Anda di halaman 1dari 10

Acta Biomaterialia 71 (2018) 261–270

Contents lists available at ScienceDirect

Acta Biomaterialia
journal homepage: www.elsevier.com/locate/actabiomat

Full length article

Controlled release of an HDAC inhibitor for reduction of inflammation in


dry eye disease
Michelle L. Ratay a, Stephen C. Balmert a, Ethan J. Bassin c, Steven R. Little a,b,c,d,e,⇑
a
Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, United States
b
Department of Chemical Engineering, University of Pittsburgh, Pittsburgh, PA 15216, United States
c
Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15213, United States
d
Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA 15213, United States
e
Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, United States

a r t i c l e i n f o a b s t r a c t

Article history: Dry eye disease (DED), also known as keratoconjunctivitis sicca, is an ocular surface disease characterized
Received 27 November 2017 by T-cell-mediated inflammation. Current therapeutics, such as immunosuppressive agents, act to sup-
Received in revised form 24 February 2018 press the clinical signs and inflammation. However, long-term usage of these treatments can cause severe
Accepted 1 March 2018
side effects. In this study, we present an alternative therapeutic approach that utilizes a histone deacety-
Available online 9 March 2018
lase inhibitor (HDACi) to regulate transcription of a variety of immunomodulatory genes. Specifically,
HDACi have emerged as a potential anti-inflammatory agent, which can modulate the functions of a sub-
Keywords:
set of suppressive T lymphocytes known as regulatory T cells (Tregs), enhancing FoxP3 acetylation and
HDACi
SAHA
subsequently guarding the transcription factor from proteasomal degradation. Here, a specific HDACi
Microspheres known as SAHA (suberoylanilide hydroxamic acid) was formulated to controllably release in the lacrimal
PLGA gland. Intralacrimal gland injection of PLGA-based SAHA microspheres prevented clinical signs of DED in
Dry eye disease mice with Concanavalin A-induced DED, reduced expression of pro-inflammatory cytokines, and
increased expression of FoxP3 in the lacrimal glands. Murine T cell culture experiments also revealed that
SAHA decreased effector T cell proliferation and enhanced suppressive function of Tregs in co-cultures of
Tregs and effector T cells.

Statement of Significance

In this study, we demonstrate a therapeutic approach that utilizes a histone deactylase inhibitor (HDACi)
to regulate transcription of a variety of immunomodulatory genes. HDACi have emerged as a potential
anti-inflammatory agent, which can modulate the functions of a subset of suppressive T lymphocytes
known as regulatory T cells (Tregs). Here, HDACi microspheres composed of a biocompatible and
biodegradable polymer (poly(lactic-co-glycolic acid) (PLGA)), were able to locally release the HDACi
and prevent clinical signs of DED. This work is timely given the recent shift in treatments of DED towards
immunological based therapies to reduce ocular inflammation. However, notably, many of these treat-
ments require large amounts of drug, and non-specifically suppress the immune system, leading to sev-
eral systemic side effects. Instead of merely suppressing or blocking inflammation, the formulation
described herein intends to balance the microenvironment promoting immunological homeostasis.
This particular drug delivery system may also have broad implications in the field of inflammatory medi-
ated ocular disorders such as uveitis, Sjögren’s syndrome, allergic conjunctivitis.
Ó 2018 Acta Materialia Inc. Published by Elsevier Ltd. All rights reserved.

1. Introduction of the immunological homeostasis of the lacrimal functional unit


(LFU) [6–9]. This disruption stems from persistent infiltration of
Dry eye disease (DED) affects more than 10 million individuals pro-inflammatory effector CD4+ T lymphocytes (Teff) into the
in the United States [1–5]. The disease originates from disruption LFU, and reduced numbers and/or functionally less suppressive
anti-inflammatory regulatory T cells (Tregs) [10,11]. Current ther-
⇑ Corresponding author at: Department of Chemical and Petroleum Engineering, apies are aimed at blocking pro-inflammatory signaling by sup-
University of Pittsburgh, Pittsburgh, PA 15261, United States. pressing T-cell proliferation, or preventing T-cell infiltration
E-mail address: srlittle@pitt.edu (S.R. Little).

https://doi.org/10.1016/j.actbio.2018.03.002
1742-7061/Ó 2018 Acta Materialia Inc. Published by Elsevier Ltd. All rights reserved.
262 M.L. Ratay et al. / Acta Biomaterialia 71 (2018) 261–270

