Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Feb 1.
Published in final edited form as: Am J Transplant. 2019 Oct 30;20(2):389–398. doi: 10.1111/ajt.15631

Regulatory T cells promote corneal endothelial cell survival following transplantation via Interleukin-10

Giulia Coco 1,2, William Foulsham 1,3, Takeshi Nakao 1, Jia Yin 1, Afsaneh Amouzegar 1, Yukako Taketani 1, Sunil K Chauhan 1, Reza Dana 1
PMCID: PMC6984989  NIHMSID: NIHMS1054259  PMID: 31587452

Abstract

The functional competence of corneal endothelial cells (CEnCs) is critical for survival of corneal allografts, but these cells are often targets of the immune response mediated by graft-attacking effector T cells. Although regulatory T cells (Tregs) have been studied for their role in regulating host’s alloimmune response towards the graft, the cytoprotective function of these cells on CEnCs has not been investigated. The aim of this study was to determine whether Tregs suppress effector T cell-mediated and inflammatory cytokine-induced CEnC death, and to elucidate the mechanism by which this cytoprotection occurs. Using two well-established models of corneal transplantation (low-risk and high-risk models), we show that Tregs derived from low-risk graft recipients have a superior capacity in protecting CEnCs against effector T cell-mediated and IFN-γ and TNF-x-induced cell death compared to Tregs derived from high-risk hosts. We further demonstrate that the cytoprotective function of Tregs derived from low-risk hosts occurs independent of direct cell-cell contact and is mediated by the immunoregulatory cytokine IL-10. Our study is the first to report that Tregs provide cytoprotection for CEnCs through secretion of IL-10, indicating potentially novel therapeutic targets for enhancing CEnC survival following corneal transplantation.

1. Introduction

Corneal transplantation is the most common form of tissue grafting worldwide1. When performed on an uninflamed and non-vascularized host bed, the graft survival rate exceeds 90%2. However, inflammation and vascularization of the host bed disrupt the cornea’s relatively tolerant immune microenvironment, and graft survival rates fall to approximately 50% despite maximum immunosuppression in high-risk recipients35. Immune rejection is the leading cause of corneal graft failure6. Corneal endothelial cells (CEnCs) are the target of the effector immune response mediated primarily by graft-targeting CD4+IFNγ+ T helper type 1 (Th1) cells7,8. When the CEnC count decreases beyond a certain threshold, they are no longer capable of maintaining corneal graft transparency and the graft fails9,10.

Substantial progress has been made in determining the mechanisms by which host factors, such as recipient bed vascularity and inflammation influence the immune response to corneal allografts1115. Forkhead box protein 3 (Foxp3)-positive regulatory T cells (Tregs) are a subset of CD4+ T cells that play a critical role in maintaining immune tolerance16. Studies performed by our laboratory1724 and others2527 indicate the critical contribution of Foxp3+ Tregs to immune tolerance in the setting of corneal transplantation. Furthermore, studies have demonstrated that these cells exhibit marked phenotypic plasticity and functional adaptability according to cues they receive from their microenvironment17,2830. In the setting of corneal transplantation, we have demonstrated that Tregs derived from mice with inflamed and vascularized host beds that are prone to quick rejection (i.e. high risk hosts) have decreased expression of Foxp3 (the transcription factor that is crucial for their development and function) relative to low risk hosts that survive long term17,23,24. Furthermore, we have established that these Tregs derived from high-risk hosts have: (i) decreased ability to suppress graft-attacking CD4+CD25 effector T cells, (ii) decreased expression of CTLA-4 co-inhibitory molecule, (iii) higher expression of IFN-γ (a cytokine involved in graft rejection), and (iv) significantly diminished production of immunoregulatory cytokines such as IL-10 and transforming growth factor (TGF)-β17,23.

The mechanisms by which Tregs exert their immunosuppressive effects remain an area of intense investigation3138. In addition to modulating dendritic cell function, Tregs are known to suppress effector T cells through different mechanisms, including cytolysis, competition for metabolites, and release of soluble immunosuppressive cytokines such as IL-1018,38. Of note, there is evidence that IL-10 itself provides cytoprotection for hepatocytes, neurons and brain microvascular endothelial cells3942. However, the cytoprotective effects of Tregs or IL-10 on CEnCs have not been previously studied.

This study aimed to investigate the capacity of Tregs to suppress effector T cell-mediated and inflammatory cytokine-induced CEnC death. Furthermore, we evaluated whether the cytoprotective effects of Tregs are dependent upon the host immune microenvironment, i.e. whether the Treg cytoprotective effects are reduced in the high-risk setting, and determined whether direct cell-cell contact is necessary for such cytoprotection. Finally, we examined the contribution of the immunosuppressive cytokine IL-10 to Treg-mediated CEnC cytoprotection.

2. Materials and Methods

2.1. Animals

Six- to eight-week-old BALB/c and C57BL/6 male mice were purchased from Charles River Laboratories (Wilmington, MA). The study was approved by the Institutional Animal Care and Use Committee, and all animals were treated in accordance with the Association for Research in Vision and Ophthalmology Statement for the Use of Animals in Ophthalmic and Vision Research.

2.2. Orthotopic Corneal Transplantation

Two well-established models of allogeneic corneal transplantation were used, as described previously4346. For the low-risk model, C57BL/6 donor corneas were grafted onto normal BALB/c host beds. For the high-risk model, three intrastromal 11–0 sutures were placed in the central cornea of BALB/c recipient mice 14 days prior to transplantation to induce an inflamed and vascularized host bed. Mice exhibiting substantial corneal neovascularization at the time of transplant surgery were selected as high-risk graft recipients. A standard protocol for murine orthotopic corneal transplantation was followed, as described previously4346. Briefly, a 2-mm central corneal button was excised from donor C57BL/6 mice, grafted onto a 1.5-mm BALB/c recipient bed and secured with eight interrupted 11–0 nylon sutures. All procedures were performed under anesthesia with intraperitoneal injection of Ketamine (120 mg/kg) and Xylazine (20 mg/kg). Post-operatively, buprenorphine (0.05–0.1 mg/kg) was injected subcutaneously to minimize pain. Sutures were removed 7 days after surgery. Grafts that were opaque at postoperative week 2 were excluded from further analysis, as were eyes that underwent complications during or after surgery such as cataract, infection, intraocular hemorrhage or synechiae.

