, S. A case of sterile corneal perforation after bone marrow transplantation. Eye 2007, 21, 114–116. [Google Scholar] [CrossRef] [Green Version]
  80. Zeiser, R.; Penack, O.; Holler, E.; Idzko, M. Danger signals activating innate immunity in graft-versus-host disease. J. Mol. Med. 2011, 89, 833–845. [Google Scholar] [CrossRef]
  81. An, S.; Raju, I.; Surenkhuu, B.; Kwon, J.-E.; Gulati, S.; Karaman, M.; Pradeep, A.; Sinha, S.; Mun, C.; Jain, S. Neutrophil extracellular traps (NETs) contribute to pathological changes of ocular graft-vs.-host disease (oGVHD) dry eye: Implications for novel biomarkers and therapeutic strategies. Ocul. Surf. 2019, 17, 589–614. [Google Scholar] [CrossRef] [PubMed]
  82. Tibrewal, S.; Sarkar, J.; Jassim, S.H.; Gandhi, S.; Sonawane, S.; Chaudhary, S.; Byun, Y.-S.; Ivanir, Y.; Hallak, J.; Horner, J.H.; et al. Tear Fluid Extracellular DNA: Diagnostic and Therapeutic Implications in Dry Eye Disease. Investig. Ophthalmol. Vis. Sci. 2013, 54, 8051–8062. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Yamane, M.; Sato, S.; Shimizu, E.; Shibata, S.; Hayano, M.; Yaguchi, T.; Tsubota, K. Senescence-associated secretory phenotype promotes chronic ocular graft-vs-host disease in mice and humans. FASEB J. 2020, 34, 10778–10800. [Google Scholar] [CrossRef]
  84. Zhang, M.; Zhang, S. T Cells in Fibrosis and Fibrotic Diseases. Front. Immunol. 2020, 11, 1142. [Google Scholar] [CrossRef]
  85. Ogawa, Y.; Razzaque, M.S.; Kameyama, K.; Hasegawa, G.; Shimmura, S.; Kawai, M.; Okamoto, S.; Ikeda, Y.; Tsubota, K.; Kawakami, Y.; et al. Role of Heat Shock Protein 47, a Collagen-Binding Chaperone, in Lacrimal Gland Pathology in Patients with cGVHD. Investig. Ophthalmol. Vis. Sci. 2007, 48, 1079–1086. [Google Scholar] [CrossRef] [PubMed]
  86. Razzaque, M.S.; Ahmed, B.S.; Foster, C.S.; Ahmed, A.R. Effects of IL-4 on Conjunctival Fibroblasts: Possible Role in Ocular Cicatricial Pemphigoid. Investig. Ophthalmol. Vis. Sci. 2003, 44, 3417–3423. [Google Scholar] [CrossRef] [Green Version]
  87. Razzaque, M.S.; Foster, C.S.; Ahmed, A.R. Role of connective tissue growth factor in the pathogenesis of conjunctival scarring in ocular cicatricial pemphigoid. Investig. Ophthalmol. Vis. Sci. 2003, 44, 1998–2003. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Razzaque, M.S.; Foster, C.S.; Ahmed, A.R. Role of collagen-binding heat shock protein 47 and transforming growth factor-beta1 in conjunctival scarring in ocular cicatricial pemphigoid. Investig. Ophthalmol. Vis. Sci. 2003, 44, 1616–1621. [Google Scholar] [CrossRef] [Green Version]
  89. Chesney, J.; Bacher, M.; Bender, A.; Bucala, R. The peripheral blood fibrocyte is a potent antigen-presenting cell capable of priming naive T cells in situ. Proc. Natl. Acad. Sci. USA 1997, 94, 6307–6312. [Google Scholar] [CrossRef] [Green Version]
  90. Hu, X.; DeBiasi, E.M.; Herzog, E.L. Flow Cytometric Identification of Fibrocytes in the Human Circulation. Advanced Structural Safety Studies 2015, 1343, 19–33. [Google Scholar] [CrossRef]
  91. Yamazaki, K.; Eyden, B.P. Ultrastructural and immunohistochemical observations on intralobular fibroblasts of human breast, with observations on the CD34 antigen. J. Submicrosc. Cytol. Pathol. 1995, 27, 309–323. [Google Scholar]
  92. Yamazaki, K.; Eyden, B.P. Ultrastructural and immunohistochemical studies of intralobular fibroblasts in human submandibular gland: The recognition of a ‘CD34 positive reticular network’ connected by gap junctions. J. Submicrosc. Cytol. Pathol. 1996, 28, 471–483. [Google Scholar]
  93. Yamazaki, K.; Eyden, B.P. Interfollicular fibroblasts in the human thyroid gland: Recognition of a CD34 positive stromal cell network communicated by gap junctions and terminated by autonomic nerve endings. J. Submicrosc. Cytol. Pathol. 1997, 29, 461–476. [Google Scholar] [PubMed]
  94. Bernardo, M.E.; Fibbe, W.E. Mesenchymal Stromal Cells: Sensors and Switchers of Inflammation. Cell Stem Cell 2013, 13, 392–402. [Google Scholar] [CrossRef] [Green Version]
  95. Martínez-Carrasco, R.; Sánchez-Abarca, L.I.; Nieto-Gómez, C.; García, E.M.; Sánchez-Guijo, F.; Argüeso, P.; Aijón, J.; Hernández-Galilea, E.; Velasco, A. Subconjunctival injection of mesenchymal stromal cells protects the cornea in an experimental model of GVHD. Ocul. Surf. 2019, 17, 285–294. [Google Scholar] [CrossRef]