[12,13]. However, in a healthy state, the body utilizes sophisticated 0.16–0.24 dL/g at 0.1% w/v), Concanavalin A (Con A), and Tween80
regulatory mechanisms that include Tregs, which play an integral were purchased from Sigma Aldrich (St. Louis, MO). Poly(vinyl
role in the immune system by suppressing the proliferation of alcohol) (PVA; MW 25 kDa, 98% hydrolyzed) was purchased from
pathogenic effector T cells and promoting the maintenance of the PolySciences (Warrington, PA). Dichloromethane, methanol, and
immunological microenvironment [14–16]. Due to the vital role neutral buffered formalin were obtained from Fisher Scientific
Tregs play in suppressing aberrant inflammation, our group previ- (Hampton, NH). Phosphate-buffered saline (PBS or ‘‘saline”) with-
ously developed a method to bolster the overall pool of Tregs, uti- out calcium and magnesium from Corning (Corning, NY) was used
lizing a sustained release of a combination of Treg-inducing factors for injections and release assays. RPMI-1640 media (Gibco via
(two cytokines and a small molecule drug) [17–19]. This therapeu- Thermo Fisher Scientific, Waltham, MA) and the following supple-
tic approach yielded promising results in an inflammatory murine ments were used for cell culture experiments: fetal bovine serum
model of DED, and ultimately shifted the immunological microen- (FBS; Atlanta Biologicals, Atlanta, GA), non-essential amino acids
vironment in the lacrimal gland tissue. Although, clinical signs of (NEAA; Lonza, Walkersville, MD), HEPES buffer (Lonza), L-
DED were prevented by the combined delivery of these three fac- glutamine (Gibco), 2-mercaptoethanol (Gibco), sodium pyruvate
tors, treatment with a single small molecule drug, instead of a (Sigma), and antibiotic-antimycotic solution (Sigma). Fluorescein,
combination of biologics and small molecule drug, would drasti- CellTrace CFSE, CellTrace Far Red, Dynabeads Mouse T-Activator
cally reduce the regulatory hurdles and translational potential of anti-CD3/CD28, and eBioscience Fixable Viability Dye eFluor 780
such a therapy. were obtained from Thermo Fisher. The following fluorescently
Histone deacetylase inhibitors (HDACi) are a family of small labeled antibodies were obtained from BD Biosciences (San Jose,
molecule drugs with the ability to shift Treg-Teff balance [20]. CA) and used for FACS and flow cytometry: CD4 (RM4-5), CD25
HDACi are best known for their ability induce differentiation and (PC61.5), and CD45RB (16A). Finally, qRT-PCR reagents included
cell cycle arrest in cancer cells [21]. A specific HDACi, known as TRI-reagent (Molecular Research Center, Cincinnati, OH), Quanti-
Vorinostat, or SAHA (N-hydroxy-N0 -phenyl-octanediamide, Tect Reverse Transcription Kit (Qiagen, Valencia, CA), VeriQuest
suberoylanilide hydroxamic acid), has been previously approved Probe qPCR Mastermix (Affymetrix, Santa Clara, CA), and TaqMan
by the FDA for treatment of cutaneous T-cell lymphoma (marketed primer/probes (Thermo Fisher).
by Merck as ZolinzaTM) [21]. In addition to use of SAHA as an anti-
cancer therapeutic, this small molecule drug has recently attracted 2.2. Fabrication of SAHA microspheres
interest as a potential anti-inflammatory therapeutic [22]. HDACi,
such as SAHA, have also recently been shown to enrich or enhance HDACi microspheres were fabricated using a single-emulsion
local populations of Tregs through several mechanisms [20]. HDACi evaporation technique due to the hydrophobic nature of SAHA.
are capable of directly acting on Tregs to increase their suppressive Specifically, 200 mg of PLGA and 40 mg of SAHA were added to a
function by promoting FoxP3 acetylation and binding to DNA mixture of 2.68 ml dichloromethane and 1.32 ml methanol, and
[23,24], or by increasing expression of regulatory molecules like sonicated for 1 h. Subsequently, this emulsion was then mixed
CTLA-4 [22]. SAHA can also increase the number of Tregs through with 60 ml of 2% PVA and homogenized (L4RT-A, Silverson, pro-
multiple routes. For instance, HDACi, including SAHA, can cause cured through Fisher Scientific) at 3000 rpm for 1 min. Subse-
conventional T cells to express FoxP3, thereby converting them quently, the homogenized mixtures were added to 80 ml of 1%
into induced Tregs [25]. Additionally, HDACi can also promote PVA on a stir plate and left for approximately 1.5 h in order for
the generation of tolerogenic dendritic cells (DCs), which can then the organic solvent to evaporate. After 1.5 h, the microspheres
induce Tregs [22,26]. This effect of HDACi is mediated by increased were centrifuged (200g, 5 min, 4 °C), washed 4 times with deion-
acetylation of STAT3, which leads to reduced expression of costim- ized water, and lyophilized for 48 h (Virtis Benchtop K freeze dryer,
ulatory molecules and production of pro-inflammatory cytokines, Gardiner, NY). Microspheres without drug, referred to as ‘‘Blank
as well as increased expression of the immunosuppressive enzyme MS,” were fabricated similarly, except without the addition of
indoleamine 2,3-dioxygenase (IDO) [26,27]. Finally, it has also SAHA.
been suggested that SAHA can inhibit T effector (Teff) proliferation,
independent of Tregs [28]. 2.3. Characterization of SAHA microspheres
Given that HDACi can both expand Tregs and enhance their sup-
pressive function, we hypothesized that the controlled release of The morphology of the microspheres were characterized using
SAHA from degradable, polymer-based microspheres injected into scanning electron microscopy (SEM) (JEOL, JSM-6330F, Peabody,
the lacrimal gland would cause local induction of Tregs and prevent MA) and volume impedance measurements were performed on a
clinical symptoms associated with DED. In order to test this hypoth- Beckman Coulter Counter (Multisizer-3, Beckman Coulter, Fuller-
esis, SAHA was formulated into degradable microspheres made ton, CA). In order to determine the release kinetics of the SAHA
from a polymer with an excellent track record of prior FDA approval microspheres, 10 mg of SAHA MS or Blank MS (unloaded control)
(poly (lactic-co-glycolic) acid). These microspheres are capable of were added to 1 ml of 0.2% Tween 80 in PBS, which was placed
sustainably releasing SAHA for several days. Local release of SAHA onto a rotator at 37 °C. The supernant was sampled daily and the
from these formulations in the lacrimal gland of mice prevent dam- release profile was determined using a NanoDrop 2000 Spec-
age to the ocular tissue, enhance FoxP3 mRNA expression in the trophotometer (ThermoFisher Scientific). The wavelength utilized
lacrimal gland, and reduce the pro-inflammatory microenviron- for this test was 242 nm. A standard curve was completed on the
ment observed in a model of murine DED. NanoDrop to determine the corresponding absorbance values and
the range of concentrations detected in this study, which were
approximately 2 ng/ml to 0.375 mg/ml. The samples were diluted
2. Experimental methods as necessary to be within the detectable range of the standard
curve.
2.1. Materials and reagents
2.4. Mice
Suberoylanilide Hydroxamic Acid (SAHA) was acquired from
Selleck Chem (Houston, TX). Poly(lactic-co-glycolic acid) (PLGA; Female Balb/c mice aged 6–8 weeks from Charles Rivers Labora-
65:35 lactide:glycolide, acid terminated, MW 7–17 kDa, viscosity tories (Wilmington, MA) were utilized for this experimental study.
M.L. Ratay et al. / Acta Biomaterialia 71 (2018) 261–270 263