2.3. Cornea-in-the-cup Assay

In order to evaluate the cytoprotective function of Tregs derived from low-risk or high-risk hosts in the context of effector T cell-mediated and IFN-γ and TNF-α-induced CEnC death, a cornea-in-the-cup assay was used47. All cell types were harvested from draining cervical lymph nodes ipsilateral to the grafted eye 14 days after corneal transplantation, and purified using magnetic cell sorting (CD4+CD25+ Regulatory T cell Isolation kit, Miltenyi Biotec, Auburn, CA, USA) as per the manufacturer’s instructions. The purity of isolated cells was investigated by evaluating their Foxp3 expression using flow cytometry and CD4+CD25+Foxp3+ cell purity of >90% was confirmed in sorted cells. To examine the efficacy of Tregs from low-risk or high-risk hosts in suppressing effector T cell-mediated CEnC death, naïve C57BL/6 (graft donor) corneal cups were cultured in RPMI 1640 medium with 10% fetal bovine serum either (i) alone, (ii) with 100,000 effector T cells from low-risk BALB/c recipients or (iii) with 100,000 effector T cells from low-risk BALB/c recipients and 100,000 Tregs derived from either low-risk or high-risk graft recipients. To examine the protective function of Tregs derived from low-risk or high-risk hosts against inflammatory cytokine-induced CEnC death, naïve C57BL/6 corneal cups were cultured in RPMI 1640 medium with 10% fetal bovine serum either (i) alone, (ii) with a combination of recombinant mouse IFN-γ (100ng/mL; Biolegend) and TNF-α (100ng/mL; Biolegend, Recombinant mouse TNF-α, carrier-free) or (iii) with a combination of IFN-γ and TNF-α and Tregs derived from either low-risk or high-risk graft recipients.

To determine whether Tregs were able to protect CEnCs in a non-contact-dependent manner, the same experimental approach was performed using a 1-μm pore size Transwell cell culture insert (Sigma Aldrich, HST Transwell-96 Permeable support with 1.0-μm pore polyester membrane, TC-treated, sterile). Naïve C57BL/6 corneal cups in RPMI 1640 medium with 10% fetal bovine serum and IFN-γ (100ng/mL) and TNF-α (100ng/mL) were placed in the bottom chamber while Tregs (derived from low-risk or high-risk hosts) were placed in the upper chamber.

To control for the possibility of Tregs acting as a sink38, 48, 49 for inflammatory cytokines, Tregs derived from low-risk or high-risk graft recipients were stimulated in anti-CD3ε precoated plates (10 μg/mL; Biolegend, LEAF™ purified anti-mouse CD3ε, clone 145–2C11, for 3 hours at 37°C, then washed three times with PBS) with IL-2 (4ng/mL, Recombinant mouse IL-2 carrier-free, Biolegend) for 24 hours at 37°C. The supernatant of stimulated Tregs was collected and cultured with naïve C57BL/6 corneal cups in RPMI 1640 medium with 10% fetal bovine serum in the presence of IFN-γ (100ng/mL) and TNF-α (100ng/mL).

To determine whether IL-10 antagonism interferes with the suppressive effect of Tregs derived from low-risk hosts on inflammatory cytokine-induced CEnC death, an anti-IL-10 antibody was used in the co-culture (1μg/mL; Mouse IL-10 Antibody, Monoclonal Rat IgG1 clone #JES052A5, R&D System). Naïve C57BL/6 corneal cups were cultured in RPMI 1640 medium with 10% fetal bovine serum either (i) alone, (ii) with a combination of IFN-γ and TNF-α, (iii) with IFN-γ, TNF-α and Tregs from low-risk hosts or (iv) with IFN-γ, TNF-α, Tregs from low-risk hosts and anti IL-10 antibodies (1μg/mL). To test the hypothesis that IL-10 can restore the impaired function of Tregs from high-risk hosts in suppressing inflammatory cytokine-induced-CEnC death, we added recombinant IL-10 (10 μg/mL, BioLegend) to cultures with naïve C57BL/6 corneal cups, IFN-γ and TNF-α and Tregs from high-risk hosts. Finally, to examine the direct role of IL-10 on inflammatory cytokine-induced CEnC death independent of Tregs, we added recombinant IL-10 (10 μg/mL) to the naïve C57BL/6 corneal cups cultured with IFN-γ and TNF-α. All cultures were incubated for 24 hours in 5% CO2 at 37°. Experiments were performed three times with 5–6 corneas in each group.

2.4. Immunohistochemistry

To determine CEnC death, corneal cups were stained and analyzed by confocal microscopy. After incubation, corneal cups were washed in PBS and then fixed in absolute ethanol for 20 minutes in 96-well plates at room temperature. Fixed corneas were then washed in PBS (3x), incubated in 5% bovine serum albumin for 60 min at room temperature and stained with a FITC-conjugated zonula occludens-1 (ZO-1) monoclonal antibody (1:200; ThermoFisher Scientific) overnight at 4°C in the dark. After another washing, corneas were permeabilized with 0.1% Triton X in 0.1% sodium citrate for 10 min at room temperature. Terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) was performed using the In Situ Cell Death Detection Kit, TMR red (Sigma Aldrich) for 60 minutes in humidified atmosphere at 37°C in the dark. To evaluate the expression of IL-10RA by naïve CEnCs and inflammatory cytokine-stimulated CEnCs, after fixation corneas were stained with a primary antibody for IL-10RA (Anti-IL10RA antibody, ab225820, Abcam) and a FITC-conjugated secondary antibody (Goat anti-Rabbit IgG [H+L] Cross-Adsorbed Secondary Antibody, FITC; Invitrogen, Thermofisher). The control corneas were stained with secondary antibody only. Corneas were mounted on glass slides with the endothelium facing up, cut on four sides to flatten them, and embedded with DAPI-containing mounting medium. Flat mount corneas were analyzed using a confocal microscope (x400 magnification; Leica TCS-SP2, Leica, Wetzlar, Germany) and two images were taken from the central area of each cornea.

2.5. Image analysis

Live cells were defined as ZO-1-positive TUNEL-negative cells. ZO-1 staining (rather than DAPI) was used to determine live CEnCs to avoid staining of other cell types such as immune cells and keratocytes, as described previously47. Apoptotic cells were defined as ZO-1-positive and TUNEL-positive. In each experiment, the number of live cells in the naïve C57BL/6 corneal cups cultured in medium alone was set as the reference. The number of live cells in each sample was then divided by the reference number, and expressed as a percentage of live cells. Percentage of cell death was derived from the percentage of live cells as per the following equation: % cell death = (100-% alive cells). The percentage of suppressive function was calculated per the following equation: % suppression of CEnC death = (1-% cell deathi / % cell deathr) x100, where i= the group analyzed and r=reference group47. A masked observer was responsible for capturing images by confocal microscopy and counting cells using the ImageJ software.