  96. Sánchez-Abarca, L.I.; Hernández-Galilea, E.; Lorenzo, R.; Herrero, C.; Velasco, A.; Carrancio, S.; Caballero-Velázquez, T.; Rodríguez-Barbosa, J.I.; Parrilla, M.; Del Cañizo, C.; et al. Human Bone Marrow Stromal Cells Differentiate into Corneal Tissue and Prevent Ocular Graft-Versus-Host Disease in Mice. Cell Transplant. 2015, 24, 2423–2433. [Google Scholar] [CrossRef] [Green Version]
  97. Tiwari, A.; Swamynathan, S.; Jhanji, V.; Swamynathan, S.K. KLF4 Coordinates Corneal Epithelial Apical-Basal Polarity and Plane of Cell Division and Is Downregulated in Ocular Surface Squamous Neoplasia. Investig. Ophthalmol. Vis. Sci. 2020, 61, 15. [Google Scholar] [CrossRef]
  98. Rajić, J.; Dinić, S.; Uskoković, A.; Jovanović, J.A.; Tolić, A.; Đorđević, M.; Đorđević, M.; Poznanović, G.; Mihailović, M.; Inic-Kanada, A.; et al. DNA methylation of miR-200 clusters promotes epithelial to mesenchymal transition in human conjunctival epithelial cells. Exp. Eye Res. 2020, 197, 108047. [Google Scholar] [CrossRef] [PubMed]
  99. Shibata, S.; Hayashi, R.; Okubo, T.; Kudo, Y.; Baba, K.; Honma, Y.; Nishida, K. The secretome of adipose-derived mesenchymal stem cells attenuates epithelial–mesenchymal transition in human corneal epithelium. Regen. Ther. 2019, 11, 114–122. [Google Scholar] [CrossRef] [PubMed]
  100. Takahashi, E.; Haga, A.; Tanihara, H. Merlin regulates the epithelial-to-mesenchymal transition of ARPE-19 cells via TAK1-p38MAPK-mediated activation. Investig. Ophthalmol. Vis. Sci. 2015, 56, 2449. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Kalluri, R.; Neilson, E.G. Epithelial-mesenchymal transition and its implications for fibrosis. J. Clin. Investig. 2003, 112, 1776–1784. [Google Scholar] [CrossRef] [PubMed]
  102. Tsukamoto, H.; Shibata, K.; Kajiyama, H.; Terauchi, M.; Nawa, A.; Kikkawa, F. Irradiation-induced epithelial–mesenchymal transition (EMT) related to invasive potential in endometrial carcinoma cells. Gynecol. Oncol. 2007, 107, 500–504. [Google Scholar] [CrossRef]
  103. Higgins, D.F.; Kimura, K.; Bernhardt, W.M.; Shrimanker, N.; Akai, Y.; Hohenstein, B.; Saito, Y.; Johnson, R.S.; Kretzler, M.; Cohen, C.D.; et al. Hypoxia promotes fibrogenesis in vivo via HIF-1 stimulation of epithelial-to-mesenchymal transition. J. Clin. Investig. 2007, 117, 3810–3820. [Google Scholar] [CrossRef]
  104. Radisky, D.C.; Levy, D.D.; Littlepage, L.E.; Liu, H.; Nelson, C.M.; Fata, J.E.; Leake, D.; Godden, E.L.; Albertson, D.G.; Nieto, M.A.; et al. Rac1b and reactive oxygen species mediate MMP-3-induced EMT and genomic instability. Nat. Cell Biol. 2005, 436, 123–127. [Google Scholar] [CrossRef] [Green Version]
  105. Yang, J.; Antin, P.; Berx, G.; Blanpain, C.; Brabletz, T.; Bronner, M.; Campbell, K.; Cano, A.; Casanova, J.; Christofori, G.; et al. Guidelines and definitions for research on epithelial–mesenchymal transition. Nat. Rev. Mol. Cell Biol. 2020, 21, 341–352. [Google Scholar] [CrossRef] [Green Version]
  106. Shintani, Y.; Maeda, M.; Chaika, N.; Johnson, K.R.; Wheelock, M.J. Collagen I promotes epithelial-to-mesenchymal transition in lung cancer cells via transforming growth factor-beta signaling. Am. J. Respir. Cell Mol. Biol. 2008, 38, 95–104. [Google Scholar] [CrossRef] [Green Version]
  107. Kawai, M.; Ogawa, Y.; Shimmura, S.; Ohta, S.; Suzuki, T.; Kawamura, N.; Kuwana, M.; Kawakami, Y.; Tsubota, K. Expression and localization of aging markers in lacrimal gland of chronic graft-versus-host disease. Sci. Rep. 2013, 3, 2455. [Google Scholar] [CrossRef] [Green Version]
  108. Sofi, M.H.; Wu, Y.; Schutt, S.D.; Dai, M.; Daenthanasanmak, A.; Voss, J.H.; Nguyen, H.; Bastian, D.; Iamsawat, S.; Selvam, S.P.; et al. Thioredoxin-1 confines T cell alloresponse and pathogenicity in graft-versus-host disease. J. Clin. Investig. 2019, 129, 2760–2774. [Google Scholar] [CrossRef] [PubMed]
  109. Harman, D. The Free Radical Theory of Aging. Antioxidants Redox Signal. 2003, 5, 557–561. [Google Scholar] [CrossRef] [PubMed]