All murine experiments were approved by the Institutional Animal using a QuantiTect Reverse Transcription Kit. Quantitative real-time
Care and Use Committee, University of Pittsburgh, Pittsburgh, Pa. PCR was then performed using VeriQuest Probe qPCR Mastermix
and TaqMan primer/probes specific for IFN-c (Mm01168134_m1),
2.5. Murine model IL-12 (Mm01288989_m1), IL-6 (Mm00446190_ml), FoxP3
(Mm00475162_m1), and Gusb (Mm01197698_m1). Primer/probes
To induce inflammation-mediated DED, 20 ll of a 10 mg/ml for target genes were FAM-MGB labeled, and that for the endoge-
solution of ConA in PBS was injected into the left and right lacrimal nous control (GUSB) was VIC-MGB_PL labeled. Duplex reactions
glands of mice with the aid of a dissecting microscope (Olympus (target gene + GUSB) were run and analyzed on a StepOnePlus
SZX10, Waltham, MA) [29]. For microsphere treatments, 10 mg/ Real-Time PCR System (Applied Biosystems, Carlsbad, CA). Relative
ml of Blank MS or SAHA MS were administered together with ConA fold changes of IFN-c, IL-12, IL-6, and FoxP3 expression were calcu-
in the same total volume of 20 ll. For non-diseased saline controls, lated and normalized based upon the 2DDCt method [33], using the
20 ll of PBS was injected into the lacrimal gland, instead of ConA Saline group as the untreated control.
and/or microspheres. Prior to lacrimal gland injections, mice were
anesthetized by intraperitoneal injection of ketamine (80–100 mg/ 2.10. Suppression assay
kg) and xylazine (5–10 mg/kg) [29].
Lymphocytes were isolated from Balb/c spleens and stained
2.6. Tear production with antibodies against CD4, CD25, and CD45RB. Flow activated
cell sorting (FACS) was performed (FACSAria, BD Bioscience) to
In order to measure tear production, phenol red cotton threads obtain Teff (CD4+CD25CD45RBhi) and Treg (CD4+CD25+CD45RBlo).
(Oasis Medical, San Dimas, CA) were utilized. Specifically, 7 days Teff and Treg were labeled with CellTrace CFSE or CellTrace Far
after lacrimal gland injections, mice were physically restrained Red, respectively. Teff were cultured in round-bottom 96 well
by hand, and the phenol red thread was placed in the lateral can- plates (5  104 cells/well) with Dynabeads Mouse T-Activator
thus of the eye for a period of 60 s. The amount of tears absorbed CD3/CD28 stimulation (1 Dynabead: 4 Teff) and 0:1, 1:1, 1:2, or
onto the thread (when tears are absorbed onto the thread a color 1:4 ratios of Treg: Teff. T cells were cultured for 72 h in RPMI media
change occurs from yellow to red) was measured using a dissecting supplemented with 10% FBS, 10 mM HEPES, 2 mM L-glutamine, 1
microscope (Olympus SZX10, Waltham, MA) [30,31]. To convert mM sodium pyruvate, 1x antibiotic–antimycotic, 1x NEAA, and
linear thread wetting measurements (mm) to tear volumes (lL), 55 lM 2-mercaptoethanol. Culture media also contained 0, 50, or
a standard curve was generated using known volumes of basic sal- 200 nM SAHA. T cells were stained with Fixable Viability Dye,
ine solution (0.9% w/v NaCl + 16.7 mM NaOH, pH  12), as and CFSE dilution was analyzed by flow cytometry.
described in [32]. The following linear regression equation was
obtained from thread wetting measurements of standard volumes
2.11. Statistical analysis
of basic saline solution using Prism GraphPad: Volume (lL) = 0.0
5106 * [thread wetting length (mm)] + 0.04596; R2 = 0.9741, n = 3
Data expressed as mean ± S.D. Comparisons between multiple
independent measurements per volume.
treatment groups were performed using one-way ANOVA, followed
by Bonferroni correction for multiple comparisons, and p < 0.05 was
2.7. Corneal fluorescein staining
considered statistically significant. For PCR data, a Grubb’s test was
performed to identify any significant outliers. If a significant outlier
Seven days after lacrimal gland injections, and after phenol red
was found (p < 0.05) it was excluded from the statistical analysis. If
thread testing, mice were physically immobilized by hand, and
assumptions of ANOVA were not met, a non-parametric Kruskal-
approximately 1 ml of fluorescein (1% solution in PBS) was applied
Wallis test, followed by Dunn’s post-hoc analysis, was performed.
to the conjunctival sac. Subsequently, the ocular surface was
All animal experiments in this study were repeated twice. The
imaged using a dissecting microscope with camera, and a masked
microsphere fabrication and characterization was repeated three
ophthalmologist used these images to score staining. Corneal fluo-
times for this study. Statistical tests were performed using GraphPad
rescein was scored 0 for no staining, score 1 for a quarter of stain-
Prism Software 6.0 (GraphPad Prism, San Diego, CA).
ing, score of 2 for less than a half, score of 3 for half, and 4 for more
than half of the cornea.
3. Results
2.8. Ocular histology
3.1. Characterization of SAHA microspheres
At the end of the one-week study, mice were euthanized by car-
bon dioxide asphyxiation and secondary cervical dislocation, and Scanning electron micrographs (SEM) (Fig. 1) illustrate that
murine eyes were exenterated and immediately fixed in 10% neu- individual, polymer particles are spherical with average size distri-
tral buffered formalin solution for a period of 48 h. Ocular tissue bution average of 17 mm (Blank MS) and an average of 17 mm
samples were embedded at the same depth and orientation to (SAHA MS), which was confirmed by utilizing a Coulter Counter
ensure comparable regions of tissue were used for histology. Then, (representative plots of volume impedance measurements shown
the paraffin embedded tissue was cut into approximately 5 mm below in Fig. 1). Additionally, the release kinetics were character-
thick sections and stained with Periodic Acid Schiff (PAS). These ized using a NanoDrop (UV–vis spectrophotometer) through detec-
histological sections were scanned and the goblet cell density tion of the absorbance of SAHA, which demonstrates a cumulative
was quantified using a Zeiss Axio Scan. Z1 (Thornwood, NY) and release of approximately 50 ng/mg of microspheres over the course
Pannoramic Viewer software (3D HISTECH Ltd.). of 5–6 days (Fig. 2).

2.9. qRT-PCR 3.2. Aqueous tear production with the administration of SAHA
microspheres
Total RNA was extracted from excised lacrimal glands using TRI-
reagent, and quantified using a NanoDrop 2000 (Thermo Scientific). To determine whether SAHA MS were capable of preventing
For the reverse transcriptase assay, 2 lg RNA was converted to cDNA loss of tear production in our model of DED, a standard phenol
264 M.L. Ratay et al. / Acta Biomaterialia 71 (2018) 261–270

Fig. 1. Characterization of SAHA Microspheres. (A) Representative scanning electron microscopy (SEM) images (1000X). (B) Volume Impedance Measurements (Coulter
Counter) of Blank Microspheres and SAHA Microspheres.

red threads test was utilized at the experimental endpoint [29]. 3.4. Impact of SAHA microspheres on corneal fluorescein staining
Compared to non-diseased mice (injected with saline), mice that
received intralacrimal gland injections of ConA, with or without Representative corneal fluorescein images were captured using
Blank MS, experienced less tear production, as shown in Fig. 3. This a fluorescent dissecting microscope and scored by a masked oph-
loss of tear production caused by ConA was prevented by the thalmologist (Fig. 5A). Compared to the ConA alone and ConA +
administration of SAHA MS (Fig. 3). Blank MS group, the uptake of fluorescein to the cornea was signif-

3.3. Impact of SAHA microsphere administration on goblet cell density

To determine whether SAHA MS could reduce loss of goblet


cells caused by ConA-induced inflammation, Periodic Acid Schiff
(PAS staining) of the ocular tissue was used to identify mucin-
producing goblet cells. SAHA MS treatment of ConA-induced DED
reduced the loss of goblet cells compared to ConA alone (Fig. 4A),
thereby potentially preserving the goblet cells (pink/purple cells)
located in the conjunctiva. Notably, there was a significant preser-
vation of goblet cell density with the administration of SAHA MS as
compared to the ConA alone group shown in Fig. 4B.