2.6. Real Time PCR

Corneas were collected in Trizol (Ambion, Life Technologies, Grand Island, NY), and then homogenized on ice. Briefly, the RNA was extracted and purified by using RNeasy Micro kit (Qiagen, Valencia, CA). Total RNA was reverse transcripted using a Superscript III kit (Invitrogen, Carlsbad, CA). Real-time PCR was performed using TaqMan Universal PCR Mastermix (Applied Biosystems, Foster City, CA) and preformulated primers for IL-10RA (ThermoFisher Scientific, Mm00434147/ Mm00434151_m1) and Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) (ThermoFisher Scientific, Mm99999915_g1).

2.7. Statistical analysis

The ANOVA test for multiple comparisons was used to determine differences among groups. Mann-Whitney U test was then used to compare both percentage of CEnC death and percentage of suppression of CEnC death between each pair of groups. Data are presented as the mean ± standard error of mean (SEM). P <0.05 was considered statistically significant.

3. Results

3.1. Regulatory T cells suppress effector T cell-mediated corneal endothelial cell death

Naïve C57BL/6 corneal cups were cultured in medium alone, with effector T cells from low-risk BALB/c recipients, or with effector T cells cells and Tregs derived from either low-risk or high-risk hosts. Effector T cells alone induced 45.1±4.3% CEnC death. The percentage of cell death was reduced when either Tregs from low-risk grafted hosts (16.5±1.5%; p<0.001) or from high-risk grafted hosts (32.2±2.8%; p=0.034) were added to the culture (Fig. 1A and 1B). However, Tregs derived from low-risk hosts exerted a more significant suppression in Teff-induced CEnC death compared to Tregs derived from high-risk hosts (63.3±3.3% vs. 28.5±6.2% suppression, p<0.001).

Figure 1. Regulatory T cells suppress effector T cell-mediated corneal endothelial cell death.

Figure 1.

Naïve C57BL/6 corneal cups were cultured with 1×105 effector T cells (Teff) from low-risk BALB/c recipients, and with 1×105 regulatory T cells (Tregs) derived from either low-risk or high-risk transplant recipients. Teff and Tregs were isolated from the draining lymph nodes (dLNs) of low-risk or high-risk recipients at day 14 post-transplantation. (A) Corneas were fixed and stained with ZO-1 (Green) and TUNEL (Red), and were analyzed using confocal microscope (X400 magnification; Scale bar=50μm) (B) Bar graph showing percentage of corneal endothelial cell (CEnC) death in different groups. The number of live cells in the group in which naïve C57BL/6 corneal cups were cultured alone served as the reference. The number of live cells in each of the remaining groups was divided by the reference number, and expressed as a percentage of live cells. % CEnC death was derived from the percentage of live cells, as explained in the Methods section. Tregs from low-risk hosts and, to a lesser extent, Tregs from high-risk hosts were able to suppress Teff mediated-CEnC death (p<0.001 and p=0.03, respectively). Data are presented as mean ± SEM of three independent experiments (n= 4–6/group). Mann-Whitney Test. * p<0.05, *** p<0.001.

3.2. Regulatory T cells from low-risk graft recipients suppress IFN-γ and TNF-α-induced corneal endothelial cell death

To determine the capacity of Tregs derived from low-risk or high-risk hosts to suppress CEnC death induced by inflammatory cytokines, naïve C57BL/6 corneal cups were cultured in the presence of IFN-γ (100ng/mL) and TNF-α (100ng/mL) with or without Tregs derived from either low-risk or high-risk graft recipients. As shown in Fig. 2A and 2B, the combination of IFN-γ and TNF-α induced 50.6±1.8% CEnC death. Tregs derived from low-risk hosts significantly reduced CEnC death induced by IFN-γ and TNF-α by 41.8±5.2% (p<0.001). However, percentage of dead CEnCs in cultures containing Tregs derived from high-risk recipients did not differ significantly from the number of dead CEnCs induced by IFN-γ and TNF-α (Fig. 2B), demonstrating a significant difference in the capacity of Tregs to suppress CEnC death based on whether they were derived from low-risk or high-risk graft recipients.

Figure 2. Regulatory T cells derived from low-risk hosts suppress inflammatory cytokine-induced corneal endothelial cell death.

Figure 2.

Naïve C57BL/6 corneal cups were cultured with IFN-γ (100ng/mL) and TNF-α (100ng/mL), as well as 1×105 Tregs from either low-risk (LR) or high-risk (HR) hosts. (A) Corneas were fixed and stained with ZO-1 (Green) and TUNEL (Red), and were analyzed using confocal microscope (X400 magnification; Scale bar=50μm) (B) Bar graph showing the percentage of corneal endothelial cell (CEnC) death in different groups. The number of live cells in naïve C57BL/6 corneal cups cultured alone was set as the reference. The number of live cells in each of the other samples was divided by the reference number and expressed as a percentage of live cells. % CEnC death was derived from the percentage of live cells, as explained in the Methods section. Tregs derived from low-risk hosts suppressed inflammatory cytokine-induced CEnC death (p<0.001) in contrast to Tregs from high-risk hosts (p=0.61). Data are presented as mean ± SEM of three independent experiments (n= 4–6/group). Mann-Whitney Test. * p<0.05, ***p<0.001, ns: not significant.

3.3. Regulatory T cells are able to protect corneal endothelial cells in a non-contact dependent manner

Next to determine whether Tregs suppress inflammatory cytokine-induced CEnC death in a cell-cell contact dependent manner or through secretory factors, cornea cups were separated from Tregs in the culture using Transwell cell culture inserts. Our results demonstrated that Tregs derived from low-risk hosts retained their protective effect on CEnC when separated from the cornea cups by a Transwell insert and reduced CEnC death by 31.4±7.4% (p=0.002). However, Tregs derived from high-risk hosts failed to exert a significant suppressive effect on CEnC death induced by IFN-γ and TNF-α (Fig. 3A). Of note, the efficacy of Tregs derived from low-risk hosts in reducing IFN-γ and TNF-α-induced CEnC death did not differ significantly in the Transwell and non-Transwell culture systems (% suppression of CEnC death of 41.8±5.2% and 31.4±7.4%, respectively; p=0.221; Fig. 3B), indicating that Tregs derived from low-risk graft recipients exert their cytoprotective function through soluble factors and independent of cell-cell contact.

Figure 3. The protective effect of regulatory T-cells on corneal endothelial cells does not depend on direct cell-to-cell contact.