  110. Yaguchi, S.; Ogawa, Y.; Shimmura, S.; Kawakita, T.; Hatou, S.; Satofuka, S.; Nakamura, S.; Imada, T.; Miyashita, H.; Yoshida, S.; et al. Angiotensin II Type 1 Receptor Antagonist Attenuates Lacrimal Gland, Lung, and Liver Fibrosis in a Murine Model of Chronic Graft-Versus-Host Disease. PLoS ONE 2013, 8, e64724. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  111. Mukai, S.; Ogawa, Y.; Kawakami, Y.; Mashima, Y.; Tsubota, K. Inhibition of Vascular Adhesion Protein-1 for Treatment of Graft-Versus-Host Disease in Mice. FASEB J. 2018, 32, 4085–4095. [Google Scholar] [CrossRef] [Green Version]
  112. Mukai, S.; Ogawa, Y.; Urano, F.; Kudo-Saito, C.; Kawakami, Y.; Tsubota, K. Novel Treatment of Chronic Graft-Versus-Host Disease in Mice Using the ER Stress Reducer 4-Phenylbutyric Acid. Sci. Rep. 2017, 7, 41939. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Yoshimoto, S.; Loo, T.M.; Atarashi, K.; Kanda, H.; Sato, S.; Oyadomari, S.; Iwakura, Y.; Oshima, K.; Morita, H.; Hattori, M.; et al. Obesity-induced gut microbial metabolite promotes liver cancer through senescence secretome. Nature 2013, 499, 97–101. [Google Scholar] [CrossRef]
  114. Lin, J.B.; Séne, A.; Santeford, A.; Fujiwara, H.; Sidhu, R.; Ligon, M.M.; Shankar, V.A.; Ban, N.; Mysorekar, I.U.; Ory, D.S.; et al. Oxysterol Signatures Distinguish Age-Related Macular Degeneration from Physiologic Aging. EBioMedicine 2018, 32, 9–20. [Google Scholar] [CrossRef] [PubMed]
  115. Zhou, R.; Yazdi, A.S.; Menu, P.; Tschopp, J. A role for mitochondria in NLRP3 inflammasome activation. Nature 2011, 469, 221–225. [Google Scholar] [CrossRef]
  116. Bakeeva, L.E.; Eldarov, C.M.; Vangely, I.M.; Kolosova, N.G.; Vays, V.B. Mitochondria-targeted antioxidant SkQ1 reduces age-related alterations in the ultrastructure of the lacrimal gland. Oncotarget 2016, 7, 80208–80222. [Google Scholar] [CrossRef] [Green Version]
  117. Batista, T.; Tomiyoshi, L.M.; Dias, A.C.; Roma, L.P.; Módulo, C.M.; Malki, L.T.; Filho, E.B.M.; Deminice, R.; Jordão, A.A.; Cunha, D.A.; et al. Age-dependent changes in rat lacrimal gland anti-oxidant and vesicular related protein expression profiles. Mol. Vis. 2012, 18, 194–202. [Google Scholar]
  118. Uchino, Y.; Kawakita, T.; Miyazawa, M.; Ishii, T.; Onouchi, H.; Yasuda, K.; Ogawa, Y.; Shimmura, S.; Ishii, N.; Tsubota, K. Oxidative Stress Induced Inflammation Initiates Functional Decline of Tear Production. PLoS ONE 2012, 7, e45805. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Dogru, M.; Kojima, T.; Simsek, C.; Tsubota, K. Potential Role of Oxidative Stress in Ocular Surface Inflammation and Dry Eye Disease. Investig. Ophthalmol. Vis. Sci. 2018, 59, DES163–DES168. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Vajapey, R.; Rini, D.; Walston, J.; Abadir, P. The impact of age-related dysregulation of the angiotensin system on mitochondrial redox balance. Front. Physiol. 2014, 5, 439. [Google Scholar] [CrossRef] [Green Version]
  121. Benigni, A.; Cassis, P.; Remuzzi, G. Angiotensin II revisited: New roles in inflammation, immunology and aging. EMBO Mol. Med. 2010, 2, 247–257. [Google Scholar] [CrossRef] [PubMed]
  122. Shay, J.W. Accelerated telomere shortening in bone-marrow recipients. Lancet 1998, 351, 153–154. [Google Scholar] [CrossRef]
  123. Garg, A.D.; Kaczmarek, A.; Krysko, O.; Vandenabeele, P.; Krysko, D.; Agostinis, P. ER stress-induced inflammation: Does it aid or impede disease progression? Trends Mol. Med. 2012, 18, 589–598. [Google Scholar] [CrossRef]
  124. Howcroft, T.K.; Campisi, J.; Louis, G.B.; Smith, M.T.; Wise, B.; Wyss-Coray, T.; Augustine, A.D.; McElhaney, J.E.; Kohanski, R.; Sierra, F. The role of inflammation in age-related disease. Aging 2013, 5, 84–93. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Lee, A.S. The ER chaperone and signaling regulator GRP78/BiP as a monitor of endoplasmic reticulum stress. Methods 2005, 35, 373–381. [Google Scholar] [CrossRef] [PubMed]
  126. Brozzi, F.; Nardelli, T.R.; Lopes, M.; Millard, I.; Barthson, J.; Igoillo-Esteve, M.; Grieco, F.A.; Villate, O.; Oliveira, J.M.O.J.; Casimir, M.; et al. Cytokines induce endoplasmic reticulum stress in human, rat and mouse beta cells via different mechanisms. Diabetol. 2015, 58, 2307–2316. [Google Scholar] [CrossRef] [Green Version]