Fig. 3. Tear production in mice with ConA-induced DED is restored by adminis-


tration of SAHA MS. Tear production was measured by phenol red thread test and
converted to tear volumes using a standard curve. Data represent mean ± SD for
saline (non-diseased) (n = 5), ConA (diseased) (n = 5), Blank MS (w/ ConA) (n = 5),
and SAHA MS (w/ ConA) (n = 4) groups. A significant difference between mice with
ConA-induced DED that were treated with Blank MS (vehicle control) and SAHA MS
Fig. 2. Release kinetics of SAHA from microspheres (n = 3). was identified by one-way ANOVA, followed by Bonferroni post-hoc test (*p < 0.05).
M.L. Ratay et al. / Acta Biomaterialia 71 (2018) 261–270 265

Fig. 4. SAHA MS Preserve Goblet Cell Density in the Conjunctival Epithelial Layer. (A) Quantification of the Number of Goblet Cells per field shown as mean ± S.D. (n = 5 per
group). (B) Representative Histological Images of the goblet cells in the conjunctiva (10X) of Saline (non-diseased), ConA (diseased), Blank MS (diseased + unloaded
microspheres), SAHA MS (diseased + SAHA microspheres). Significant differences were determined by one-way ANOVA, followed by Bonferroni post-hoc tests, and are
indicated by *p < 0.05, ****p < 0.0001.

Fig. 5. Corneal Fluorescein Staining is Reduced with the Administration of SAHA MS. (A) Corneal Fluorescein Staining scored on a scale of (0–4) shown as mean ± S.D. (n = 6).
(B) Representative Corneal Fluorescein Images of Saline (non-diseased), ConA (diseased), Blank MS (w/ ConA), and SAHA MS (w/ ConA). Significant differences were
determined by one-way ANOVA, followed by Bonferroni post-hoc tests, and are indicated by *p < 0.05, **p < 0.01, ***p < 0.001.
266 M.L. Ratay et al. / Acta Biomaterialia 71 (2018) 261–270

icantly reduced in the Saline and SAHA MS groups shown in Fig. 5B. labeled Teff co-cultured with Tregs were treated with SAHA and
Ultimately, the local administration of SAHA MS to lacrimal gland proliferation was assessed by CFSE dilution. It was observed that
demonstrated that the preventative therapy was able to reduce the Teff alone, treated with 200 nM SAHA, exhibited significantly
corneal fluorescein scores by approximately 4-fold compared to reduced proliferation compared to Teff alone that were not treated
the ConA alone group. with SAHA (Fig. 7A). Teff co-cultured with Tregs exhibited less pro-
liferation, and this was further reduced in a dose-dependent man-
3.5. mRNA expression altered in the lacrimal gland with SAHA ner by the addition of SAHA as shown in Fig. 7B. Although 200 nM
microspheres SAHA inhibited the proliferation of Teff alone by 18% and a 1:1
Teff: Treg ratio (SAHA not treated) inhibited proliferation by 27%,
To determine if SAHA MS could reduce production of inflamma- the combination of 200 nM SAHA and a 1:1 Treg: Teff ratio inhib-
tory cytokines, mRNA expression levels were examined in the ited T cell proliferation by 66%. Overall, the impact of SAHA alone
lacrimal gland tissue after ConA-induced inflammation. Quantita- on solely T effector cells appears to be smaller compared to the
tive RT-PCR data suggested that IL-12, IFN-c, and IL-6 pro- co-culture of Tregs and Teff cells.
inflammatory cytokines were significantly reduced in the lacrimal
gland of the SAHA MS treated as compared to the ConA (diseased) 4. Discussion
group as shown in Fig. 6. In contrast, the level of FoxP3 (transcrip-
tion factor and marker of regulatory T cells) mRNA expression was Recently, immunosuppressive agents such as corticosteroids
significantly higher in the SAHA MS group as compared to the ConA have been investigated as a therapeutic to reduce inflammation
(diseased) group [34]. Though mice treated with Blank MS had associated with DED [37,38]. However, long-term topical use of
intermediate levels of FoxP3 expression (greater than ConA alone, corticosteroids has been implicated in conditions such as glaucoma
but less than SAHA MS treatment), these differences were not sig- and retinopathy [39]. As an alternative to non-specific corticos-
nificant. Together, this data suggests that the SAHA MS was able to teroids and associated side effects, a histone deactylase inhibitor
reduce the pro-inflammatory microenvironment initiated by ConA (HDACi) was utilized to alter the adaptive immune response to
and enhance the expression levels of a Treg-associated transcrip- restore immunological homeostasis. Specifically, the HDACi, SAHA,
tion factor. was selected due to its ability to cause epigenetic modifications
that regulate gene expression and protein function to modulate
3.6. Lymphocyte suppression assay the function of immune cells such as T lymphocytes [21,22]. In par-
ticular, SAHA and other HDACi can stimulate thymic production of
Various mechanisms by which SAHA can influence the Treg-Teff anti-inflammatory Tregs, enhance Treg suppressive function, and
balance have been proposed including direct suppression of Teff promote the peripheral conversion of CD4+ naïve T cells into Tregs
proliferation and enhancement of Treg function [20,22,28,35,36]. [22,36]. Although, HDACi have shown promising immunomodula-
In order to gain insight into the mechanism of inflammation reduc- tory effects on T lymphocytes in vitro and in pre-clinical inflamma-
tion in our model of murine DED, CFSE labeled Teff alone or CFSE tory models at a dose of 0.1 mg/kg/d [20], this class of small

Fig. 6. mRNA Expression is altered in the lacrimal gland tissue with the administration of SAHA MS, shown as mean ± S.D. (n  5 mice per group). Relative fold changes of
IFN-c, IL-12, IL-6, and FoxP3 mRNA expression were calculated and normalized based upon the 2DDCt method, with the Saline group as the untreated control. Significant
differences were determined by one-way ANOVA, followed by Bonferroni post-hoc test, or Kruskal-Wallis test, followed by Dunn’s post-hoc test (for IL-6), and are indicated
by *p < 0.05; **p < 0.01.
M.L. Ratay et al. / Acta Biomaterialia 71 (2018) 261–270 267

Fig. 7. Lymphocyte Suppression Assay. (A) Representative Flow Cytometry Histograms of T effector cells only untreated (no SAHA) and cells treated with 50 nM and 200 nM
SAHA and bar graphs shown as mean ± S.D. (B) Representative Flow Cytometry Histograms of 1:1 Treg: T effector cells untreated (no SAHA) and cells treated with 50 nM and
200 nM SAHA and bar graphs shown as mean ± S.D. Significant differences were determined by one-way ANOVA, followed by Bonferroni post-hoc test, and are indicated by
*
p < 0.05; **p < 0.01; ****p < 0.0001.