Figure 3.

Naïve C57BL/6 corneal cups treated with IFN-γ (100ng/mL) and TNF-α (100ng/mL) were placed in the lower chamber of a Transwell insert (1-μm pore size) and cultured with 1×105 Tregs derived from either low-risk (LR) or high-risk (HR) hosts that were placed in the upper chamber. (A) Bar graph showing the percentage of corneal endothelial cell (CEnC) death in different groups. The number of live cells in the naïve C57BL/6 corneal cups cultured alone was set as the reference. The number of live cells in each of the other samples was divided by the reference number, and expressed as the percentage of live cells. % CEnC death was derived from the percentage of live cells, as explained in the Methods section. Tregs from low-risk hosts maintained their ability to suppress CEnC death when separated from corneas by Transwell inserts (p=0.002). (B) Bar graph showing comparable capacity of Tregs in suppressing inflammatory cytokine-induced CEnC death in direct co-culture and Transwell culture systems (p=0.22). (C) Representative confocal images of corneas cultured with IFN-γ and TNF-α with or without Tregs in the Transwell culture system (ZO-1 [Green] and TUNEL [Red]; X400 magnification; Scale bar=50μm). (D) 1×105 Tregs were stimulated in anti-CD3ε precoated plates with IL-2 (4ng/mL) for 24 hours at 37°C, and the supernatant of stimulated Tregs was then collected and used instead of Tregs in the culture system. Bar graph demonstrates the percentage of CEnC death in different groups. The supernatant of Tregs from low-risk hosts was significantly more effective in suppressing inflammatory cytokine-induced CEnC death compared to the supernatant from their high-risk counterparts (p<0.001). Data are presented as mean ± SEM of three independent experiments (n= 4–6/group). Mann-Whitney Test. * p<0.05, ** p<0.01, *** p<0.001, ns: not significant.

To control for the possibility of cells binding to the inflammatory cytokines in the culture and thereby suppressing their bioavailability (e.g. a “sink” mechanism), we substituted Tregs with the supernatant of anti-CD3ε-stimulated Tregs. Our data demonstrated that the supernatants of both Tregs derived from low-risk and high-risk hosts were effective in suppressing IFN-γ and TNF-α-induced CEnC death. While we observed 34.2±2.8% CEnC death in corneas cultured in the presence of IFN-γ and TNF-α, addition of supernatants of stimulated Tregs derived from low-risk and high-risk hosts reduced CEnC death to 11.0±1.6% (p=0.001) and 23.0±2.8 (p=0.020), respectively (Fig. 3D). Similar to our previous observations, Tregs derived from low-risk graft recipients demonstrated a significantly greater capacity in suppressing CEnC death (67.7±4.8%) as compared to Tregs derived from high-risk hosts (32.7±8.3%; p=0.004).

3.4. IL-10 is a key factor in suppressing IFN-γ and TNF-α-induced corneal endothelial cell death

IL-10 is a key regulatory factor secreted by Tregs, which mediates the alloantigen-specific function of Tregs. Previous work from our laboratory has demonstrated that Tregs derived from high-risk graft recipients secrete significantly lower levels of IL-10 compared to their counterparts derived from low-risk hosts23. We thus next investigated the role of IL-10 in mediating the cytoprotective function of Tregs. We first confirmed the expression of the IL-10 receptor by murine CEnCs using qPCR (data not shown) and confocal microscopy (Fig. 4A). Next, to determine the role of IL-10 in Treg-mediated suppression of CEnC death, IL-10 neutralizing antibody was added to the cornea cups cultured with Tregs derived from low-risk hosts in the presence of IFN-γ and TNF-α. Our results demonstrated that IL-10 neutralization completely abrogated the efficacy of Tregs from low-risk hosts in suppressing IFN-γ and TNF-α-induced CEnC death (61% vs. 2% [with IL-10 blockade] suppression in CEnC death; p<0.001; Fig. 4B). Since we consistently observed a lower cytoprotective function by Tregs derived from high-risk hosts, we next set to determine whether addition of IL-10 could restore the suppressive function of these Tregs against IFN-γ and TNF-α-induced CEnC death. To this aim, recombinant IL-10 was added to the culture of cornea cups with Tregs derived from high-risk hosts in the presence of IFN-γ and TNF-α. While Tregs derived from high-risk hosts did not exert a protective effect against CEnC death (p=0.953), as we observed earlier, supplementation of the culture with IL-10 significantly improved their capacity to suppress CEnC death resulting from exposure to IFN-γ and TNF-α (p<0.001; Fig. 4C).

Figure 4. IL-10 is a key factor in suppressing inflammatory cytokine-induced corneal endothelial cell death.

Figure 4.

(A) Confocal images showing IL-10 receptor (IL-10R) expression in [A] isotype control, [B] naïve C57BL/6 corneal cups and [C] naïve C57BL/6 corneal cups cultured with IFNγ (100ng/mL)+TNFα (100ng/mL) for 24 hours at 37°C and 5%CO2. Corneal endothelial cells (CEnCs) are shown to express IL-10R (green), expression of which is increased after exposure to inflammatory stimuli. (B) Naïve C57BL/6 corneal cups were cultured with Tregs from low-risk hosts in the presence of IFN-γ (100ng/mL) and TNF-α (100ng/mL) with or without anti-IL-10 (1μg/mL) antibody. Bar graph showing percentage of CEnC death in different groups. The number of live cells in the naïve C57BL/6 corneal cups cultured alone was set as the reference. The number of live cells in each of the other samples was divided by the reference number and expressed as a percentage of live cells. %CEnC death was derived from the percentage of live cells, as explained in the Methods section. The capacity of Tregs from low-risk hosts to suppress CEnC death was abrogated by neutralization of IL-10 in the culture (p<0.001). (C) Naïve C57BL/6 corneal cups were cultured with Tregs from high-risk hosts in the presence of IFN-γ (100ng/mL) and TNF-α (100ng/mL) with or without recombinant IL-10 (10μg/mL). Bar graph showing the percentage of CEnC death in different groups. Addition of IL-10 potentiated the cytoprotective function of dysfunctional Tregs derived from high-risk hosts on CEnCs (p<0.001). (D) Naïve C57BL/6 corneal cups were directly cultured with or without IL-10 (10μg/mL) in the presence of IFN-γ (100ng/mL) and TNF-α (100ng/mL). Bar graph demonstrates the capacity of IL-10 alone in suppressing inflammatory cytokine-induced CEnC death (p<0.001). Data are presented as mean ± SEM of three independent experiments (n= 4–6/group). Mann-Whitney Test. *p<0.05, **p<0.01, ***p<0.001, ns: not significant.