  127. Anthony, T.G.; Wek, R.C. TXNIP Switches Tracks toward a Terminal UPR. Cell Metab. 2012, 16, 135–137. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Kiss, J.; Jalkanen, S.; Fülöp, F.; Savunen, T.; Salmi, M. Ischemia-reperfusion injury is attenuated in VAP-1-deficient mice and by VAP-1 inhibitors. Eur. J. Immunol. 2008, 38, 3041–3049. [Google Scholar] [CrossRef]
  129. Salmi, M.; Jalkanen, S. Cell-surface enzymes in control of leukocyte trafficking. Nat. Rev. Immunol. 2005, 5, 760–771. [Google Scholar] [CrossRef]
  130. Foot, J.S.; Yow, T.T.; Schilter, H.; Buson, A.; Deodhar, M.; Findlay, A.D.; Guo, L.; McDonald, I.A.; Turner, C.I.; Zhou, W.; et al. PXS-4681A, a Potent and Selective Mechanism-Based Inhibitor of SSAO/VAP-1 with Anti-Inflammatory Effects In Vivo. J. Pharmacol. Exp. Ther. 2013, 347, 365–374. [Google Scholar] [CrossRef] [Green Version]
  131. Kitko, C.L.; Levine, J.E.; Storer, B.E.; Chai, X.; Fox, D.A.; Braun, T.M.; Couriel, D.R.; Martin, P.J.; Flowers, M.E.; Hansen, J.A.; et al. Plasma CXCL9 elevations correlate with chronic GVHD diagnosis. Blood 2014, 123, 786–793. [Google Scholar] [CrossRef] [Green Version]
  132. Tung, C.I. Graft versus host disease: What should the oculoplastic surgeon know? Curr. Opin. Ophthalmol. 2017, 28, 499–504. [Google Scholar] [CrossRef]
  133. Tung, C.I. Current Approaches to Treatment of Ocular Graft-Versus-Host Disease. Int. Ophthalmol. Clin. 2017, 57, 65–88. [Google Scholar] [CrossRef]
  134. Espana, E.M.; Shah, S.; Santhiago, M.R.; Singh, A.D. Graft versus host disease: Clinical evaluation, diagnosis and management. Graefe’s Arch. Clin. Exp. Ophthalmol. 2013, 251, 1257–1266. [Google Scholar] [CrossRef] [PubMed]
  135. Riemens, A.; Boome, L.T.; Imhof, S.; Kuball, J.; Rothova, A. Current insights into ocular graft-versus-host disease. Curr. Opin. Ophthalmol. 2010, 21, 485–494. [Google Scholar] [CrossRef]
  136. Vanathi, M.; Nair, S.; Mukhija, R.; Tandon, R.; Jain, S.; Ogawa, Y. Update on ocular graft-versus-host disease. Indian J. Ophthalmol. 2021, 69, 1038–1050. [Google Scholar] [CrossRef]
  137. Hessen, M.; Akpek, E.K. Ocular graft-versus-host disease. Curr. Opin. Allergy Clin. Immunol. 2012, 12, 540–547. [Google Scholar] [CrossRef] [PubMed]
  138. Yamane, M.; Ogawa, Y.; Fukui, M.; Kamoi, M.; Uchino, M.; Saijo-Ban, Y.; Kozuki, N.; Mukai, S.; Mori, T.; Okamoto, S.; et al. Long-Term Topical Diquafosol Tetrasodium Treatment of Dry Eye Disease Caused by Chronic Graft-Versus-Host Disease: A Retrospective Study. Eye Contact Lens Sci. Clin. Pr. 2018, 44, S215–S220. [Google Scholar] [CrossRef] [PubMed]
  139. Sabti, S.; Halter, J.P.; Braun Fränkl, B.C.; Goldblum, D. Punctal occlusion is safe and efficient for the treatment of keratoconjunctivitis sicca in patients with ocular GvHD. Bone Marrow Transpl. 2012, 47, 981–984. [Google Scholar] [CrossRef] [Green Version]
  140. Yaguchi, S.; Ogawa, Y.; Kamoi, M.; Uchino, M.; Tatematsu, Y.; Ban, Y.; Ohba, E.; Okamoto, S.; Goto, E.; Tsubota, K. Surgical management of lacrimal punctal cauterization in chronic GVHD-related dry eye with recurrent punctal plug extrusion. Bone Marrow Transplant. 2012, 47, 1465–1469. [Google Scholar] [CrossRef] [Green Version]
  141. Agha-Hosseini, F.; Mirzaii-Dizgah, I.; Ghavamzadeh, L.; Ghavamzadeh, A.; Tohidast-Acrad, Z. Effect of pilocarpine hydrochloride on unstimulated whole saliva flow rate and composition in patients with chronic graft-versus-host disease (cGVHD). Bone Marrow Transpl. 2007, 39, 431–434. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Yamane, M.; Ogawa, Y.; Fukui, M.; Kamoi, M.; Saijo-Ban, Y.; Yaguchi, S.; Mukai, S.; Kawakita, T.; Simmura, S.; Tsubota, K. Long-term Rebamipide and Diquafosol in Two Cases of Immune-Mediated Dry Eye. Optom. Vis. Sci. 2015, 92, S25–S32. [Google Scholar] [CrossRef] [Green Version]
  143. Dietrich-Ntoukas, T. Diagnosis and treatment of ocular chronic graft-versus-host disease: Report from the German-Austrian-Swiss Consensus Conference on Clinical Practice in chronic GVHD. Cornea 2012, 31, 299–310. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Abud, T.B.; Amparo, F.; Saboo, U.S.; Di Zazzo, A.; Dohlman, T.H.; Ciolino, J.; Hamrah, P.; Dana, R. A Clinical Trial Comparing the Safety and Efficacy of Topical Tacrolimus versus Methylprednisolone in Ocular Graft-versus-Host Disease. Ophthalmol. 2016, 123, 1449–1457. [Google Scholar] [CrossRef] [Green Version]