molecules possesses a narrow therapeutic window (both efficacy ocular tissue in DED is reportedly due to an increase in the pro-
and toxicity) [40], which limit their broader applicability. The abil- inflammatory milieu [44]. Thus, in this study, histological sections
ity to locally and sustainably deliver the HDACi would in theory of the conjunctiva were examined to determine whether SAHA MS
permit a much lower drug dosage, reducing toxicity concerns, as treatment preserved mucin-producing goblet cells. Indeed, there
well as permit reduced frequency of injections. Therefore, in this was a significantly greater goblet cell density in SAHA MS-
study a controlled release system was fabricated to provide local treated mice, compared to diseased (ConA) mice (Fig. 4). Although,
administration of SAHA to the lacrimal gland in an inflammatory the preservation of goblet cell density was not as profound as other
murine model of DED. measurements (ex: corneal fluorescein staining) there was a statis-
SAHA releasing microspheres (MS) were fabricated using the tically significant preservation of goblet cell numbers in the SAHA
biodegradable and biocompatible polymer poly (lactic-co- MS as compared to the ConA (diseased) group as can be seen in
glycolic) acid (PLGA) (Fig. 1), and showed the ability to release Fig. 4. These results are in agreement with a previous report by
SAHA in vitro over the course of approximately five days (as shown DeZoeten et al., which demonstrated that administration of an
in Fig. 2) with the intention of altering the local microenvironment HDACi from the same class as SAHA, led to preservation of intesti-
in the ocular tissue [17,41]. Notably, the data suggest that local nal goblet cells in an inflammatory colitis model [47].
administration of SAHA MS in the ocular tissue prevented several Since loss of aqueous tear production and/or mucin-producing
clinical signs of DED. For instance, a common clinical sign of DED goblet cells in DED ultimately leads to damage and increased per-
is a reduction of overall tear production, which subsequently can meability of the ocular surface [48,49–51], we investigated
lead to ocular dryness and irritation [42]. While aqueous tear whether SAHA MS, which prevented both of the former patholog-
secretion was significantly reduced in diseased (ConA ± Blank ical features, also maintained integrity of the corneal epithelium.
MS), as expected [29], SAHA MS treatment prevented ConA- Corneal integrity was assessed with fluorescein [12,48], a dye that
induced loss of aqueous tear secretion (Fig. 3). In addition to pre- stains dead epithelial cells and can diffuse into areas where cellular
serving aqueous tear production, effective therapeutics should also tight junctions are compromised [52]. Corneal fluorescein staining
maintain the composition of tears by protecting gel-forming, is a standard diagnostic indicator of ocular surface damage and
mucin-producing goblet cells [2]. These goblet cells are located measurement for DED severity [48]. Fluorescein staining was
on the apical surface of the conjunctiva, within the stratified observed in mice with DED induced by ConA (with or without
columnar conjunctival epithelial cells, and produce important Blank MS), as shown in Fig. 5. Notably, there was a 4-fold reduction
non-aqueous components of tears, such as mucin [43,44,45]. in the average fluorescein staining score in the SAHA MS group,
Mucin 5AC (MUC5AC), a glycoprotein produced by goblet cells, acts compared to ConA alone, suggesting that the HDACi was able to
as a gel layer to trap pollen and allergens [44,46]. Depletion of gob- prevent damage to the ocular tissue initiated by ConA.
let cells due to chronic inflammation in conditions, such as DED, In order to demonstrate that the prevention of clinical signs of
can ultimately lead to conjunctival epithelial squamous metaplasia DED was attributed to changes in the underlying T cell mediated
and abnormal tear film [46]. Interestingly, loss of goblet cells in immune response, the lacrimal gland microenvironment was eval-
268 M.L. Ratay et al. / Acta Biomaterialia 71 (2018) 261–270