Finally, to evaluate the direct cytoprotective effects of IL-10 on CEnCs in the absence of Tregs, IL-10 was added to naïve corneal cups cultured with IFN-γ and TNF-α, and CEnC death was determined. Our data demonstrated that IL-10 alone was effective in reducing IFN-γ and TNF-α-induced CEnC death (22.1±2.5% CEnC death with IL-10 added compared to 44.9±3.4% without IL-10; p<0.001; Fig. 4D), indicating the central role of IL-10 in protecting CEnCs from death induced by inflammatory cytokines.

4. Discussion

Tregs have a critical function in suppressing alloimmune responses in the setting of tissue transplantation21,49,50. However, the functional status of Tregs demonstrates significant plasticity, and is determined in part by cues from their microenvironment29,51. In this study, we investigated the mechanisms by which Tregs suppress effector T cell-mediated and inflammatory cytokine-induced CEnC death, and evaluated whether these effects were abrogated in Tregs derived from high-risk graft recipients. Our data show that Tregs from low-risk hosts are able to protect CEnCs against both effector T cell-mediated and IFN-γ and TNF-α-induced cell death, and that this function is significantly compromised in Tregs derived from high-risk hosts. Furthermore, we demonstrate that the cytoprotective function of Tregs occurs irrespective of direct cell-cell contact and is mediated by the immunomodulatory cytokine IL-10.

Host alloimmunity is the principal cause of donor CEnC loss after corneal transplantation52. Although multiple alloimmune mechanisms, including humoral antibody-mediated CEnC loss53 and CD8+ T cell cytotoxicity54 have been implicated in CEnC death following transplantation, the principal effector cells mediating CEnC loss have been shown to be CD4+ T helper 1 (Th1) cells7,8,52. Th1 cells are notable for their secretion of signature cytokine IFN-γ, as well as other inflammatory cytokines such as TNF-α and IL-155. These pro-inflammatory cytokines have also been shown to cause CEnC death independent of Th1 cells56. Our data demonstrate that Tregs are capable of rescuing CEnC from effector T cell-mediated death in vitro. Indeed, we show that both Tregs from low-risk and high-risk hosts are effective in protecting CEnCs, with a greater cytoprotective capacity demonstrated by Tregs derived from low-risk hosts. Previous work from our group has shown that Tregs derived from both low-risk and high-risk corneal allograft recipients effectively suppress the proliferation of effector CD4+CD25 T cells, although Tregs derived from low-risk hosts exhibit 20% more suppressive capacity relative to Tregs from high-risk hosts23. It is therefore conceivable that in these experiments both Treg populations are limiting CEnC loss through modulating effector T cell responses and not necessarily by exerting a direct effect on endothelial cells. In order to delineate whether Tregs have a direct cytoprotective effect on CEnCs or whether increased CEnC survival is due to modulation of the effector T cell response, we developed an in vitro system with only two cell types – CEnCs and Tregs. Previous work from our laboratory and others has shown that CEnC death can be induced by effector T cell-associated inflammatory cytokines, such as IFN-γ and TNF-α52,56. Thus, we added IFN-γ and TNF-α to our in vitro culture system to induce CEnC death. Our data show that Tregs from low-risk graft recipients, but not from high-risk hosts, suppress inflammatory cytokine-induced CEnC death. It has been shown that Tregs exert their immunoregulatory function via both contact-dependent and non-contact dependent mechanisms38. Thus, we employed a Transwell system to determine whether the cytoprotective effects of Tregs on CEnCs occurs in a contact-dependent manner or is mediated through soluble factors. We show that Tregs from low-risk hosts continue to exert their cytoprotective effects on CEnCs in the presence of Transwell inserts, indicating that the observed phenomenon is mediated through soluble factors rather than direct cell-cell contact.

We considered the possibility that Tregs were binding inflammatory cytokines in the culture (i.e. acting as a cytokine sink). Accordingly, we conducted experiments in which Tregs were removed from the culture and replaced by the supernatant of stimulated Tregs. Our results show that even in the absence of Tregs, the supernatant of stimulated Tregs exerts a protective function on CEnCs, further confirming that Treg promotion of CEnC survival is mediated through soluble factors. Interestingly, we observed that supernatant derived from both LR and HR Tregs were able to suppress CEnC death more than Tregs themselves. We believe that this finding is related to Tregs acting as a cytokine sink, reducing the amount of IL-10 or other immunoregulatory cytokines in the culture supernatant. However, by removing actual Tregs from the culture, and only using their supernatant, we circumvent the cells acting as cytokine sinks and focus on the function of their expressed cytokines.

In a previous study, our group reported that Tregs derived from low-risk allograft recipients secrete comparably higher levels of IL-10 compared to Tregs derived from high-risk hosts23. IL-10 has been shown to protect various cell types from biological activities induced by other cytokines through the activation of transcription factors57. This cytoprotective effect has been documented in a variety of conditions including diabetes58 and microvascular endothelial cell function in hypertension59, neuronal cells after brain infarction40 and hepatic cells after hepatic cold ischemia/reperfusion injury39. The observation of higher levels of IL-10 secreted by Tregs derived from low-risk hosts coupled with studies that have demonstrated the protective role of IL-10 prompted us to hypothesize that the promotion of CEnC survival by Tregs occurs through their secretion of IL-10. Using PCR and immunohistochemistry staining, we confirmed the expression of IL-10 receptor by murine CEnCs. Furthermore, we observed an upregulated expression of IL-10 receptor by CEnCs following exposure to inflammatory stimuli. Our data indicate that IL-10 plays a critical role in the cytoprotective function of Tregs, as neutralization of IL-10 completely abrogates the protective effects of Tregs from low-risk hosts on CEnC, while addition of IL-10 to cultures with less cytoprotective Tregs from high-risk hosts enhances their suppressive effect against CEnC death.

We note that IL-10 has been reported to expand Foxp3+ Tregs, to promote CTLA-4 expression by Tregs and increase their suppressive function60. In order to determine whether IL-10 was having a direct effect on CEnC survival, or whether IL-10 was in fact upregulating non-IL-10-mediated Treg cytoprotective mechanisms, we excluded Tregs from the co-culture system and cultured CEnCs directly with inflammatory cytokines and IL-10. Our data clearly indicate that IL-10 alone is effective in protecting CEnCs from inflammatory cytokine-induced cell death. Interestingly, previous studies have reported that CEnCs promote the generation of Tregs through their expression of CTLA-2α and TGF-β61,62. Our study provides evidence that these cellular interactions may in fact be bidirectional, with Tregs promoting CEnC survival by IL-10-mediated cytoprotection against inflammatory cytokine-induced death. However, these data shed further light on the diverse mechanisms by which regulatory mechanisms conspire to promote the observed high survival of corneal grafts, even without use of immunosuppressive medications.