  145. Sato, S.; Shimizu, E.; He, J.; Ogawa, M.; Asai, K.; Yazu, H.; Rusch, R.; Yamane, M.; Yang, F.; Fukuda, S.; et al. Positive Effects of Oral Antibiotic Administration in Murine Chronic Graft-Versus-Host Disease. Int. J. Mol. Sci. 2021, 22, 3745. [Google Scholar] [CrossRef] [PubMed]
  146. Jung, J.W.; Lee, Y.J.; Yoon, S.C.; Kim, T.-I.; Kim, E.K.; Seo, K.Y. Long-term Result of Maintenance Treatment With Tacrolimus Ointment in Chronic Ocular Graft-Versus-Host Disease. Am. J. Ophthalmol. 2015, 159, 519–527.e1. [Google Scholar] [CrossRef]
  147. Poe, J.C.; Jia, W.; Di Paolo, J.A.; Reyes, N.J.; Kim, J.Y.; Su, H.; Sundy, J.S.; Cardones, A.R.; Perez, V.L.; Chen, B.J.; et al. SYK inhibitor entospletinib prevents ocular and skin GVHD in mice. JCI Insight 2018, 3. [Google Scholar] [CrossRef]
  148. Ogawa, Y.; Okamoto, S.; Mori, T.; Yamada, M.; Mashima, Y.; Watanabe, R.; Kuwana, M.; Tsubota, K.; Ikeda, Y.; Oguchi, Y. Autologous serum eye drops for the treatment of severe dry eye in patients with chronic graft-versus-host disease. Bone Marrow Transplant. 2003, 31, 579–583. [Google Scholar] [CrossRef] [Green Version]
  149. Tahmaz, V.; Gehlsen, U.; Sauerbier, L.; Holtick, U.; Engel, L.; Radojska, S.; Petrescu-Jipa, V.-M.; Scheid, C.; Hallek, M.; Gathof, B.; et al. Treatment of severe chronic ocular graft-versus-host disease using 100% autologous serum eye drops from a sealed manufacturing system: A retrospective cohort study. Br. J. Ophthalmol. 2017, 101, 322–326. [Google Scholar] [CrossRef]
  150. Yoon, K.-C.; Jeong, I.-Y.; Im, S.-K.; Park, Y.-G.; Kim, H.-J.; Choi, J. Therapeutic effect of umbilical cord serum eyedrops for the treatment of dry eye associated with graft-versus-host disease. Bone Marrow Transplant. 2007, 39, 231–235. [Google Scholar] [CrossRef] [Green Version]
  151. Jones, L.; Downie, L.; Korb, D.; Benitez-Del-Castillo, J.M.; Dana, R.; Deng, S.X.; Dong, P.N.; Geerling, G.; Hida, R.Y.; Liu, Y.; et al. TFOS DEWS II Management and Therapy Report. Ocul. Surf. 2017, 15, 575–628. [Google Scholar] [CrossRef]
  152. Pezzotta, S.; Del Fante, C.; Scudeller, L.; Rossi, G.C.; Perotti, C.; E Bianchi, P.; Antoniazzi, E. Long-term safety and efficacy of autologous platelet lysate drops for treatment of ocular GvHD. Bone Marrow Transplant. 2017, 52, 101–106. [Google Scholar] [CrossRef]
  153. Valentini, C.G.; Nuzzolo, E.R.; Orlando, N.; Metafuni, E.; Bianchi, M.; Chiusolo, P.; Zini, G.; Teofili, L. Cytokine profile of autologous platelet-derived eye drops in patients with ocular chronic graft-versus-host disease. Vox Sang. 2016, 110, 189–192. [Google Scholar] [CrossRef] [PubMed]
  154. Ikarashi, H.; Aketa, N.; Shimizu, E.; Takano, Y.; Kawakita, T.; Uchino, Y.; Matsumoto, Y.; Ogawa, J.; Tsubota, K.; Ogawa, Y. Two case reports of continued progression of chronic ocular graft-versus-host disease without concurrent systemic comorbidities treated by amniotic membrane transplantation. BMC Ophthalmol. 2021, 21, 1–5. [Google Scholar] [CrossRef]
  155. Ma, K.N.; Thanos, A.; Chodosh, J.; Shah, A.S.; Mantagos, I.S. A Novel Technique for Amniotic Membrane Transplantation in Patients with Acute Stevens-Johnson Syndrome. Ocul. Surf. 2016, 14, 31–36. [Google Scholar] [CrossRef] [PubMed]
  156. Mohammadpour, M.; Maleki, S.; Hashemi, H.; Beheshtnejad, A.H. Recurrent Corneal Perforation due to Chronic Graft versus Host Disease; a Clinicopathologic Report. J. Ophthalmic Vis. Res. 2016, 11, 108–111. [Google Scholar] [CrossRef] [Green Version]
  157. Yen, P.-T.; Hou, Y.-C.; Lin, W.-C.; Wang, L.-J.; Hu, F.-R. Recurrent Corneal Perforation and Acute Calcareous Corneal Degeneration in Chronic Graft-Versus-Host Disease. J. Formos. Med Assoc. 2006, 105, 334–339. [Google Scholar] [CrossRef]
  158. Meller, D.; Fuchsluger, T.; Pauklin, M.; Steuhl, K.-P. Ocular Surface Reconstruction in Graft-Versus-Host Disease With HLA-Identical Living-Related Allogeneic Cultivated Limbal Epithelium After Hematopoietic Stem Cell Transplantation from the Same Donor. Cornea 2009, 28, 233–236. [Google Scholar] [CrossRef] [PubMed]
  159. DeLoss, K.S.; Le, H.-G.; Gire, A.; Chiu, G.B.; Jacobs, D.S.; Carrasquillo, K.G. PROSE Treatment for Ocular Chronic Graft-Versus-Host Disease as a Clinical Network Expands. Eye Contact Lens: Sci. Clin. Pr. 2016, 42, 262–266. [Google Scholar] [CrossRef] [PubMed]