uated using PCR analysis. The underlying pathogenesis of DED may fail to capture some of the multifactorial pathophysiology of
involves effector T cells that infiltrate the ocular tissue and secrete chronic and/or autoimmune forms of DED. Therefore, future stud-
pro-inflammatory cytokines, which can directly affect the health of ies in chronic DED models (e.g. induction by chronic desiccating
the ocular microenvironment [9,53–55]. Since increases in pro- stress) will be needed to determine whether restoring tear and
inflammatory cytokines have been found in inflamed lacrimal mucin production (e.g. by resolving lacrimal gland inflammation
gland tissues of mice, the microenvironment of the tissue was with SAHA MS) could also help to reduce ocular inflammation
investigated to determine whether the SAHA MS-mediated preven- mediated by immune cells in the conjunctiva [9,66,67]. In such
tion of clinical signs of DED was due to a reduced pro- chronic models, use of SAHA MS with longer release kinetics, or
inflammatory milieu in the ocular tissue [56–58]. Specifically, novel delivery systems that could provide sustained topical deliv-
mRNA expression levels of pro-inflammatory cytokines (IL-12, ery of SAHA to the conjunctiva (as in [68]), rather than lacrimal
IFN-c, and IL-6) in lacrimal gland tissue were evaluated by qRT- gland, could be investigated. Chronic DED models would also allow
PCR. Expression of IL-12 and IFN-c was induced by ConA injection, us to evaluate treatment of ongoing DED and to study the kinetics
relative to saline controls, as shown in Fig. 6. This is likely attribu- of disease progression and resolution following treatment with
ted to ConA acting as a T-cell mitogen, ultimately leading to the SAHA MS (or soluble SAHA). Additional clinical tests for DED (e.g.
production of Th1 cytokines [59,60]. Both of these findings in the tear osmolarity and evaporation rate) could also provide useful
ConA-induced murine model of DED are consistent with clinical information regarding the status of the ocular surface, and could
reports that IL-12 and IFN-c are upregulated in tears of patients be performed throughout the course of disease progression. In
with DED [42,43,56]. In addition to increases in IL-12 and IFN-c the future, large animal pre-clinical rabbit models would also be
in the lacrimal gland, IL-6 was also significantly elevated by ConA, needed to move toward potential clinical translation and to better
which is consistent with other reports of increased IL-6 production test local toxicity of SAHA MS and/or topical SAHA sustained deliv-
in lacrimal glands of DED murine models [61]. Importantly, treat- ery formulations in various compartments of the eye. Additionally,
ment with SAHA MS significantly inhibited ConA-induced expres- by ameliorating inflammation in the lacrimal glands, SAHA MS
sion of IL-12, IFN-c, and IL-6 (Fig. 6). These reductions in the could also potentially be used to treat Sjögren’s syndrome, an
expression levels of pro-inflammatory cytokines prompted the autoimmune disease targeting the lacrimal and salivary glands
question of whether SAHA MS were altering the immunological that causes a severe form of DED and dry mouth. Accordingly,
microenvironment by increasing anti-inflammatory Tregs. Previ- future studies using SAHA MS in murine models of Sjögren’s syn-
ous studies have demonstrated that HDACi can expand FoxP3+ drome would be of interest [69,70]. Such studies would also pro-
Tregs in vitro and in vivo [20,28]. Interestingly, we observed a sig- vide additional insight into the various types of DED (e.g.
nificant increase in FoxP3 mRNA expression in the lacrimal gland evaporative, aqueous deficient, Sjögren’s, non-Sjögren’s) for which
tissue of SAHA MS-treated mice (Fig. 6), suggesting that the reduc- this therapeutic approach may be more or less effective. Ulti-
tion of signs of DED and pro-inflammatory microenvironment in mately, the controlled delivery of SAHA may also have pharmaco-
the lacrimal gland may be due to Tregs suppressing ocular inflam- logical benefits for other inflammation-mediated conditions (based
mation. Future studies will be needed to determine whether on previous literature reports) [23,25,28,71] by locally inducing
increased FoxP3 expression in lacrimal glands of SAHA MS- FoxP3+ Tregs and suppressing the pro-inflammatory milieu.
treated mice is due to an increase in the number of Tregs in the tis-
sue, or enhanced expression of FoxP3 by Tregs, which is associated
with greater suppressive function [62]. Acknowledgements
In vitro experiments suggest, however, that SAHA may enhance
Treg function in addition to directly inhibiting Teff (Fig. 7). Notably, This work was supported in part by the National Institutes of
neither Treg induction, nor increased expression of FoxP3 by Tregs Health (NIH) CORE Grant P30 EY008098, the University of Pitts-
was observed with SAHA treatment in vitro in our hands (data not burgh, Pittsburgh, PA; an unrestricted grant from Research to Pre-
shown). It is possible that this discrepancy between in vitro and vent Blindness, NY. Research reported in this publication was
in vivo results regarding FoxP3 expression is due to the fact that supported by the National Center for Advancing Translational
increased FoxP3 mRNA in the lacrimal gland may not necessarily Sciences of the National Institutes of Health under Award Number
correspond to increases in FoxP3 protein, as was measured TL1R001858. The content is solely the responsibility of the authors
in vitro experiments. Alternatively, enhanced FoxP3 expression and does not necessarily represent the official view of the National
in vivo could be due to the effects of dendritic cells (DCs) and other Institutes of Health. We would also like to acknowledge Dr. Julia K.
antigen-presenting cells on T cells in vivo [22]. In addition to direct Polat for scoring the corneal fluorescein images and Katherine A.
effects on T cells, SAHA has also been shown to cause DCs to reduce Davoil for assisting with ocular histology. Finally, thanks to the
costimulatory molecule expression and increase IDO production, Department of Dermatology at the University of Pittsburgh for
both of which can promote Treg induction [26]. Due to the ubiqui- use of their facilities and equipment for RNA isolation and qRT-
tous nature of histone acetylation and the dynamic nature of epi- PCR.
genetic regulation, it is not surprising that HDACi shift the Teff-
Treg balance through multiple direct and indirect mechanisms.
The results presented here are consistent with several previous References
reports of SAHA directly inhibiting Teff proliferation and enhancing
the suppressive function of Tregs [22,25,28,36]. [1] R.D. William Stevenson, Sunil K. Chauhan, Dry Eye Disease, Arch. Ophthamol.
130 (2012) 90–100..
In summary, administration of SAHA MS preserved aqueous [2] M. Rolando, M. Zierhut, The ocular surface and tear film and their dysfunction
tear production (Fig. 3), reduced inflammation-induced loss of in dry eye disease, Surv. Ophthalmol. 45 (Suppl 2) (2001) S203–S210.
mucin-producing goblet cells (Fig. 4), and prevented damage to [3] F. Brewitt, H. Sistani, Dry eye disease: the scale of the problem, Surv.
Ophthalmol. 45 (2001) 199–202.
ocular tissue (Fig. 5), likely by reducing the pro-inflammatory
[4] J.L. Gayton, Etiology, prevalence, and treatment of dry eye disease, Clin.
milieu in the lacrimal gland (Fig. 6) and potentially by enhancing Ophthalmol. 3 (2009) 405–412.
numbers and/or suppressive function of FoxP3+ Tregs (Figs. 6–7). [5] J. Yu, C.V. Asche, C.J. Fairchild, The economic burden of dry eye disease in the
While the ConA-induced DED model in these studies is frequently United States: a decision tree analysis, Cornea. 30 (2011) 379–387.
[6] M.E. Stern, J. Gao, K.F. Siemasko, R.W. Beuerman, S.C. Pflugfelder, The role of
used as an acute model of inflammation-mediated DED for pre- the lacrimal functional unit in the pathophysiology of dry eye, Exp. Eye Res. 78
clinical assessment of preventative therapies [18,29,30,63–65], it (2004) 409–416.
M.L. Ratay et al. / Acta Biomaterialia 71 (2018) 261–270 269