Acknowledgments

This work was supported by the National Institutes of Health R01 EY12963 (to RD) and Core grant P30EY003790.

Abbreviations:

CEnCs

corneal endothelial cells

Foxp3

Forkhead box protein 3

IFN-γ

interferon-gamma

IL-10

Interleukin-10

Teff

effector T cell

Th1

T helper 1

TNF-α

Tumor necrosis factor-alpha

Treg

regulatory T cell

Footnotes

Disclosure

The authors of this manuscript have no conflicts of interest to disclose as described by the American Journal of Transplantation.

Data Availability Statement

The data that support the findings of this study are available from the corresponding author upon reasonable request.

References

  • 1.Gain P, Jullienne R, He Z, et al. Global Survey of Corneal Transplantation and Eye Banking. JAMA Ophthalmol. 2016;134(2):167. doi: 10.1001/jamaophthalmol.2015.4776. [DOI] [PubMed] [Google Scholar]
  • 2.Williams KA, Muehlberg SM, Lewis RF, Coster DJ. How successful is corneal transplantation? A report from the Australian corneal graft register. Eye. 1995;9(2):219–227. [DOI] [PubMed] [Google Scholar]
  • 3.Dana MR, Qian Y, Hamrah P. Twenty-five-year panorama of corneal immunology: emerging concepts in the immunopathogenesis of microbial keratitis, peripheral ulcerative keratitis, and corneal transplant rejection. Cornea. 2000;19(5):625–643. http://www.ncbi.nlm.nih.gov/pubmed/11009315. Accessed October 20, 2018. [DOI] [PubMed] [Google Scholar]
  • 4.Maguire MG, Stark WJ, Gottsch JD, et al. Risk Factors for Corneal Graft Failure and Rejection in the Collaborative Corneal Transplantation Studies. Ophthalmology. 1994;101(9):1536–1547. [DOI] [PubMed] [Google Scholar]
  • 5.Kamp MT, Fink NE, Enger C, Maguire MG, Stark WJ, Stulting RD. Patient-reported symptoms associated with graft reactions in high-risk patients in the collaborative corneal transplantation studies. Collaborative Corneal Transplantation Studies Research Group. Cornea. 1995;14(1):43–48. http://www.ncbi.nlm.nih.gov/pubmed/7712736. Accessed October 20, 2018. [PubMed] [Google Scholar]
  • 6.Coster DJ, Williams KA. The impact of corneal allograft rejection on the long-term outcome of corneal transplantation. Am J Ophthalmol. 2005;140(6):1112–1122. doi: 10.1016/j.ajo.2005.07.024. [DOI] [PubMed] [Google Scholar]
  • 7.Boisgérault F, Liu Y, Anosova N, et al. Role of CD4+ and CD8+ T cells in allorecognition: lessons from corneal transplantation. J Immunol. 2001;167(4):1891–1899. [DOI] [PubMed] [Google Scholar]
  • 8.Hegde S, Beauregard C, Mayhew E, Niederkorn JY. CD4(+) T-cell-mediated mechanisms of corneal allograft rejection: role of Fas-induced apoptosis. Transplantation. 2005;79(1):23–31. http://www.ncbi.nlm.nih.gov/pubmed/15714165. Accessed November 1, 2018. [DOI] [PubMed] [Google Scholar]
  • 9.Claerhout I, Beele H, Kestelyn P. Graft failure: I. Endothelial cell loss. Int Ophthalmol. 2008;28(3):165–173. doi: 10.1007/s10792-007-9087-0. [DOI] [PubMed] [Google Scholar]
  • 10.Patel SV, Hodge DO, Bourne WM. Corneal endothelium and postoperative outcomes 15 years after penetrating keratoplasty. Trans Am Ophthalmol Soc. 2004;102:57–65–6. http://www.ncbi.nlm.nih.gov/pubmed/15747745. Accessed October 20, 2018. [PMC free article] [PubMed] [Google Scholar]
  • 11.Liu Y, Hamrah P, Zhang Q, Taylor AW, Dana MR. Draining lymph nodes of corneal transplant hosts exhibit evidence for donor major histocompatibility complex (MHC) class II-positive dendritic cells derived from MHC class II-negative grafts. J Exp Med. 2002;195(2):259–268. http://www.ncbi.nlm.nih.gov/pubmed/11805152. Accessed October 21, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Huq S, Liu Y, Benichou G, Dana MR. Relevance of the direct pathway of sensitization in corneal transplantation is dictated by the graft bed microenvironment. J Immunol. 2004;173(7):4464–4469. http://www.ncbi.nlm.nih.gov/pubmed/15383577. Accessed October 21, 2018. [DOI] [PubMed] [Google Scholar]
  • 13.Hamrah P, Huq SO, Liu Y, Zhang Q, Dana MR. Corneal immunity is mediated by heterogeneous population of antigen-presenting cells. J Leukoc Biol. 2003;74(2):172–178. http://www.ncbi.nlm.nih.gov/pubmed/12885933. Accessed October 21, 2018. [DOI] [PubMed] [Google Scholar]
  • 14.Hamrah P, Liu Y, Zhang Q, et al. Alterations in corneal stromal dendritic cell phenotype and distribution in inflammation. 2003;121(8). doi: 10.1001/archopht.121.8.1132. [DOI] [PubMed] [Google Scholar]
  • 15.Dana MR. Angiogenesis and lymphangiogenesis-implications for corneal immunity. Semin Ophthalmol. 2006;21(1):19–22. doi: 10.1080/08820530500509358. [DOI] [PubMed] [Google Scholar]
  • 16.Sakaguchi S, Sakaguchi N, Asano M, Itoh M, Toda M. Immunologic self-tolerance maintained by activated T cells expressing IL-2 receptor alpha-chains (CD25). Breakdown of a single mechanism of self-tolerance causes various autoimmune diseases. J Immunol. 1995;155(3):1151–1164. http://www.ncbi.nlm.nih.gov/pubmed/7636184. Accessed October 31, 2018. [PubMed] [Google Scholar]