  160. Inamoto, Y.; Sun, Y.-C.; Flowers, M.E.; Carpenter, P.A.; Martin, P.J.; Li, P.; Wang, R.; Chai, X.; Storer, B.E.; Shen, T.T.; et al. Bandage Soft Contact Lenses for Ocular Graft-versus-Host Disease. Biol. Blood Marrow Transplant. 2015, 21, 2002–2007. [Google Scholar] [CrossRef] [Green Version]
  161. Russo, P.A.; Bouchard, C.S.; Galasso, J.M. Extended-Wear Silicone Hydrogel Soft Contact Lenses in the Management of Moderate to Severe Dry Eye Signs and Symptoms Secondary to Graft-Versus-Host Disease. Eye Contact Lens Sci. Clin. Pr. 2007, 33, 144–147. [Google Scholar] [CrossRef]
  162. Ohigashi, H.; Hashimoto, D.; Hayase, E.; Takahashi, S.; Ara, T.; Yamakawa, T.; Sugita, J.; Onozawa, M.; Nakagawa, M.; Teshima, T. Ocular instillation of vitamin A–coupled liposomes containing HSP47 siRNA ameliorates dry eye syndrome in chronic GVHD. Blood Adv. 2019, 3, 1003–1010. [Google Scholar] [CrossRef] [Green Version]
  163. Steven, P.; Faust, C.; Holtick, U.; Scheid, C.; Tahmaz, V.; Stern, M.E.; Gehlsen, U. Adverse environmental conditions are a risk factor for ocular GvHD after allogeneic hematopoietic stem cell transplantation. Bone Marrow Transplant. 2020, 55, 1851–1853. [Google Scholar] [CrossRef]
  164. Dulz, S.; Wagenfeld, L.; Richard, G.; Schrum, J.; Muschol, N.; Keserü, M. A Case of a Bilateral Cicatricial Upper Eyelid Entropion After Hematopoietic Stem Cell Transplantation in Mucopolysaccharidosis Type I. Ophthalmic Plast. Reconstr. Surg. 2017, 33, S75–S77. [Google Scholar] [CrossRef] [PubMed]
  165. Frucht-Pery, J.; Sagi, E.; Hemo, I.; Ever-Hadani, P. Efficacy of Doxycycline and Tetracycline in Ocular Rosacea. Am. J. Ophthalmol. 1993, 116, 88–92. [Google Scholar] [CrossRef]
  166. Robinson, M.R.; Lee, S.S.; I Rubin, B.; Wayne, A.S.; Pavletic, S.Z.; Bishop, M.R.; Childs, R.; Barrett, A.J.; Csaky, K.G. Topical corticosteroid therapy for cicatricial conjunctivitis associated with chronic graft-versus-host disease. Bone Marrow Transplant. 2004, 33, 1031–1035. [Google Scholar] [CrossRef] [Green Version]
  167. Chun, Y.H.; Beak, J.U.; Kim, H.-S.; Na, K.-S. Topical Cyclosporine Pretreatment of Ocular Surface in Allogeneic Hematopoietic Stem Cell Transplant Recipients. J. Ocul. Pharmacol. Ther. 2018, 34, 628–632. [Google Scholar] [CrossRef]
  168. Yin, J.; Kheirkhah, A.; Dohlman, T.; Saboo, U.; Dana, R. Reduced Efficacy of Low-dose Topical Steroids in Dry Eye Disease Associated With Graft-versus-Host Disease. Am. J. Ophthalmol. 2018, 190, 17–23. [Google Scholar] [CrossRef]
  169. Chhabra, S.; Jerkins, J.H.; Conto, J.E.; Zellner, K.; Shah, N.N.; Hari, P.N.; Hamadani, M. Lifitegrast ophthalmic solution for treatment of ocular chronic graft-versus-host disease. Leuk. Lymphoma 2019, 61, 869–874. [Google Scholar] [CrossRef] [PubMed]
  170. Ferrete, T.; Rocher, F.; Elmaleh, V.; Loschi, M.; Tieulie, N.; Baillif, S.; Martel, A. Eye amputation following lifitegrast treatment for ocular graft-versus-host disease: First case report. J. Fr. d’Ophtalmol. 2021, 44, 652–657. [Google Scholar] [CrossRef]
  171. Amparo, F. Topical interleukin 1 receptor antagonist for treatment of dry eye disease: A randomized clinical trial. JAMA Ophthalmol. 2013, 131, 715–723. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  172. Darakhshan, S.; Pour, A.B. Tranilast: A review of its therapeutic applications. Pharmacol. Res. 2015, 91, 15–28. [Google Scholar] [CrossRef]
  173. Hertenstein, A.; Schumacher, T.; Litzenburger, U.; Opitz, C.A.; Falk, C.S.; Serafini, T.; Wick, W.; Platten, M. Suppression of human CD4+ T cell activation by 3,4-dimethoxycinnamonyl-anthranilic acid (tranilast) is mediated by CXCL9 and CXCL10. Biochem. Pharmacol. 2011, 82, 632–641. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Ogawa, Y.; Dogru, M.; Uchino, M.; Tatematsu, Y.; Kamoi, M.; Yamamoto, Y.; Ogawa, J.; Ishida, R.; Kaido, M.; Hara, S.; et al. Topical tranilast for treatment of the early stage of mild dry eye associated with chronic GVHD. Bone Marrow Transplant. 2010, 45, 565–569. [Google Scholar] [CrossRef] [Green Version]
  175. Rocha, E.; Pelegrino, F.S.A.; De Paiva, C.S.; Vigorito, A.C.; De Souza, C.A.; Rocha, E. GVHD dry eyes treated with autologous serum tears. Bone Marrow Transplant. 2000, 25, 1101–1103. [Google Scholar] [CrossRef] [Green Version]