[7] S.K. Chauhan, J. El Annan, T. Ecoiffier, S. Goyal, Q. Zhang, D.R. Saban, R. Dana, [37] C.S. De Paiva, R.M. Corrales, A.L. Villarreal, W.J. Farley, D.Q. Li, M.E. Stern, S.C.
Autoimmunity in dry eye is due to resistance of Th17 to Treg suppression, J. Pflugfelder, Corticosteroid and doxycycline suppress MMP-9 and
Immunol. 182 (2009) 1247–1252. inflammatory cytokine expression, MAPK activation in the corneal
[8] J.Y. Niederkorn, M.E. Stern, S.C. Pflugfelder, C.S. De Paiva, R.M. Corrales, J. Gao, epithelium in experimental dry eye, Exp. Eye Res. 83 (2006) 526–535,
K. Siemasko, Desiccating stress induces T cell-mediated Sjogren ’ s Syndrome- https://doi.org/10.1016/j.exer.2006.02.004.
like lacrimal keratoconjunctivitis, J. Immunol. 176 (2006) 3950–3957. [38] H.D. Perry, Evaluation of topical cyclosporine for the treatment of dry eye
[9] Y. Chen, S.K. Chauhan, H.S. Lee, D.R. Saban, R. Dana, Chronic dry eye disease is disease, Arch. Ophthalmol. 126 (2008) 1046.
principally mediated by effector memory Th17 cells, Mucosal Immunol. 7 [39] J.P. Kersey, D.C. Broadway, Corticosteroid-induced glaucoma: a review of the
(2014) 38–45. literature, Eye 20 (2006) 407–416.
[10] M. Miyara, S. Sakaguchi, Natural regulatory T cells: mechanisms of [40] J.R. Whittle, J. Desai, Histone deacetylase inhibitors in cancer: What have we
suppression, Trends Mol. Med. 13 (2007) 108–116. learned?, Cancer 121 (2015) 1164–1167
[11] J. Stein-Streilein, A.W. Taylor, An eye’s view of T regulatory cells, J. Leukoc. Biol. [41] X. Rong, W. Yuan, Y. Lu, X. Mo, Safety evaluation of poly(lactic-co-glycolic
81 (2007) 593–598. acid)/poly(lactic-acid) microspheres through intravitreal injection in rabbits,
[12] S. Goyal, S.K. Chauhan, Q. Zhang, R. Dana, Amelioration of murine dry eye Int. J. Nanomed. 9 (2014) 3057–3068.
disease by topical antagonist to chemokine receptor 2, Arch. Ophthalmol. 127 [42] J.M. Tiffany, Tears in health and disease, Eye (Lond). 17 (2003) 923–926.
(2009) 882–887. [43] T.G. Coursey, J.T. Henriksson, F.L. Barbosa, C.S. de Paiva, S.C. Pflugfelder,
[13] C.A. Dinarello, J.W.M. van der Meer, Treating inflammation by blocking Interferon-c–induced unfolded protein response in conjunctival goblet cells as
interleukin-1 in humans, Semin. Immunol. 25 (2013) 469–484. a cause of mucin deficiency in Sjögren Syndrome, Am. J. Pathol. 186 (2016) 1–
[14] M. Yadav, S. Stephan, J.A. Bluestone, Peripherally induced Tregs-role in 12.
immune homeostasis and autoimmunity, Front. Immunol. 4 (2013) 1–12. [44] Z. Zhang, W.Z. Yang, Z.Z. Zhu, Q.Q. Hu, Y.F. Chen, H. He, Y.X. Chen, Z.G. Liu,
[15] A. Schmidt, N. Oberle, P.H. Krammer, Molecular mechanisms of Treg-mediated Therapeutic effects of topical doxycycline in a benzalkonium chloride-induced
cell suppression, Front. Immunol. 3 (2012) 1–20. mouse dry eye model, Investig. Ophthalmol. Vis. Sci. 55 (2014) 2963–2974.
[16] B.D. Singer, L.S. King, F.R. D’Alessio, Regulatory T cells as immunotherapy, [45] C. Baudouin, P. Aragona, E.M. Messmer, A. Tomlinson, M. Calonge, K.G.
Front. Immunol. 5 (2014) 1–10. Boboridis, Y.A. Akova, G. Geerling, M. Labetoulle, M. Rolando, Role of
[17] S. Jhunjhunwala, S.C. Balmert, G. Raimondi, E. Dons, E.E. Nichols, A.W. Thomson, hyperosmolarity in the pathogenesis and management of dry eye disease:
S.R. Little, Controlled release formulations of IL-2, TGF-b1 and rapamycin for the Proceedings of the ocean group meeting, Ocul. Surf. 11 (2013) 246–258.
induction of regulatory T cells, J. Control. Release. 159 (2012) 78–84. [46] Vamsi K. Gangaraju, Lin Haifan, Conjunctival epithelial and goblet cell function
[18] M.L. Ratay, S.C. Balmert, A.P. Acharya, A.C. Greene, T. Meyyappan, S.R. Little, in chronic inflammation and ocular allergic inflammation, Curr. Opin. Clin.
TRI Microspheres prevent key signs of dry eye disease in a murine, Immunol. 14 (2014) 467–470.
inflammatory model, Sci. Rep. 7 (2017) 17527. [47] Edwin F. De Zoeten, Liqing Wang, Hong Sai, Wolfgang H. Dillmann, W.W.
[19] S.C. Balmert, C. Donahue, J.R. Vu, G. Erdos, L.D. Falo, S.R. Little, In vivo induction Hancock, Inhibition of HDAC9 increases T regulatory cell function and
of regulatory T cells promotes allergen tolerance and suppresses allergic prevents colitis in mice, Gastroenterology 138 (2010) 583–594.
contact dermatitis, J. Control. Release. 261 (2017) 223–233. [48] Z. Lin, X. Liu, T. Zhou, Y. Wang, L. Bai, H. He, Z. Liu, A mouse dry eye model
[20] R. Tao, E.F. De Zoeten, E. Ozkaynak, C. Chen, L. Wang, P.M. Porrett, B. Li, L.A. induced by topical administration of benzalkonium chloride, Mol. Vis. 17
Turka, E.N. Olson, M.I. Greene, A.D. Wells, W.W. Hancock, Deacetylase (2011) 257–264.
inhibition promotes the generation and function of regulatory T cells, Nat. [49] E. Badaro, E.A. Novais, F.M. Penha, M. Maia, M.E. Farah, E.B. Rodrigues, S. Paulo,
Med. 13 (2007) 1299–1307. Vital dyes in ophthalmology: a chemical perspective, Curr. Eye Res. (2014) 1–
[21] K. Ververis, A. Hiong, T.C. Karagiannis, P.V. Licciardi, Histone deacetylase 10.
inhibitors (HDACIS): Multitargeted anticancer agents, Biol. Targets Ther. 7 [50] D. Zoukhri, A. Fix, J. Alroy, C.L. Kublin, Mechanisms of murine lacrimal gland
(2013) 47–60. repair after experimentally induced inflammation, Investig. Ophthalmol. Vis.
[22] T. Akimova, U.H. Beier, Y. Liu, L. Wang, W.W. Hancock, Histone/protein Sci. 49 (2008) 4399–4406.
deacetylases and T-cell immune responses, Blood 119 (2012) 2443–2451. [51] J.D. Nelson, H. Helms, R. Fiscella, Y. Southwell, J.D. Hirsch, A new look at dry
[23] C. Chen, L. Wang, P.M. Porrett, B. Li, R. Tao, E.F. De Zoeten, O. Engin, L.A. Turka, eye disease and its treatment, Adv. Ther. 17 (2000) 84–93.
E.N. Olson, M.I. Greene, A.D. Wells, W.W. Hancock, Deacetylase inhibition [52] A.J. Bron, P. Argueso, M. Irkec, F.V. Bright, Clinical staining of the ocular surface:
promotes the generation and function of regulatory T cells, Nat. Med. 13 Mechanisms and interpretations, Prog. Retin. Eye Res. 44 (2015) 36–61.
(2007) 1299–1307. [53] J. El Annan, S.K. Chauhan, T. Ecoiffier, Q. Zhang, D.R. Saban, R. Dana,
[24] B. Li, A. Samanta, X. Song, K.T. Iacono, K. Bembas, R. Tao, S. Basu, J.L. Riley, W.W. Characterization of effector T cells in dry eye disease, Invest. Ophthalmol.