  • 17.Hua J, Inomata T, Chen Y, et al. Pathological conversion of regulatory T cells is associated with loss of allotolerance. Sci Rep. 2018;8(1):7059. doi: 10.1038/s41598-018-25384-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Foulsham W, Marmalidou A, Amouzegar A, Coco G, Chen Y, Dana R. The function of regulatory T cells at the ocular surface: Review. Ocul Surf. May 2017. doi: 10.1016/j.jtos.2017.05.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Tahvildari M, Omoto M, Chen Y, et al. In Vivo Expansion of Regulatory T Cells by Low-Dose Interleukin-2 Treatment Increases Allograft Survival in Corneal Transplantation. Transplantation. 2016;100(3):525–532. doi: 10.1097/TP.0000000000001044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Shao C, Chen Y, Nakao T, et al. Local Delivery of Regulatory T Cells Promotes Corneal Allograft Survival. Transplantation. September 2018:1. doi: 10.1097/TP.0000000000002442. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Chauhan SK, Saban DR, Dohlman TH, Dana R. CCL-21 conditioned regulatory T cells induce allotolerance through enhanced homing to lymphoid tissue. J Immunol. 2014;192(2):817–823. doi: 10.4049/jimmunol.1203469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Jin Y, Chauhan SK, Saban DR, Dana R. Role of CCR7 in facilitating direct allosensitization and regulatory T-cell function in high-risk corneal transplantation. Invest Ophthalmol Vis Sci. 2010;51(2):816–821. doi: 10.1167/iovs.09-3952. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Inomata T, Hua J, Di Zazzo A, Dana R. Impaired Function of Peripherally Induced Regulatory T Cells in Hosts at High Risk of Graft Rejection. Sci Rep. 2016;6(1):39924. doi: 10.1038/srep39924. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Chauhan SK, Saban DR, Lee HK, Dana R. Levels of Foxp3 in regulatory T cells reflect their functional status in transplantation. J Immunol. 2009;182(1):148–153. http://www.ncbi.nlm.nih.gov/pubmed/19109145. Accessed October 20, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Cunnusamy K, Chen PW, Niederkorn JY. IL-17A-Dependent CD4+CD25+ Regulatory T Cells Promote Immune Privilege of Corneal Allografts. J Immunol. 2011;186(12):6737–6745. doi: 10.4049/jimmunol.1100101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Cunnusamy K, Niederkorn JY. IFN-γ blocks CD4+CD25+ Tregs and abolishes immune privilege of minor histocompatibility mismatched corneal allografts. Am J Transplant. 2013;13(12):3076–3084. doi: 10.1111/ajt.12466. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Hildebrand A, Jarsch C, Kern Y, Böhringer D, Reinhard T, Schwartzkopff J. Subconjunctivally applied naïve Tregs support corneal graft survival in baby rats. Mol Vis. 2014;20:1749–1757. [PMC free article] [PubMed] [Google Scholar]
  • 28.DuPage M, Bluestone JA. Harnessing the plasticity of CD4(+) T cells to treat immune-mediated disease. Nat Rev Immunol. 2016;16(3):149–163. doi: 10.1038/nri.2015.18. [DOI] [PubMed] [Google Scholar]
  • 29.Sawant DV, Vignali DAA. Once a Treg, always a Treg? Immunol Rev. 2014;259(1):173–191. doi: 10.1111/imr.12173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Campbell DJ. Control of Regulatory T Cell Migration, Function, and Homeostasis. J Immunol. 2015;195(6):2507–2513. doi: 10.4049/jimmunol.1500801. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Takahashi T, Kuniyasu Y, Toda M, et al. Immunologic self-tolerance maintained by CD25+CD4+ naturally anergic and suppressive T cells: induction of autoimmune disease by breaking their anergic/suppressive state. Int Immunol. 1998;10(12):1969–1980. http://www.ncbi.nlm.nih.gov/pubmed/9885918. Accessed November 3, 2018. [DOI] [PubMed] [Google Scholar]
  • 32.Thornton AM, Shevach EM. CD4+CD25+ immunoregulatory T cells suppress polyclonal T cell activation in vitro by inhibiting interleukin 2 production. J Exp Med. 1998;188(2):287–296. http://www.ncbi.nlm.nih.gov/pubmed/9670041. Accessed November 3, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Gondek DC, Lu L-F, Quezada SA, Sakaguchi S, Noelle RJ. Cutting edge: contact-mediated suppression by CD4+CD25+ regulatory cells involves a granzyme B-dependent, perforin-independent mechanism. J Immunol. 2005;174(4):1783–1786. http://www.ncbi.nlm.nih.gov/pubmed/15699103. Accessed November 3, 2018. [DOI] [PubMed] [Google Scholar]
  • 34.Bopp T, Becker C, Klein M, et al. Cyclic adenosine monophosphate is a key component of regulatory T cell-mediated suppression. J Exp Med. 2007;204(6):1303–1310. doi: 10.1084/jem.20062129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Deaglio S, Dwyer KM, Gao W, et al. Adenosine generation catalyzed by CD39 and CD73 expressed on regulatory T cells mediates immune suppression. J Exp Med. 2007;204(6):1257–1265. doi: 10.1084/jem.20062512. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Paust S, Lu L, McCarty N, Cantor H. Engagement of B7 on effector T cells by regulatory T cells prevents autoimmune disease. Proc Natl Acad Sci U S A. 2004;101(28):10398–10403. doi: 10.1073/pnas.0403342101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Oderup C, Cederbom L, Makowska A, Cilio CM, Ivars F. Cytotoxic T lymphocyte antigen-4-dependent down-modulation of costimulatory molecules on dendritic cells in CD4+ CD25+ regulatory T-cell-mediated suppression. Immunology. 2006;118(2):240–249. doi: 10.1111/j.1365-2567.2006.02362.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Vignali DAA, Collison LW, Workman CJ. How regulatory T cells work. Nat Rev Immunol. 2008;8(7):523–532. doi: 10.1038/nri2343. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Li J, Qi H, He Z, et al. Cytoprotective effects of human interleukin-10 gene transfer against necrosis and apoptosis induced by hepatic cold ischemia/reperfusion injury. J Surg Res. 2009;157(1):e71–8. doi: 10.1016/j.jss.2009.03.004. [DOI] [PubMed] [Google Scholar]
  • 40.Ooboshi H, Ibayashi S, Shichita T, et al. Postischemic gene transfer of interleukin-10 protects against both focal and global brain ischemia. Circulation. 2005;111(7):913–919. doi: 10.1161/01.CIR.0000155622.68580.DC. [DOI] [PubMed] [Google Scholar]