  176. Chiang, C.-C.; Lin, J.-M.; Chen, W.-L.; Tsai, Y.-Y. Allogeneic Serum Eye Drops for the Treatment of Severe Dry Eye in Patients With Chronic Graft-Versus-Host Disease. Cornea 2007, 26, 861–863. [Google Scholar] [CrossRef]
  177. Tahmaz, V.; Wiesen, M.H.J.; Gehlsen, U.; Sauerbier, L.; Stern, M.E.; Holtick, U.; Gathof, B.; Scheid, C.; Müller, C.; Steven, P. Detection of systemic immunosuppressants in autologous serum eye drops (ASED) in patients with severe chronic ocular graft versus host disease. Graefe’s Arch. Clin. Exp. Ophthalmol. 2021, 259, 121–128. [Google Scholar] [CrossRef]
  178. Flowers, M.E.D.; Apperley, J.F.; van Besien, K.; Elmaagacli, A.; Grigg, A.; Reddy, V.; Bacigalupo, A.; Kolb, H.-J.; Bouzas, L.; Michallet, M.; et al. A multicenter prospective phase 2 randomized study of extracorporeal photopheresis for treatment of chronic graft-versus-host disease. Blood 2008, 112, 2667–2674. [Google Scholar] [CrossRef] [PubMed]
  179. Perotti, C.; Del Fante, C.; Tinelli, C.; Viarengo, G.; Scudeller, L.; Zecca, M.; Locatelli, F.; Salvaneschi, L. Extracorporeal photochemotherapy in graft-versus-host disease: A longitudinal study on factors influencing the response and survival in pediatric patients. Transfus. 2010, 50, 1359–1369. [Google Scholar] [CrossRef]
  180. Nair, S.; Vanathi, M.; Mahapatra, M.; Seth, T.; Kaur, J.; Velpandian, T.; Ravi, A.; Titiyal, J.S.; Tandon, R. Tear inflammatory mediators and protein in eyes of post allogenic hematopoeitic stem cell transplant patients. Ocul. Surf. 2018, 16, 352–367. [Google Scholar] [CrossRef] [PubMed]
  181. Cocho, L.; Fernandez, I.; Calonge, M.; Martínez, V.; González-García, M.J.; Caballero, L.; López-Corral, L.; García-Vázquez, C.; Vazquez, L.; Stern, M.E.; et al. Gene Expression–Based Predictive Models of Graft Versus Host Disease–Associated Dry Eye. Investig. Ophthalmol. Vis. Sci. 2015, 56, 4570. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  182. Yucebay, F. Tocilizumab as first-line therapy for steroid-refractory acute graft-versus-host-disease: Analysis of a single-center experience. Leuk. lymphoma 2019, 60, 2223–2229. [Google Scholar] [CrossRef]
  183. Kattner, A.-S.; Holler, E.; Holler, B.; Klobuch, S.; Weber, D.; Martinovic, D.; Edinger, M.; Herr, W.; Wolff, D. IL6-receptor antibody tocilizumab as salvage therapy in severe chronic graft-versus-host disease after allogeneic hematopoietic stem cell transplantation: A retrospective analysis. Ann. Hematol. 2020, 99, 847–853. [Google Scholar] [CrossRef] [Green Version]
  184. Smolen, J.S. EULAR recommendations for the management of rheumatoid arthritis with synthetic and biological disease-modifying antirheumatic drugs: 2019 update. Ann. Rheum. Dis. 2020, 79, 685–699. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Kheirkhah, A. A Pilot Randomized Trial on Safety and Efficacy of a Novel Topical Combined Inhibitor of Janus Kinase 1/3 and Spleen Tyrosine Kinase for GVHD-Associated Ocular Surface Disease. Cornea 2017, 36, 799–804. [Google Scholar] [CrossRef]
  186. Mukai, S.; Ogawa, Y.; Saya, H.; Kawakami, Y.; Tsubota, K. Therapeutic potential of tranilast for the treatment of chronic graft-versus-host disease in mice. PLoS ONE 2018, 13, e0203742. [Google Scholar] [CrossRef]
  187. Ogawa, Y.; He, H.; Mukai, S.; Imada, T.; Nakamura, S.; Su, C.-W.; Mahabole, M.; Tseng, S.C.G.; Tsubota, K. Heavy Chain-Hyaluronan/Pentraxin 3 from Amniotic Membrane Suppresses Inflammation and Scarring in Murine Lacrimal Gland and Conjunctiva of Chronic Graft-versus-Host Disease. Sci. Rep. 2017, 7, 42195. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. A variety of biological areas orchestrate in ocular GVHD, dry eye disease, which are representative of a manifestation of ocular GVHD and are related to various aspects of the study area including the tear film, microbiome, mucosal and exocrine gland immunity, stem cell biology on HSC or MSCs, acute and chronic inflammation, pathogenic fibrosis including fibroblasts origin, and stress-induced senescence. Those pathways are connected timely and spatially to each other and the development of ocular GVHD, MSC; mesenchymal stem cells, HSC; hematopoietic stem cells, EMT; epithelial mesenchymal transition.