Hancock, Y. Shen, S.J. Saouaf, M.I. Greene, H.T. N-terminal, FOXP3 interactions Vis. Sci. 50 (2009) 3802–3807.
with histone acetyltransferase and class II histone deacetylases are required [54] T.G. Coursey, N.B. Gandhi, E.A. Volpe, S.C. Pflugfelder, C.S. De Paiva, Chemokine
for repression, Proc. Natl. Acad. Sci. USA 104 (2007) 4571–4576. receptors CCR6 and CXCR3 are necessary for CD4+ T cell mediated ocular
[25] J.L. Lucas, P. Mirshahpanah, E. Haas-Stapleton, K. Asadullah, T.M. Zollner, R.P. surface disease in experimental dry eye disease, PLoS One 8 (2013).
Numerof, Induction of Foxp3+ regulatory T cells with histone deacetylase [55] M.E. Stern, C.S. Schaumburg, S.C. Pflugfelder, Dry eye as a mucosal
inhibitors, Cell. Immunol. 257 (2009) 97–104. autoimmune disease, Int. Rev. Immunol. 32 (2013) 19–41.
[26] J. Frikeche, Z. Peric, E. Brissot, M. Grégoire, B. Gaugler, M. Mohty, Impact of [56] M.L. Massingale, X. Li, M. Vallabhajosyula, D. Chen, Y. Wei, P.A. Asbell, Analysis
HDAC inhibitors on dendritic cell functions, Exp. Hematol. 40 (2012) 783–791. of inflammatory cytokines in the tears of dry eye patients, Cornea 28 (2009)
[27] Y. Sun, Y.E. Chin, E. Weisiger, C. Malter, I. Tawara, T. Toubai, E. Gatza, P. 1023–1027.
Mascagni, C.A. Dinarello, P. Reddy, Cutting edge: negative regulation of [57] K.S. Na, J.W. Mok, J.Y. Kim, C.R. Rho, C.K. Joo, Correlations between tear
dendritic cells through acetylation of the nonhistone protein STAT-3 1, J. cytokines, chemokines, and soluble receptors and clinical severity of dry eye
Immunol. 182 (2009) 5899–5903. disease, Investig. Ophthalmol. Vis. Sci. 53 (2012) 5443–5450.
[28] J. Johnson, A. Pahuja, M. Graham, B. Hering, W.W. Hancock, P. Bansal-Pakala, [58] S.C. Pflugfelder, R.M. Corrales, C.S. de Paiva, T helper cytokines in dry eye
Effects of histone deacetylase inhibitor SAHA on effector and FOXP3 disease, Exp. Eye Res. 117 (2013) 118–125.
+Regulatory T cells in Rhesus macaques, Transplant. Proc. 40 (2008) 459–461. [59] R. Palacios, Concanavalin A triggers T lymphocytes by directly interacting with
[29] M.L. Ratay, A.J. Glowacki, S.C. Balmert, A.P. Acharya, J. Polat, L.P. Andrews, M.V. their receptors for activation, J. Immunol. 128 (1982) 337–342.
Fedorchak, J.S. Schuman, D.A.A. Vignali, S.R. Little, Treg-recruiting [60] F. Gantner, M. Leist, A.W. Lohse, P.G. Germann, G. Tiegs, Concanavalin A-
microspheres prevent inflammation in a murine model of dry eye disease, J. induced T-cell-mediated hepatic injury in mice: the role of tumor necrosis
Control. Release. 258 (2017) 208–217. factor, Hepatology 21 (1995) 190–198.
[30] M.J. Lee, D.H. Kim, J.S. Ryu, A.Y. Ko, J.H. Ko, M.K. Kim, W.R. Wee, S.I. Khwarg, J.Y. [61] X. Xiao, X. Shi, Y. Fan, X. Zhang, M. Wu, P. Lan, L. Minze, Y.-X. Fu, R.M. Ghobrial,
Oh, Topical TSG-6 administration protects the ocular surface in two mouse W. Liu, X.C. Li, GITR subverts Foxp3(+) Tregs to boost Th9 immunity through
models of inflammation-related dry eye, Investig. Ophthalmol. Vis. Sci. 56 regulation of histone acetylation, Nat. Commun. 6 (2015) 8266.
(2015) 5175–5181, https://doi.org/10.1167/iovs.14-16307. [62] S.K. Chauhan, D.R. Saban, H.K. Lee, R. Dana, Levels of Foxp3 in regulatory T cells
[31] D. Dursun, M. Wang, D. Monroy, D.-Q. Li, B.L. Lokeshwar, M.E. Stern, S.C. reflect their functional status in transplantation, J. Immunol. 182 (2009) 148–
Pflugfelder, A mouse model of keratoconjunctivitis sicca, Invest. Ophthalmol. 153.
Vis. Sci. 43 (2002) 632–638. [63] M.J. Seo, J.M. Kim, M.J. Lee, Y.S. Sohn, K.K. Kang, M. Yoo, The therapeutic effect
[32] P. Stewart, Z. Chen, W. Farley, L. Olmos, S.C. Pflugfelder, Effect of experimental of DA-6034 on ocular inflammation via suppression of MMP-9 and
dry eye on tear sodium concentration in the mouse, Eye Contact Lens. 31 inflammatory cytokines and activation of the MAPK signaling pathway in an
(2005) 175–178. experimental dry eye model, Curr. Eye Res. 35 (2010) 165–175.
[33] K.J. Livak, T.D. Schmittgen, Analysis of relative gene expression data using real- [64] M.J. Lee, A.Y. Ko, J.H. Ko, H.J. Lee, M.K. Kim, W.R. Wee, S.I. Khwarg, J.Y. Oh,
time quantitative PCR and the 2-DDCT method, Methods 25 (2001) 402–408. Mesenchymal stem/stromal cells protect the ocular surface by suppressing
[34] Q. Tang, J.A. Bluestone, The Foxp3+ regulatory T cell: a jack of all trades, master inflammation in an experimental dry eye, Mol. Ther. (2014) 1–8.
of regulation, Nat. Immunol. 9 (2008) 239–244. [65] M.T.T.J. Nagelhout, D.A. Gamache, L. Roberts, J.M. Yanni Brady, Preservation of
[35] F. Blanchard, C. Chipoy, Histone deacetylase inhibitors: new drugs for the Tear Film Integrity and Inhibition of Corneal Injury by Dexamethasone in a
treatment of inflammatory diseases?, Drug Discov Today. 10 (2005) 197–204. Rabbity Model of Lacrimal Gland Inflammation -Induced Dry Eye, J. Ocul.
Pii s1359-. Pharmacol. Ther. 21 (2005).
[36] T. Akimova, G. Ge, T. Golovina, T. Mikheeva, L. Wang, J.L. Riley, W.W. Hancock, [66] C.S.D.P. Zhang, E.Al. Xiaobo, Topical interferon-gamma neutralization prevents
Histone/protein deacetylase inhibitors increase suppressive functions of conjunctival goblet cell loss in experimental murine dry eye, Exp. Eye Res. 118
human FOXP3+ Tregs, Clin. Immunol. 136 (2010) 348–363. (2014) 117–124.
270 M.L. Ratay et al. / Acta Biomaterialia 71 (2018) 261–270

[67] V.L. Perez, S.C. Pflugfelder, S. Zhang, A. Shojaei, R. Haque, Lifitegrast, a novel [70] T.N. Lavoie, B.H. Lee, C.Q. Nguyen, Current concepts: mouse models of
integrin antagonist for treatment of dry eye disease, Ocul. Surf. 14 (2016) 207– Sjögren’s syndrome, J. Biomed. Biotechnol. 2011 (2011) 1–14.
215. [71] L. Wang, E.F. de Zoeten, M.I. Greene, W.W. Hancock, Immunomodulatory
[68] M.V. Fedorchak, I.P. Conner, J.S. Schuman, A. Cugini, S.R. Little, Long term effects of deacetylase inhibitors: therapeutic targeting of FOXP3+ regulatory T
glaucoma drug delivery using a topically retained gel/microsphere eye drop, cells, Nat. Rev. Drug Discov. 8 (2009) 969–981.
Sci. Rep. 7 (2017) 8639.
[69] Y. Park, A.E. Gauna, S. Cha, Y. Park, Mouse models of primary Sjogren ’ s
Syndrome, Curr. Pharm. 21 (2015) 2350–2364.

Anda mungkin juga menyukai