  • 41.Lin R, Chen F, Wen S, Teng T, Pan Y, Huang H. Interleukin-10 attenuates impairment of the blood-brain barrier in a severe acute pancreatitis rat model. J Inflamm. 2018;15(1):4. doi: 10.1186/s12950-018-0180-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Yue Y, Garikipati VNS, Verma SK, Goukassian DA, Kishore R. Interleukin-10 Deficiency Impairs Reparative Properties of Bone Marrow-Derived Endothelial Progenitor Cell Exosomes. Tissue Eng Part A. 2017;23(21–22):1241–1250. doi: 10.1089/ten.TEA.2017.0084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Inomata T, Mashaghi A, Di Zazzo A, Dana R. Ocular surgical models for immune and angiogenic responses. J Biol methods. 2015;2(3):27. doi: 10.14440/jbm.2015.78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Inomata T, Mashaghi A, Di Zazzo A, Lee S-M, Chiang H, Dana R. Kinetics of Angiogenic Responses in Corneal Transplantation. Cornea. 2017;36(4):491–496. doi: 10.1097/ICO.0000000000001127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Tahvildari M, Emami-Naeini P, Omoto M, Mashaghi A, Chauhan SK, Dana R. Treatment of donor corneal tissue with immunomodulatory cytokines: a novel strategy to promote graft survival in high-risk corneal transplantation. Sci Rep. 2017;7(1):971. doi: 10.1038/s41598-017-01065-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Dohlman TH, Omoto M, Hua J, et al. VEGF-trap Aflibercept Significantly Improves Long-term Graft Survival in High-risk Corneal Transplantation. Transplantation. 2015;99(4):678–686. doi: 10.1097/TP.0000000000000512. [DOI] [PubMed] [Google Scholar]
  • 47.Satitpitakul V, Sun Z, Suri K, et al. Vasoactive Intestinal Peptide Promotes Corneal Allograft Survival. Am J Pathol. 2018;188(9):2016–2024. doi: 10.1016/j.ajpath.2018.05.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Vignali DAA. Mechanisms of Treg suppression: still a long way to go. Frontiers in Immunol. 2012;3(191):1–3. 10.3389/fimmu.2012.00191 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Gupta S. Immune homeostasis: regulatory T cells (Treg) and molecules. J Clin Immunol. 2008;28(6):617–618. doi: 10.1007/s10875-008-9259-3. [DOI] [PubMed] [Google Scholar]
  • 50.Wood KJ, Sakaguchi S. Regulatory Lymphocytes: Regulatory T cells in transplantation tolerance. Nat Rev Immunol. 2003;3(3):199–210. doi: 10.1038/nri1027. [DOI] [PubMed] [Google Scholar]
  • 51.Sakaguchi S, Vignali DAA, Rudensky AY, Niec RE, Waldmann H. The plasticity and stability of regulatory T cells. Nat Rev Immunol. 2013;13(6):461–467. doi: 10.1038/nri3464. [DOI] [PubMed] [Google Scholar]
  • 52.Chauhan SK, Jurkunas U, Funaki T, Dastjerdi M, Dana R. Quantification of allospecific and nonspecific corneal endothelial cell damage after corneal transplantation. Eye. 2015;29(1):136–144. doi: 10.1038/eye.2014.248. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Hargrave SL, Mayhew E, Hegde S, Niederkorn J. Are corneal cells susceptible to antibody-mediated killing in corneal allograft rejection? Transpl Immunol. 2003;11(1):79–89. doi: 10.1016/S0966-3274(02)00082-5. [DOI] [PubMed] [Google Scholar]
  • 54.Niederkorn JY, Stevens C, Mellon J, Mayhew E. Differential roles of CD8+ and CD8- T lymphocytes in corneal allograft rejection in “high-risk” hosts. Am J Transplant. 2006;6(4):705–713. doi: 10.1111/j.1600-6143.2006.01237.x. [DOI] [PubMed] [Google Scholar]
  • 55.Luckheeram RV, Zhou R, Verma AD, Xia B. CD4+T cells: differentiation and functions. Clin Dev Immunol. 2012;2012:925135. doi: 10.1155/2012/925135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Sagoo P, Chan G, Larkin DFP, George AJT. Inflammatory cytokines induce apoptosis of corneal endothelium through nitric oxide. Invest Ophthalmol Vis Sci. 2004;45(11):3964–3973. doi: 10.1167/iovs.04-0439. [DOI] [PubMed] [Google Scholar]
  • 57.Pestka S, Krause CD, Sarkar D, Walter MR, Shi Y, Fisher PB. Interleukin-10 and Related Cytokines and Receptors. Annu Rev Immunol. 2004;22(1):929–979. doi: 10.1146/annurev.immunol.22.012703.104622. [DOI] [PubMed] [Google Scholar]
  • 58.Gunnett CA, Heistad DD, Faraci FM. Interleukin-10 protects nitric oxide-dependent relaxation during diabetes: role of superoxide. Diabetes. 2002;51(6):1931–1937. http://www.ncbi.nlm.nih.gov/pubmed/12031983. Accessed February 28, 2019. [DOI] [PubMed] [Google Scholar]
  • 59.Kassan M, Galan M, Partyka M, Trebak M, Matrougui K. Interleukin-10 Released by CD4(+)CD25(+) Natural Regulatory T Cells Improves Microvascular Endothelial Function Through Inhibition of NADPH Oxidase Activity in Hypertensive Mice. Arterioscler Thromb Vasc Biol. 2011;31(11):2534–2542. doi: 10.1161/ATVBAHA.111.233262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Hsu P, Santner-Nanan B, Hu M, et al. IL-10 Potentiates Differentiation of Human Induced Regulatory T Cells via STAT3 and Foxo1. J Immunol. 2015;195(8):3665–3674. doi: 10.4049/jimmunol.1402898. [DOI] [PubMed] [Google Scholar]
  • 61.Sugita S, Yamada Y, Horie S, et al. Induction of T regulatory cells by cytotoxic T-lymphocyte antigen-2α on corneal endothelial cells. Invest Ophthalmol Vis Sci. 2011;52(5):2598–2605. doi: 10.1167/iovs.10-6322. [DOI] [PubMed] [Google Scholar]
  • 62.Yamada Y, Sugita S, Horie S, Yamagami S, Mochizuki M. Mechanisms of Immune Suppression for CD8 + T Cells by Human Corneal Endothelial Cells via Membrane-Bound TGFβ. Investig Opthalmology Vis Sci. 2010;51(5):2548. doi: 10.1167/iovs.09-4233. [DOI] [PubMed] [Google Scholar]

RESOURCES