Figure 1. A variety of biological areas orchestrate in ocular GVHD, dry eye disease, which are representative of a manifestation of ocular GVHD and are related to various aspects of the study area including the tear film, microbiome, mucosal and exocrine gland immunity, stem cell biology on HSC or MSCs, acute and chronic inflammation, pathogenic fibrosis including fibroblasts origin, and stress-induced senescence. Those pathways are connected timely and spatially to each other and the development of ocular GVHD, MSC; mesenchymal stem cells, HSC; hematopoietic stem cells, EMT; epithelial mesenchymal transition.
Ijms 22 06114 g001
Figure 2. Migration of senescent macrophages play a key role in ocular GVHD. A pro-inflammatory state related to the conditioning regimen, and previous acute GVHD, may lead to an accumulation of cell debris, resulting in the activation of RAS, ER stress, oxidative stress, and senescent cell involving SASP accumulation on the ocular surface and in the lacrimal gland. 8-OHdG; 8-hydroxy-2′-deoxyguanosine, 4-HNE; 4-hydroxy-2-nonenal, HEL; hexonoyl lesion, IL-6, interleukine-6, CXCL9; C-X-C motif ligand 9, OPN; osteopontin, CD; clustered of differentiation.
Figure 2. Migration of senescent macrophages play a key role in ocular GVHD. A pro-inflammatory state related to the conditioning regimen, and previous acute GVHD, may lead to an accumulation of cell debris, resulting in the activation of RAS, ER stress, oxidative stress, and senescent cell involving SASP accumulation on the ocular surface and in the lacrimal gland. 8-OHdG; 8-hydroxy-2′-deoxyguanosine, 4-HNE; 4-hydroxy-2-nonenal, HEL; hexonoyl lesion, IL-6, interleukine-6, CXCL9; C-X-C motif ligand 9, OPN; osteopontin, CD; clustered of differentiation.
Ijms 22 06114 g002
Table 1. Possible treatments/prophylaxis of ocular cGVHD at present and in the future [3,6,12,132,133,134,136,137].
Table 1. Possible treatments/prophylaxis of ocular cGVHD at present and in the future [3,6,12,132,133,134,136,137].
StrategyMedicine or Device
Retention of tear fluidPreservative-free artificial tear, Diquafosol [138], Punctal plug [139], Surgical punctal occlusion [140], Oral muscarinic agonists (pilocarpine [141], cevimeline [12]),
Mucin producing, tear film stabilizationDiquafosol [138], Rebamipide [142]
Reduction of inflammation and by-products Preservative-free corticosteroid [143], Cyclosporin [143], Tacrolimus [144,145,146], Tranilast, IL-1 receptor inhibitor, Lifitegrast, DNase [82], immunoglobulin [32], (Basic study; Heparin [81], Systemic vascular adhesion protein-1 inhibitor [111], Phenyl butyric acid [112], Senolytic agent [83,112]. Small molecule inhibitors of lymphocyte signaling [147], Anti-IL-6 receptor antagonist [83])
Epithelial supportHyaluronic acid, Autologeous sera [148,149], Cord blood sera [150,151], Platelet lysate [152,153], Amniotic membrane transplantation [4,154,155,156,157], Cultured epithelial cell transplantation, Medical use contact lenses [158,159,160,161].
Prevention of tear evaporationMoisture goggle, Tetracycline, Warm compression, Lid hygiene.
Inhibition of refractory ocular and systemic GVHD Systemic administration (Tacrolimus, Cyclosporin, Corticosteroid, Extracorporeal photopheresis, Rituximab, Sirolimus (Rapamycin), Mycophenolate mofetil) [6]
Reduction of fibrosis(Basic study, Angiotensin type I receptor antagonist [110], Vitamin A-coupled liposomes containing HSP47 siRNA [162])
Supportive careMoisture goggle, Prevention of infection (Doxycycline, Minocycline) [3], Maintenance of environmental factor [163]
Other surgical treatment [5,132]Epithelial cell debridement [5]
Surgical correction of entropion [132,164]
Partial tarsorrhaphy
Fornix reconstruction
Penetrating keratoplasty
Lamellar keratoplasty
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Ogawa, Y.; Kawakami, Y.; Tsubota, K. Cascade of Inflammatory, Fibrotic Processes, and Stress-Induced Senescence in Chronic GVHD-Related Dry Eye Disease. Int. J. Mol. Sci. 2021, 22, 6114. https://doi.org/10.3390/ijms22116114

AMA Style

Ogawa Y, Kawakami Y, Tsubota K. Cascade of Inflammatory, Fibrotic Processes, and Stress-Induced Senescence in Chronic GVHD-Related Dry Eye Disease. International Journal of Molecular Sciences. 2021; 22(11):6114. https://doi.org/10.3390/ijms22116114

Chicago/Turabian Style

Ogawa, Yoko, Yutaka Kawakami, and Kazuo Tsubota. 2021. "Cascade of Inflammatory, Fibrotic Processes, and Stress-Induced Senescence in Chronic GVHD-Related Dry Eye Disease" International Journal of Molecular Sciences 22, no. 11: 6114. https://doi.org/10.3390/ijms22116114

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop