Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

A C-terminal peptide from type I interferon protects the retina in a mouse model of autoimmune uveitis

  • Chulbul M. Ahmed,

    Roles Conceptualization, Formal analysis, Investigation, Writing – original draft

    Affiliation Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL, United States of America

  • Cristhian J. Ildefonso,

    Roles Formal analysis, Visualization, Writing – review & editing

    Affiliation Department of Ophthalmology, University of Florida, Gainesville, FL, United States of America

  • Howard M. Johnson,

    Roles Supervision, Writing – review & editing

    Affiliation Department of Microbiology and Cell Science, University of Florida, Gainesville, FL, United States of America

  • Alfred S. Lewin

    Roles Formal analysis, Supervision, Writing – review & editing

    lewin@ufl.edu

    Affiliation Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL, United States of America

Abstract

Experimental autoimmune uveitis (EAU) in rodents recapitulates many features of the disease in humans and has served as a useful tool for the development of therapeutics. A peptide from C-terminus of interferon α1, conjugated to palmitoyl-lysine for cell penetration, denoted as IFNα-C, was tested for its anti-inflammatory properties in ARPE-19 cells, followed by testing in a mouse model of EAU. Treatment with IFNα-C and evaluation by RT-qPCR showed the induction of anti-inflammatory cytokines and chemokine. Inflammatory markers induced by treatment with TNFα were suppressed when IFNα-C was simultaneously present. TNF-α mediated induction of NF-κB and signaling by IL-17A were attenuated by IFNα-C. Differentiated ARPE-19 cells were treated with TNFα in the presence or absence IFNα-C and analyzed by immmunhistochemistry. IFNα-C protected against the disruption integrity of tight junction proteins. Similarly, loss of transepithelial resistance caused by TNFα was prevented by IFNα-C. B10.RIII mice were immunized with a peptide from interphotoreceptor binding protein (IRBP) and treated by gavage with IFNα-C. Development of uveitis was monitored by histology, fundoscopy, SD-OCT, and ERG. Treatment with IFNα-C prevented uveitis in mice immunized with the IRBP peptide. Splenocytes isolated from mice with ongoing EAU exhibited antigen-specific T cell proliferation that was inhibited in the presence of IFNα-C. IFNα-C peptide exhibits anti-inflammatory properties and protects mice against damage to retinal structure and function suggesting that it has therapeutic potential for the treatment of autoimmune uveitis.

Introduction

Uveitis refers to an inflammation affecting the uveal and retinal layers of the eye that accounts for 10–15% of blindness in US[1]. In rodent models of the disease, histological analysis shows injury to retinal layers, retinal folds, subretinal fluid, damage to retinal blood vessels and the retinal pigment epithelium (RPE), and choroiditis. Uveitis may affect both the anterior and posterior chambers of the eye [2,3]. Left untreated, it can lead to permanent retinal damage, cystoid macular edema and vitreous opacity[4]. Uveitis can arise from an infection, systemic inflammation, or from a local autoimmune response. It may also be a component of a systemic autoimmune response involving multiple organs. Several conditions can cause uveitis secondarily, including sarcoidosis [5], Behcet’s disease [6], and Vogt-Koyanagi-Harada (VKH) syndrome [7]. Autoimmune disorders such as Crohn’s disease [8], rheumatoid arthritis [9], and multiple sclerosis (MS) [10,11], can also have an overlapping uveitis. After the initial trigger, the immunogenic pathways in these different diseases share many common features. Both the innate and adaptive immune responses are activated in autoimmune uveitis.[12,13]. An animal model of the disease, experimental autoimmune uveitis (EAU) is established by inoculating mice with specific antigens including peptides derived from interphotoreceptor retinoid binding protein (IRBP), RPE65, rhodopsin, retinal arrestin, recoverin, and phosducin. Transgenic lines of mice have been established that express a T cell receptor that is specific for immunogenic peptides derived from IRBP, and one of these lines develops uveitis spontaneously, providing a model that avoids the need for immunization [14]. Uveitis may also be induced by adoptive transfer of uveitic antigen-specific T cells [2]. Such models have been important in deciphering the mechanisms of disease and in testing therapeutic approaches.

Aside from external factors, the host microbiome has been identified in recent years as a contributing factor to both infectious and autoimmune uveitis. Commensal microbes from intestine, saliva, or ocular source have been implicated in the initiation of uveitis [1517]. It is believed that through a process involving antigen mimicry followed by an adjuvant effect, these microbes can produce autoreactive lymphocytes leading to the development of uveitis. Thus, what is gleaned from rodent models of the disease can have greater relevance in alleviating the disease arising from host-microbiome interaction.

Non-infectious uveitis is currently being treated by using corticosteroids, general immunosuppressants, or specific antibodies [18,19]. While these treatments provide temporary relief, they are associated with side effects that include cataract formation, increased susceptibility to infection, and elevated intraocular pressure [20]. There is thus a critical need for treatments that are sustained and safer.

The ability of type I interferons (IFNα, IFNβ, IFNτ) to suppress autoimmune and inflammatory responses is well documented [2123]. This has been exploited to develop treatments for multiple sclerosis [24], psoriasis [25], and coeliac diseases [26]. Several studies have reported the success of use of IFNα in a number of ocular disorders, including uveitis [2729]. Type I interferon (IFNα or IFNβ) are commonly used to treat relapsing multiple sclerosis (MS) [24]. Nevertheless, its use is associated with lymphopenia, flu-like symptoms, depression, and weight loss. To circumvent these undesirable effects, we have developed a C-terminal peptide of IFNα(residues 152–189), which upon conjugation with palmitoyl-lysine can penetrate the plasma membrane. The resulting modified peptide is denoted as IFNα-C. IFNα-C can recapitulate nearly all the activities of intact IFN [30], including protection against the severe remitting/relapsing paralysis in a mouse model of multiple sclerosis (experimental allergic encephalomyelitis or EAE). Between the regions 152–189, there is 70% homology and 90% conservation between human and mouse IFNα1 sequences. Of note, the IFNα-C peptide lacks the toxicity associated with the parent IFN [30], and may thus represent an improved alternative to parent IFN. Since MS and autoimmune uveitis share common characteristics, we were interested in testing the therapeutic efficacy of IFNα-C peptide in experimental autoimmune uveitis (EAU). Herein, we provide evidence that oral administration of IFNα-C can prevent the intraocular damage associated with EAU.

Materials and methods

Peptide synthesis

Human lipo-IFNα1 (152–189) peptide was synthesized by GenScript (Piscataway, N.J.). Lipo refers to conjugation with the lipid moiety, palmitoyl-lysine that was conjugated to the N-terminus of the peptide to allow it to penetrate the plasma membrane. The resulting peptide was dissolved in DMSO at 10 mg/ml. Further dilutions to the desired concentration were carried out in PBS under sterile conditions, as described before [30]. In control cells, DMSO at the concentration present in the diluted IFN peptide was added to the cells and is indicated as vehicle control.

Cell culture

ARPE-19 cells were obtained from ATCC (Manassas, VA) and propagated in DMEM/F-12 medium containing 10% FBS and 1% each of Penicillin, Streptomycin in a humidified incubator at 37°C.

Relative quantitation of mRNA expression by RNA extraction and qPCR

ARPE-19 cells were grown overnight in a 12 well plate seeded at 70% confluence. The next day, the medium was changed to DMEM/F-12 containing low FBS (1%). Cells were pre-treated with 3 μM IFNα-C for 4 hr. Cells were then washed with PBS and total RNA was extracted using the RNeasy Mini Kit from QIAGEN, as described before [31]. One microgram of RNA was used to synthesize first strand cDNA using the iScript cDNA synthesis kit from Bio-Rad (Hercules, CA). The primers used for qPCR were synthesized by IDT (Coralville, Iowa) and are listed in Table 1.

The PCR reaction mixture contained cDNA template, SsoFastEvaGreen Super mix containing SYBR green (Bio-Rad, Hercules, CA), and 3 μM target specific primers. After denaturation at 95°C for 2 min, 40 cycles of reaction including denaturation at 94°C for 15 sec followed by annealing at 60°C for 30 sec were carried out using C1000 thermal cycler CFX96 real-time system (Bio-Rad). Gene expression was normalized to beta actin. Relative gene expression was compared with untreated samples and determined using the CFX96 software from Bio-Rad. To investigate the anti-inflammatory properties of IFNα-C, ARPE-19 cells were seeded as above, and pretreated with IFNα-C (3 μM) for 2 hr, followed by TNFα (50 ng/ml) for 4 hr. RNA extraction and qPCR were carried out, as described above. The primers used are listed in Table 1. In these experiments the Ct value for β-actin in untreated samples was 16.1 with a standard deviation of 0.18, and the Ct values for the cytokine/chemokine analytes ranged between 19.03 (CCL-2 following TNFα treatment) to 33.1 (IL-1β in untreated cells).

Quantitation of IL-1β using ELISA

ARPE-19 cells were seeded at 70% confluence in 12 well plate and grown overnight. They were then incubated in medium containing 1% FBS and treated with IFNα-C (3 μM) followed by treatment with TNFα (50 ng/ml) for 24 hr. Supernatants were harvested and used for quantitation of IL-1β in an ELISA format, using a kit from BioLegend (San Diego, CA).

Immunohistochemistry

ARPE-19 cells were seeded in 8 well chambered slides and grown overnight in regular DMEM/F12 medium with 10% FBS. Next day, the medium was changed to DMEM/F12 medium without the serum and pre-treated with IFNα-C (3 μM) for 2 hr followed by addition of TNFα or IL-17A (both at 50 ng/ml) for 30 min. For immunohistochemistry, cells were fixed with 4% paraformaldehyde for 30 min at room temperature and washed with PBS, followed by permeabilization of cells in PBS with 1% Triton X-100 for 30 min at room temperature. Cells were then blocked in 10% normal goat serum in PBS containing 0.5% Triton X-100 for 30 min at room temperature followed by washing in 0.2% Triton X-100 in PBS (wash buffer). Antibody to p65 (cat no. 8242S, Cell Signaling Technology), the active subunit of NF-kB, pSTAT3 (cat no. 9145S, Cell Signaling Technology), or STAT3 (cat no. sc-8019, Santa Cruz Biotechnology) were added to cells and incubated for 2 hr, followed by washing and staining with secondary antibody conjugated with Cy3 (cat no. A10520, Invitrogen) (for p65), or Alexa488 (cat no. A32371, Invitrogen) for pSTAT3 and STAT3, and DAPI for 1 hr. Cells were then washed. For ARPE-19 cells grown for 4 weeks and treatment, antibody to ZO-1 (cat no. 18–7430, Invitrogen) was added and incubated overnight, followed by washing and incubation with Cy-3 conjugated anti-rabbit secondary antibody for 1 hr. After washing, mounting media was added. Cells were covered with a cover slip and imaged in a Keyence BZ-X700 fluorescence microscope.

Measurement of transepithelial electrical resistance (TEER)

We plated the ARPE19 cells on Transwell inserts (Greiner-Bio-one, surface area 33.6 mm2, 0.4 μm pore size) in DMEM/F-12 with 1% FBS for 4 weeks, with a change of media twice per week in order to obtain a differentiated monolayer characterized by tight junctions and a cobblestone appearance [32]. It should be noted that despite their ability to form tight junctions, ARPE-19 monolayers do not form melanosomes and express low levels of marker proteins such as CRALBP and RPE65 [32,33]. Cells were pre-treated with IFNα-C (3 μM) for 4 hr followed by treatment with TNFα (50 ng/ml) for 48 hr. We used an EVOM2 voltohmmeter (World Precision Instruments, Sarasota, FL) to measure transepithelial electrical resistance. The inserts were removed one at a time and placed into the EVOM2 chamber filled with DMEM/F-12 basal media. We calculated the net resistance values by subtracting the value of a blank transwell filter from the value of each filter with plated cells. The samples were run in triplicates and the values show mean with standard deviation.

Experimental autoimmune uveitis and administration of peptide

All procedures were approved by the University of Florida Institutional Animal Care and Use Committee and were conducted in accordance with the Association for Research in Vision and Ophthalmology (ARVO) Statement for the Use of Animals in Ophthalmic and Vision Research. We maintained B10.RIII mice (Jackson Laboratory) in a 12h light: 12 hour dark cycle in a barrier facility. We used B10.RIII mice since they carry H2r allele of the MHC gene and are more amenable to immunization (2, 3). B10.RIII mice (female, 6 to 8 weeks old, n = 8) were immunized with 100 μg of human IRBP160-181 peptide (GenScript, Piscataway, N.J.) dissolved 100 μl PBS emulsified in 100 μl of complete Freund’s adjuvant. Mouse numbers were selected based on the effect size expected from our previous experiments with this model [31]. While sex is not reported to influence the susceptibility to disease, female mice have been typically used in this model [3]. We administered the emulsion at the base of the tail and subcutaneously on the thighs. We administered IFNα-C (200 μg/mouse in 200 μl PBS), or PBS (200 μl) by gavage starting two days before injection of IRBP peptide and continuing daily for three weeks after IRBP injection. The peptide dose was based on our previous study in which oral gavage of IFN peptide had protected mice against lethal dose of vaccinia virus [34] Strategies to overcome barriers to oral delivery of peptide drugs has recently been reviewed by Drucker [35].

Fundoscopy and spectral domain optical coherence tomography (SD-OCT)

These procedures were conducted exactly as described in our previous paper [31]. The health of mice in this study was monitored daily. For these procedures mice were anesthetized with a mixture of ketamine (95 mg/kg) and xylazine (5–10 mg/kg), and at the conclusion of the study mice were euthanized by inhalation of CO2 followed by cervical dislocation. We measured the number of infiltrating cells in the vitreous using Image J software from NIH (URL:https://imagej.nih.gov/ij/), as described before [31]. Briefly, the area representing the vitreous was delineated in the OCT image. The infiltrating cells were converted into binary images and the software quantified the cells in the selected area. We averaged the number of cells in three different B-scans from the stack. We then averaged the number of cells in both eyes from these digital images and averaged.

Electroretinography (ERG)

Electroretinography measurements in dark adapted mice was conducted as described in Ahmed et al. [31].

Splenocyte proliferation assay

Spleens were harvested from IRBP-immunized B10.RIII mice and treated with IFNα-C, four weeks after immunization. Splenocytes were isolated and seeded at 5 x 105 cells per well in RPMI medium containing 10% FBS in a 96 well plate and grown in a humidified chamber at 37°C. IFNα-C (3 μM) was added to cells for 2 hr. IRBP (50 μg/ml) was added to each well and cells were incubated for 72 hr before proliferation was measured using CellTiter 96 Aqueous One cell proliferation assay kit from Promega (Madison, WI). The data represent the average of 5 different mice immunized and treated with IFNα-C, and are shown as average ± standard deviation.

Statistical analysis

For comparison of mean transcript levels to untreated cells, we used Student’s t test for unpaired samples. In this case, samples were not compared to each other but only to the untreated cells for each group. ERG data was analyzed using GraphPad Prism software (San Diego, CA). Two-way ANOVA followed by a post-hoc Sidak test was used to analyze differences between groups at the same light intensity using an adjusted critical value.

Results

IFNα-C induces expression of anti-inflammatory mediators in ARPE-19 cells

We used a spontaneously derived human RPE cell line, ARPE-19 to test the protective effect of IFNα-C. ARPE-19 cells were incubated in the presence or absence of IFNα-C peptide. RNA was extracted from these cells and used to synthesize cDNA and carry out qPCR for target genes. The expression of target genes was normalized to that in untreated cells. Addition of 3 μM IFNα-C to ARPE-19 cells for 4 hrs led to an increase in expression of the anti-inflammatory cytokine, TGFβ (3 fold) (Table 2). The anti-inflammatory proteins TWST1 and TTP that cause post-transcriptional downregulation of mRNA of inflammatory mediators, such as TNFα, IL-6, CCL2, and IFNγ (reviewed in [36,37]), were also induced by 3-fold. The Forkhead family transcription factor, Foxp3 that is required for the production of Tregs that are neuroprotective [38] was enhanced by 2-fold. Together, these results point to an anti-inflammatory environment generated in the presence of IFNα-C.

thumbnail
Table 2. IFNα-C added to ARPE-19 cells causes the induction of anti-inflammatory factors.

https://doi.org/10.1371/journal.pone.0227524.t002

Immunoprotective effects of IFNα-C in ARPE-19 cells treated with TNFα

Tumor necrosis factor α (TNFα) is associated with the onset of uveitis [39,40]. Therefore, we examined the effect of TNFα treatment (50 ng/ml) on ARPE-19 cells for 4 hrs, and determined whether pre-treatment with the IFNαC (3 μM) for 2 hrs suppressed markers of inflammation (Table 3).

thumbnail
Table 3. IFNα-C suppresses the inflammatory cytokines induced by TNFα treatment.

https://doi.org/10.1371/journal.pone.0227524.t003

RNA extracted from these cells was used to carry out qPCR. Inflammatory cytokines IL-1β, IL-6, and the chemokines IL-8 and CCL-2 (both attract leukocytes) were induced to varying degrees. These cytokines and the chemokines were downregulated by 30–50% by the simultaneous presence of IFNαC, thus suggesting an anti-inflammatory role for IFNαC. The induction of IL-1β and its downregulation by IFNαC was further verified by carrying out ELISA of the supernatants from these cells (Fig 1). Treatment of ARPE-19 cells with TNFα (50 ng/ml) for 24 hr resulted in the secretion of 250 ± 40 pg/ml of IL-1β, which was reduced to 71 ± 30 pg/ml, by pretreatment of cells with IFNαC (3 μM) for 2 hr followed by treatment with TNFα (50 ng/ml) for 24 hr, while the presence of solvent and TNFα caused a secretion of 210 ± 20 pg/ml of IL-1β, indicating the ability of IFNαC to reduce an inflammatory response.

thumbnail
Fig 1. IFNα-C suppressed the release of IL-1β from ARPE-19 cells induced with TNFα.

ARPE-19 cells were grown overnight in 12-well plates. They were placed in low serum medium and pre-treated with IFNα-C (3 μM) for 2 hr followed by treatment with TNFα (50 ng/ml) for 24 hr. Supernatants were collected and assayed for IL-1β using ELISA. N = 6. The error bars indicate standard deviation. *, P = 0.01.

https://doi.org/10.1371/journal.pone.0227524.g001

TNFα-associated inflammation activates the NF-κB promoter. We have determined the effect of IFNα-C on NF-κB promoter by following the nuclear translocation of p65, the active subunit of NF-κB. Addition of TNFα (50 ng/ml) to ARPE-19 cells for 30 min caused translocation of p65 to the nucleus, and translocation was prevented in cells that were pretreated with IFNα-C (Fig 2). IL-17A, the product of T helper 17 cells is a major contributor to the onset of uveitis. Since type I IFN is reported to have suppressive effect on IL-17A [41,42], we wanted to test the effect of IFNα-C on the signaling by IL-17. IL-17 acts through the JAK2/STAT3 pathway that causes the nuclear translocation of STAT3 upon its activation [43,44]

thumbnail
Fig 2. Suppression of NF-κB signaling by IFNα-C.

ARPE-19 cells were seeded on 8 well microscopic slides and grown overnight. They were taken in low serum medium and treated with IFNα-C (3 μM) for 2 hr followed by treatment with TNFα (50 ng/ml) for 30 min. Cells were fixed, permeabilized with 1% Triton X-100 (in PBS) and stained with an antibody to p65, followed by staining with Cy3-conjugated secondary antibody and DAPI and imaged using fluorescence microscopy at 40x.

https://doi.org/10.1371/journal.pone.0227524.g002

ARPE-19 cells were pretreated with IFNα-C (3 μM for 2 hr) followed by treatment with IL-17A (50 ng/ml) for 30 min (Fig 3). Nuclear translocation of pSTAT3 caused by IL-17A was prevented when IFNα-C was simultaneously present. As a control, staining carried out with antibody to total STAT3 (inactivate form) did not show any nuclear translocation, as reported in our previous publication [31]. Treatment with IFNα-C can thus reduce inflammatory response in ARPE-19 cells.

thumbnail
Fig 3. IFNα-C prevented signaling from IL-17A.

ARPE-19 cells were seeded on 8 well microscopic slides and grown overnight. They were placed in low serum medium and treated with IFNα-C (3 μM) for 2 hr followed by treatment with IL-17A (50 ng/ml) for 30 min. Cells were fixed, permeabilized with 1% Triton X-100 (in PBS) and stained with an antibody to pSTAT3, followed by staining with Alexa 488-conjugated secondary antibody and DAPI and imaged in a fluorescence microscope at 40x magnification.

https://doi.org/10.1371/journal.pone.0227524.g003

Protection against disruption of barrier properties of ARPE-19 cells by IFNαC.

Tight junction proteins provide a functional barrier on the cell surface of RPE cells. ARPE-19 cells were grown in 8 well microscopic slides for 4 weeks in low serum media until they differentiated and acquired a cobblestone appearance similar to primary RPE cells. During uveitis, elevated levels of cytokines such as TNFα or IL-17A cause disruption of RPE barrier properties [39,45]. These were added to differentiated ARPE-19 cells for 48 hr at 50 ng/ml each. Treatment here was for 48 hr as opposed to 24 hr for freshly cultured cells. We treated differentiated ARPE-19 cells for 48 hours, because they are highly resistant to damage. Some of the cells were pre-incubated with IFNα-C (3 μM) for 4 hr prior to the addition of TNFα or IL-17A. To detect the maintenance of cell boundaries, cells were incubated with a primary antibody to zona occludin 1 (ZO-1) (Fig 4).

thumbnail
Fig 4. IFNα-C prevented the loss of tight junctions caused by TNFα or IL-17A.

ARPE-19 cells grown in 8 well slides in low serum for 4 weeks were pre-treated with IFNα-C (3 μM) for 4 hr followed by TNFα or IL-17A (both at 50 ng/ml) for 48 hr. These were compared to untreated cells and cell treated with TNFα alone. Cells were fixed, permeabilized and stained with an antibody to ZO-1, followed by staining with Cy3-conjugated secondary antibody, and imaged by fluorescence microscopy at 40x magnification.

https://doi.org/10.1371/journal.pone.0227524.g004

Cells that were not treated or those treated only with IFNα-C exhibited continuous staining of ZO-1 at the interface of neighboring cells. Treatment with TNFα or IL-17A disrupted this distribution. When IFNα-C was also present in the cells, the cell surface distribution of ZO-1 was maintained. IFNα-C peptide by itself did not have any effect on the distribution of ZO-1.

The integrity of RPE cell junctions can be monitored by measuring the electrical resistance across the monolayer of cells. We measured transepithelial electrical resistance (TEER) in ARPE-19 cells that had been differentiated by growing them in transwell plates in low serum media for 4 weeks. Cells were incubated with IFNα-C (3 μM) for 4 hr, followed by exposure to TNFα (50 ng/ml) for 48 hr. We measured TEER using a voltohmmeter (Fig 5).

thumbnail
Fig 5. IFNα-C prevents the reduction of transepithelial resistance caused by TNFα.

ARPE-19 cells grown in 24 well transwell plates in low serum medium for 4 weeks were treated with IFNα-C (3 μM) for 4 hr followed by treatment with TNFα (50 ng/ml) for 48 hr. Untreated cells or those treated with TNFα alone were included alongside. TEER, in individual cells in triplicate was measured using EVOM2 voltohmmeter. The error bars indicate standard deviation. *, p = 0.001; **, p = 0.009.

https://doi.org/10.1371/journal.pone.0227524.g005

In cells without IFNα-C, treatment with TNFα led to a 75% reduction in resistance. When IFNα-C was also present, there was no significant loss in TEER caused by TNFα. This result is consistent with the maintenance of tight junction proteins detected by ZO-1 staining (Fig 4). Thus, IFNα-C preserved the distribution and function of tight junction proteins in an inflammatory environment.

Oral delivery of IFNα-C suppresses autoimmune uveitis in mice

Eight female B10.RIII mice were immunized with IRBP emulsified in complete Freund’s adjuvant. Oral gavage with IFNα-C (200 μg in 200 μl PBS per mouse) or with solvent (200 μl PBS per mouse) began on day -2 and was performed daily for 3 weeks after immunization with IRBP peptide. Starting two weeks after immunization, eyes were examined weekly by fundoscopy and SD-OCT. An example of SD-OCT B-scans from day 14 are shown in Fig 6A.

thumbnail
Fig 6. Protection against EAU by oral gavage with IFNα-C.

Eight-week old female B10.RIII mice (n = 8) were gavaged with IFNα-C (200 μg/mouse) or solvent (PBS) in 200 μl on days -2, -1 and 0. On day 0, mice were immunized with IRBP peptide emulsified in complete Freund’s adjuvant. Treatment with IFNα-C or solvent was continued for 3 weeks. A. Optical coherence tomography on day 14. Mice treated with solvent showed influx of inflammatory cells and swelling of retina, which were not seen in mice treated with IFNα-C. B. Fundoscopic images on day 14 show engorged blood vessels and hemorrhage in solvent treated mice. C. Hematoxylin and eosin staining of eyes harvested on day 14 after immunization. Infiltrating cells and retinal folding seen in solvent were not observed in mice treated with IFNα-C. Images were taken at 40x magnification. D. A clinical score, as described in Materials and Methods was used. Mice treated with IFNα-C were protected against the symptoms of EAU. The error bars represent standard deviation. The symptoms did not develop in the following two weeks after termination of treatment. **, p<0.01.

https://doi.org/10.1371/journal.pone.0227524.g006

Control mice (solvent) showed excessive infiltration of cells in the vitreous, optic nerve head, and the retina. Swelling of the retina and deposits were also noted in control eyes. In contrast, eyes from mice that were orally administered IFNα-C showed significantly fewer infiltrating cells and retinal structures were unaffected. On day 14 after immunization, the average number of inflammatory cells in B-scans from three areas of the posterior chamber in PBS treated mice was 70 ± 10, while the scans of similar areas in IFNα-C treated mice showed 4.2 ± 2 cells (p = 0.01, n = 8). Fundoscopic examination of PBS-treated mice showed perivascular deposits, engorged blood vessels, and hemorrhage (Fig 6B), while the eyes from IFNα-C treated mice did not exhibit similar damage. Fourteen days after immunization, eyes were harvested, fixed and stained with hematoxylin and eosin (Fig 6C). Infiltrates of inflammatory cells in retina and vitreous were observed, which is consistent with the observation from SD-OCT scans. We also noted retinal buckling and increased number of nuclei within the inner plexiform layer in the control-treated eyes. The IFNα-C treated eyes, in contrast, were protected from the damage. To evaluate the course of the disease, we employed a clinical score: 0, no disease; 1, numerous infiltrating cells; 2, 1 + engorged blood vessels; 2.5, 2 + hemorrhage and deposits; 3, 2.5 + retinal edema and detachment. The clinical data, including the histology, fundus images, and OCT b-scans were evaluated by 3 masked reviewers who evaluated 5 mice for each treatment at each interval. Their values were averaged for each mouse at each time point. Statistical significance at each interval was determined using the Mann-Whiney test.

Solvent treated mice had reached a clinical score of 2.7 ± 0.45 on day 14, while the IFNα-C treated mice were at 0.4 ± 0.4 (n = 8) (Fig 6D), suggesting the protective effect of IFNα-C against the development of EAU. Although, treatment with IFNα-C was stopped at 3 week, no relapse of the disease symptoms was noted up to 5 weeks when the study was terminated.

To test if the structural damage seen above affected retinal function, we conducted electroretinography (ERG) mice prior to immunization and 4 weeks after the immunization and treatment (Fig 7).

thumbnail
Fig 7. IFNα-C prevented the loss of ERG a- and b- wave amplitudes in EAU mice.

Dark adapted and light adapted electroretinograms were recorded in B10.RIII mice before immunization (pre) and 3 weeks after immunization with IRBP and treatment with solvent (top row) or IFNα-C (bottom row). Average a-wave and b-wave amplitudes are shown. The error bars represent standard deviation. The same cohort of mice were used for all data points. P values were determined by 2-way ANOVA using the Sidak method for multiple comparisons. N = 8.

https://doi.org/10.1371/journal.pone.0227524.g007

Control mice, treated only with PBS, showed 50–60% decline in amplitudes across all intensities in a- and b- waves in both dark-adapted (scotopic) and light-adapted (photopic) conditions. A two-way ANOVA followed by post-hoc Sidak test to identify differences between groups at the same light intensity gave a p value of 0.01 and <0.0001 at -10 and 0 dB, respectively. In contrast to this, mice that were treated with IFNα-C showed a modest decline in scotopic ERG amplitudes that did not reach statistical significance, suggesting the preservation of retinal function by treatment with IFNα-C.

IFNα-C inhibits IRBP-induced ex vivo proliferation of splenocytes from EAU mice

To evaluate the effect of IFNα-C on peripheral immune system, splenocytes from mice four weeks after immunization and treatment with IFNα-C were cultured and their proliferation in response to IRBP was tested in the presence or absence of IFNα-C. As shown in Fig 8, treatment of splenocytes with IRBP induced their proliferation, which was significantly inhibited when IFNα-C was simultaneously present. This result is consistent with the immunomodulation and therapeutic efficacy noted above.

thumbnail
Fig 8. IFNα-C suppressed the IRBP-induced proliferation of splenocytes.

Splenocytes were harvested from IRBP-immunized B10.RIII mice 3 weeks after immunization. Splenocytes (5x105 cells/well) in 96 well plates were grown in RPMI with 10% FBS. IFNα-C (3 μM) was added for 4 hr followed by addition of IRBP (50 μg/ml) and cells were grown for 72 hrs. Cell proliferation was measured as described in Material and Methods. The error bars indicate standard deviation. P = 0.02.

https://doi.org/10.1371/journal.pone.0227524.g008

Discussion

Recombinant type I interferons (IFNα and IFNβ), have been approved for the treatment of a number of malignancies, viral infections and multiple sclerosis [46]. In addition, IFNα has been used in Europe to treat various forms of uveitis [27,47]. However, their use in the clinic is associated with severe toxicity, including lymphopenia, depression and weight loss. As an alternative, we have developed an IFNα mimetic from its C-terminal, denoted as IFNα-C that offers two distinct advantages: 1) In a mouse model of MS, IFNα-C protected mice against the remitting/relapsing episodes of paralysis, without the attendant toxicity seen in the parent IFN [30]; 2) Since receptors for type I IFN are ubiquitous, therapeutic IFN is often “soaked up” by the unwanted cells and tissues before reaching its target organ, which may explain why a higher dose is required to attain therapeutic efficacy. The uptake of interferon by undesired tissues may contribute to its toxicity. We have shown previously that the higher the affinity of the type I IFN binding to its receptor, the greater is its toxicity. For example, IFNα2 bound to its receptor with 10-fold higher affinity than the non-toxic IFNτ [48]. Since the IFNα-C peptide acts independently of binding to the extracellular domain of its receptor, it is conceivable that this property makes it less toxic [49]. In a series of experiments carried out over twenty years, we have demonstrated for both type I and type II IFNs that the N-terminus of the ligand interacts with the extracellular domain of its cognate receptor and determines the species specificity of IFN action, while the C terminus, after endocytosis, binds to the intracellular domain of the receptor and initiates JAK/STAT signaling similar to the parent IFN (reviewed in [49,50]). This model was further tested with the poxvirus decoy receptors for both type I and type II IFNs that are secreted and code only for the extracellular domain of the receptor. The C-terminal peptides from type I or type II IFNs bypassed these decoy receptors and protected mice against lethal dose of vaccinia virus (reviewed in [51,52]). Furthermore, IFNα-C was shown to phosphorylate tyrosine kinase TYK2 and the transcription factor STAT1 in WISH cells [30], confirming the ability of these peptides to recruit the same signaling molecules within the cell as the parent IFN. In future studies, intravitreal delivery of IFNα-C peptide will be investigated to allow more of the effector molecule become available where it is needed, without losing it en route and protecting the individual from toxic side effects.

In this work, we have shown that IFNα-C increases the expression of the transcription factor Foxp3. Foxp3 is required for the generation of Tregs that suppress immune response [38]. In addition, type I IFN can increase the functional activity of regulatory T cells by enabling the conversion of conventional T cells into regulatory T cells. Although, the induction of Foxp3 here was observed in ARPE-19 cells, its effects are most relevant in regulatory T cells. Type I IFN has also been shown to cause the polarization of macrophages into the M2 subtype that have a neuroprotective role [38]. Elevated production of TGFβ as recorded above also contributes to reducing the inflammatory response.

An emerging theme in the regulation of cytokine levels is the production of the following set of proteins, tristetraprolin (TTP), and Twist 1 and 2 that bind to the AU-rich region (ARE) in the 3’-UTR of cytokine mRNAs and cause their degradation. Examples of regulation at the level of mRNA degradation include: TNFα [53], IL-2 [54], IL-6 [55], IL-12 [56], IL-23, IFNγ [57], Ccl2, and Ccl3 [36]. In Table 2, we have demonstrated the ability of IFNα-C to enhance TWST1 and TTP synthesis that are likely to down-regulate the levels of inflammatory cytokines by causing degradation of the corresponding mRNAs.

The retinal pigment epithelium constitutes the outer blood retinal barrier (BRB). Since enhanced levels of TNFα have been noted in uveitis patients, we have demonstrated that IFNα-C counteracts the effect of TNFα by down-regulating the cytokines and chemokines that are induced by TNFα (Table 3). IL-8, the chemokine that recruits neutrophils and leukocytes to the site of inflammation, is also inhibited by type I IFN [58]. Type I IFNs exert anti-inflammatory effects by using a variety of both STAT-dependent and independent mechanisms that contribute to resolution of inflammation. Acting via JAK/STAT pathway, and using a complex consisting of STAT1/STAT2/IRF9 (ISGF3), a number of genes are activated, including suppressor of cytokine signaling 1 (SOCS1). SOCS1 binds to Rac1, an intracellular GTPase, ubiquinylates and degrades it, which leads to a reduction in the generation of reactive oxygen species (ROS) and suppression of the NLRP3 inflammasome [5961]. The presence of inflammatory substances (e.g., complement) and the oxidation products from drusen and other regions in the eye initiate signals for their activation leading to the production of IL-1β and IL-18, resulting in pyroptosis [22]. Membrane rupture from pyroptosis leads to the release of IL-1β, IL-18 and the cellular oxidation products, thus enhancing chemotactic migration of T helper cells and antigen presenting cells. Aside from its role in suppression of NLRP3, SOCS1 (induced by IFN) also acts by binding to the activation domain of JAK2 and TYK2 and the adaptor protein MyD88 adaptor like (MAL), thus limiting the extent and duration of inflammatory responses eliciting from IL-6 [62], IFNγ[62,63], and MyD88 [62]. The end result is the prevention of pathological consequences of uncontrolled expression of inflammatory signals, some examples of which were noted in Table 3.

We have shown the ability of IFNα-C to suppress signaling arising from NF-kB activation. Suppression of NF-kB leads to the downregulation of inflammatory cytokines such as IL-6 and TNFα. In addition, IL-17 production as well as its downstream signaling is suppressed by type I IFN [41, 42, 64], which protects against the damage caused by IL-17 in several autoimmune disorders. We showed that IFNα-C suppressed signaling from IL-17A by blocking activation of STAT3. Type I IFN can also downregulate IFNγ, a product of Th1 cells, by binding to its 3’-UTR and degrading its mRNA [57]. In addition, SOCS 1/3 induced by IFNα is likely to inhibit Th1 response. TGFβ and IL-6 contribute to the polarization of Th17 cells [65, 66], while IL-10 has anti-inflammatory properties and protects against EAU [67]. Either Th1 or the Th17 response may cause autoimmune multiple sclerosis and uveitis [68]. However, blockade of the Th1 response leads to an elevated Th17 response, while a deficiency of Th17 response increases the Th1 response [69]. To effectively treat autoimmune disease, inhibition of both Th1 and Th17 responses is critical. IFNα-C achieves this dual inhibition.

TNFα and IL-17A have been shown to disrupt blood-brain barrier (BBB) by destabilizing tight junctions [45]. We showed that IFNα-C was able to prevent the disruption of ZO-1 distribution on ARPE-19 cells and consequently to counteract the reduction of transepithelial electrical resistance (Figs 4 and 5). Having confirmed the ability of IFNα-C to reduce inflammatory response and to protect against damage to cell permeability properties in ARPE-19 cells, we proceeded to test it ability to protect B10.RIII mice against the development of autoimmune uveitis. These beneficial effects observed on ARPE-19 cells may have contributed to the protection afforded by IFNα-C in mice with EAU, including the prevention of influx of inflammatory cells, and preservation of retinal structure and function.

Cell penetrating peptides (CPPs) offer an attractive alternative to gene therapy approaches [70]. Large amounts of peptides can be generated by chemical synthesis or by bacterial production of recombinant peptides. Attachment of basic amino acids (nine arginines, R9), or conjugation with palmitoyl-lysine allows penetration across plasma membrane. Alternatively, a peptide sequence targeting a specific tissue can be generated by attaching a sequence unique to the target tissue. We have shown previously that a R9-conjugated peptide from SOCS1 kinase inhibitor region (R9-SOCS1-KIR), used topically was effective in protecting against EAU, both prophylactically and therapeutically [31]. Palmitoyl-lysine conjugated SOCS1-KIR was reported by others as effective in preventing EAU [71, 72]. Eye drop formulations of other CPPs, vascular endothelial growth inhibitor (VEGI) [73], pigment epithelium derived factor peptide (PEDF) peptide [74, 75], and endostatin [76] have been reported.

We delivered IFNα-C by oral gavage in these experiments. We have previously shown the therapeutic efficacy of orally administered type II IFN peptide, IFNγ(95–132) in protecting against a lethal dose of vaccinia virus [34]. In earlier work we demonstrated the ability of palmitoyl-lysine conjugated to type II IFN peptide to carry the peptide across the plasma membrane by fusing it with FITC and testing in cells in culture and in mouse peritoneal cells by fluorescence microscopy [77]. However, we have not yet conducted detailed pharmacokinetic experiments of this cell-penetrating protein, and cannot comment about its stability or biodistribution in mice. Orally administered effectors of serotonin receptor were shown by us to protect retinal structure and function in a mouse model of macular degeneration [78.79]. These examples suggest the oral bioavailability of these drugs through recognition by the mucosal immune system. Also, CPP for SOCS1-KIR given i.p. was shown to cross blood-brain barrier to protect against severe form of remitting/relapsing experimental allergic encephalomyelitis (EAE), the mouse model of multiple sclerosis (MS) [62]. Systemic delivery of these peptides is likely to have therapeutic effect in CNS or retinal disorders.

Conclusion

IFNα-C had a protective role in affording an immunosuppressive environment as well in preventing damage caused by the relevant inflammatory cytokines to ARPE-19 cells. These protective properties may have contributed to the preservation of structural and functional properties during the course of autoimmune uveitis in mice. It is conceivable that these properties will also be helpful in treating other inflammatory eye diseases, such as age related macular degeneration (AMD) and autoimmune keratitis.

Supporting information

S1 Checklist. The ARRIVE guidelines checklist animal research: Reporting in vivo experiments.

https://doi.org/10.1371/journal.pone.0227524.s001

(PDF)

Acknowledgments

This project was supported by the Shaler Richardson Professorship Endowment, which had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript. The University of Florida been awarded a patent (US 9,951,111B2) governing the interferon peptide mimetic used in this study. Two of the authors (CMA and HMJ) may obtain royalties from the licensing of this technology.

References

  1. 1. Miserocchi E, Fogliato G, Modorati G, Bandello F. Review on the worldwide epidemiology of uveitis. Eur J Ophthalmol. 2013;23(5):705–17. pmid:23661536
  2. 2. Chen J, Caspi RR. Clinical and Functional Evaluation of Ocular Inflammatory Disease Using the Model of Experimental Autoimmune Uveitis. Methods Mol Biol. 2019;1899:211–27. pmid:30649775
  3. 3. Agarwal RK, Silver PB, Caspi RR. Rodent models of experimental autoimmune uveitis. Methods Mol Biol. 2012;900:443–69. pmid:22933083
  4. 4. Pan J, Kapur M, McCallum R. Noninfectious immune-mediated uveitis and ocular inflammation. Curr Allergy Asthma Rep. 2014;14(1):409. pmid:24338488
  5. 5. Frohman LP. Treatment of Neuro-Ophthalmic Sarcoidosis. J Neuroophthalmol. 2014.
  6. 6. Park UC, Kim TW, Yu HG. Immunopathogenesis of ocular Behçet's disease. J Immunol Res. 2014;2014:653539. pmid:25061613
  7. 7. Greco A, Fusconi M, Gallo A, Turchetta R, Marinelli C, Macri GF, et al. Vogt-Koyanagi-Harada syndrome. Autoimmun Rev. 2013;12(11):1033–8. pmid:23567866
  8. 8. Abbasian J, Martin TM, Patel S, Tessler HH, Goldstein DA. Immunologic and genetic markers in patients with idiopathic ocular inflammation and a family history of inflammatory bowel disease. Am J Ophthalmol. 2012;154(1):72–7. pmid:22464367
  9. 9. Walscheid K, Hennig M, Heinz C, Wasmuth S, Busch M, Bauer D, et al. Correlation between disease severity and presence of ocular autoantibodies in juvenile idiopathic arthritis-associated uveitis. Invest Ophthalmol Vis Sci. 2014;55(6):3447–53. pmid:24764059
  10. 10. Shugaiv E, Tüzün E, Kürtüncü M, Kıyat-Atamer A, Çoban A, Akman-Demir G, et al. Uveitis as a prognostic factor in multiple sclerosis. Mult Scler. 2015;21(1):105–7. pmid:24948689
  11. 11. Messenger W, Hildebrandt L, Mackensen F, Suhler E, Becker M, Rosenbaum JT. Characterisation of uveitis in association with multiple sclerosis. Br J Ophthalmol. 2015;99(2):205–9. pmid:25170065
  12. 12. Perez VL, Caspi RR. Immune mechanisms in inflammatory and degenerative eye disease. Trends Immunol. 2015;36(6):354–63. pmid:25981967
  13. 13. Forrester JV, Klaska IP, Yu T, Kuffova L. Uveitis in mouse and man. Int Rev Immunol. 2013;32(1):76–96. pmid:23360160
  14. 14. Horai R, Silver PB, Chen J, Agarwal RK, Chong WP, Jittayasothorn Y, et al. Breakdown of immune privilege and spontaneous autoimmunity in mice expressing a transgenic T cell receptor specific for a retinal autoantigen. J Autoimmun. 2013;44:21–33. pmid:23810578
  15. 15. Heissigerova J, Seidler Stangova P, Klimova A, Svozilkova P, Hrncir T, Stepankova R, et al. The Microbiota Determines Susceptibility to Experimental Autoimmune Uveoretinitis. J Immunol Res. 2016;2016:5065703. pmid:27294159
  16. 16. Horai R, Caspi RR. Microbiome and Autoimmune Uveitis. Front Immunol. 2019;10:232. pmid:30837991
  17. 17. Rowan S, Taylor A. The Role of Microbiota in Retinal Disease. Adv Exp Med Biol. 2018;1074:429–35. pmid:29721973
  18. 18. Touhami S, Diwo E, Sève P, Trad S, Bielefeld P, Sène D, et al. Expert opinion on the use of biological therapy in non-infectious uveitis. Expert Opin Biol Ther. 2019;19(5):477–90. pmid:30888881
  19. 19. Gomes Bittencourt M, Sepah YJ, Do DV, Agbedia O, Akhtar A, Liu H, et al. New treatment options for noninfectious uveitis. Dev Ophthalmol. 2012;51:134–61. pmid:22517211
  20. 20. Lee FF, Foster CS. Pharmacotherapy of uveitis. Expert Opin Pharmacother. 2010;11(7):1135–46. pmid:20367272
  21. 21. Benveniste EN, Qin H. Type I interferons as anti-inflammatory mediators. Sci STKE. 2007;2007(416):pe70. pmid:18073382
  22. 22. Guarda G, Braun M, Staehli F, Tardivel A, Mattmann C, Förster I, et al. Type I interferon inhibits interleukin-1 production and inflammasome activation. Immunity. 2011;34(2):213–23. pmid:21349431
  23. 23. González-Navajas JM, Lee J, David M, Raz E. Immunomodulatory functions of type I interferons. Nat Rev Immunol. 2012;12(2):125–35. pmid:22222875
  24. 24. Dumitrescu L, Constantinescu CS, Tanasescu R. Recent developments in interferon-based therapies for multiple sclerosis. Expert Opin Biol Ther. 2018;18(6):665–80. pmid:29624084
  25. 25. Hile GA, Gudjonsson JE, Kahlenberg JM. The influence of interferon on healthy and diseased skin. Cytokine. 2018.
  26. 26. Pott J, Stockinger S. Type I and III Interferon in the Gut: Tight Balance between Host Protection and Immunopathology. Front Immunol. 2017;8:258. pmid:28352268
  27. 27. Lewczuk N, Zdebik A, Bogusławska J. Interferon Alpha 2a and 2b in Ophthalmology: A Review. J Interferon Cytokine Res. 2019;39(5):259–72. pmid:30950678
  28. 28. Mackensen F, Max R, Becker MD. Interferons and their potential in the treatment of ocular inflammation. Clin Ophthalmol. 2009;3:559–66. pmid:19898628
  29. 29. Mackensen F, Jakob E, Springer C, Dobner BC, Wiehler U, Weimer P, et al. Interferon versus methotrexate in intermediate uveitis with macular edema: results of a randomized controlled clinical trial. Am J Ophthalmol. 2013;156(3):478-86.e1.
  30. 30. Ahmed CM, Johnson HM. Short Peptide type I interferon mimetics: therapeutics for experimental allergic encephalomyelitis, melanoma, and viral infections. J Interferon Cytokine Res. 2014;34(10):802–9. pmid:24811478
  31. 31. Ahmed CM, Massengill MT, Brown EE, Ildefonso CJ, Johnson HM, Lewin AS. A cell penetrating peptide from SOCS-1 prevents ocular damage in experimental autoimmune uveitis. Exp Eye Res. 2018;177:12–22. pmid:30048621
  32. 32. Dunn KC, Aotaki-Keen AE, Putkey FR, Hjelmeland LM. ARPE-19, a human retinal pigment epithelial cell line with differentiated properties. Exp Eye Res. 1996;62(2):155–69. pmid:8698076
  33. 33. Ablonczy Z, Dahrouj M, Tang PH, Liu Y, Sambamurti K, Marmorstein AD, et al. Human retinal pigment epithelium cells as functional models for the RPE in vivo. Invest Ophthalmol Vis Sci. 2011;52(12):8614–20. pmid:21960553
  34. 34. Ahmed CM, Martin JP, Johnson HM. IFN mimetic as a therapeutic for lethal vaccinia virus infection: possible effects on innate and adaptive immune responses. J Immunol. 2007;178(7):4576–83. pmid:17372016
  35. 35. Drucker DJ. Advances in oral peptide therapeutics. Nat Rev Drug Discov. 2019.
  36. 36. Kovarik P, Ebner F, Sedlyarov V. Posttranscriptional regulation of cytokine expression. Cytokine. 2015.
  37. 37. Palanisamy V, Jakymiw A, Van Tubergen EA, D'Silva NJ, Kirkwood KL. Control of cytokine mRNA expression by RNA-binding proteins and microRNAs. J Dent Res. 2012;91(7):651–8. pmid:22302144
  38. 38. Lee SE, Li X, Kim JC, Lee J, González-Navajas JM, Hong SH, et al. Type I interferons maintain Foxp3 expression and T-regulatory cell functions under inflammatory conditions in mice. Gastroenterology. 2012;143(1):145–54. pmid:22475534
  39. 39. Al-Gayyar MM, Elsherbiny NM. Contribution of TNF-alpha to the development of retinal neurodegenerative disorders. Eur Cytokine Netw. 2013;24(1):27–36. pmid:23608634
  40. 40. Thomas AS. Biologics for the treatment of noninfectious uveitis: current concepts and emerging therapeutics. Curr Opin Ophthalmol. 2019;30(3):138–50. pmid:30844945
  41. 41. Guo B, Chang EY, Cheng G. The type I IFN induction pathway constrains Th17-mediated autoimmune inflammation in mice. J Clin Invest. 2008;118(5):1680–90. pmid:18382764
  42. 42. Moschen AR, Geiger S, Krehan I, Kaser A, Tilg H. Interferon-alpha controls IL-17 expression in vitro and in vivo. Immunobiology. 2008;213(9–10):779–87. pmid:18926293
  43. 43. Xu Y, Xu X, Gao X, Chen H, Geng L. Shikonin suppresses IL-17-induced VEGF expression via blockage of JAK2/STAT3 pathway. Int Immunopharmacol. 2014;19(2):327–33. pmid:24521871
  44. 44. Ganesan R, Rasool M. Interleukin 17 regulates SHP-2 and IL-17RA/STAT-3 dependent Cyr61, IL-23 and GM-CSF expression and RANKL mediated osteoclastogenesis by fibroblast-like synoviocytes in rheumatoid arthritis. Mol Immunol. 2017;91:134–44. pmid:28898718
  45. 45. Weinstein JE, Pepple KL. Cytokines in uveitis. Curr Opin Ophthalmol. 2018;29(3):267–74. pmid:29521875
  46. 46. Reder AT, Feng X. How type I interferons work in multiple sclerosis and other diseases: some unexpected mechanisms. J Interferon Cytokine Res. 2014;34(8):589–99. pmid:25084175
  47. 47. Plskova J, Greiner K, Forrester JV. Interferon-alpha as an effective treatment for noninfectious posterior uveitis and panuveitis. Am J Ophthalmol. 2007;144(1):55–61. pmid:17601428
  48. 48. Subramaniam PS, Khan SA, Pontzer CH, Johnson HM. Differential recognition of the type I interferon receptor by interferons tau and alpha is responsible for their disparate cytotoxicities. Proc Natl Acad Sci U S A. 1995;92(26):12270–4. pmid:8618883
  49. 49. Johnson HM, Noon-Song E, Ahmed CM. Noncanonical IFN Signaling, Steroids, and STATs: A Probable Role of V-ATPase. Mediators Inflamm. 2019;2019:4143604. pmid:31275057
  50. 50. Johnson HM, Noon-Song EN, Dabelic R, Ahmed CM. IFN signaling: how a non-canonical model led to the development of IFN mimetics. Front Immunol. 2013;4:202. pmid:23898330
  51. 51. Ahmed CM, Johnson HM. The role of a non-canonical JAK-STAT pathway in IFN therapy of poxvirus infection and multiple sclerosis: An example of Occam's Broom? JAKSTAT. 2013;2(4):e26227. pmid:24416655
  52. 52. Ahmed CM, Johnson HM. Type I interferon mimetics bypass vaccinia virus decoy receptor virulence factor for protection of mice against lethal infection. Clin Vaccine Immunol. 2014;21(8):1178–84. pmid:24964806
  53. 53. Sauer I, Schaljo B, Vogl C, Gattermeier I, Kolbe T, Müller M, et al. Interferons limit inflammatory responses by induction of tristetraprolin. Blood. 2006;107(12):4790–7. pmid:16514065
  54. 54. Ogilvie RL, Abelson M, Hau HH, Vlasova I, Blackshear PJ, Bohjanen PR. Tristetraprolin down-regulates IL-2 gene expression through AU-rich element-mediated mRNA decay. J Immunol. 2005;174(2):953–61. pmid:15634918
  55. 55. Zhao W, Liu M, D'Silva NJ, Kirkwood KL. Tristetraprolin regulates interleukin-6 expression through p38 MAPK-dependent affinity changes with mRNA 3' untranslated region. J Interferon Cytokine Res. 2011;31(8):629–37. pmid:21457063
  56. 56. Gu L, Ning H, Qian X, Huang Q, Hou R, Almourani R, et al. Suppression of IL-12 production by tristetraprolin through blocking NF-kcyB nuclear translocation. J Immunol. 2013;191(7):3922–30. pmid:23997224
  57. 57. Ogilvie RL, Sternjohn JR, Rattenbacher B, Vlasova IA, Williams DA, Hau HH, et al. Tristetraprolin mediates interferon-gamma mRNA decay. J Biol Chem. 2009;284(17):11216–23. pmid:19258311
  58. 58. Laver T, Nozell SE, Benveniste EN. IFN-beta-mediated inhibition of IL-8 expression requires the ISGF3 components Stat1, Stat2, and IRF-9. J Interferon Cytokine Res. 2008;28(1):13–23. pmid:18370868
  59. 59. Inoue M, Shinohara ML. NLRP3 Inflammasome and MS/EAE. Autoimmune Dis. 2013;2013:859145. pmid:23365725
  60. 60. Ildefonso CJ, Biswal MR, Ahmed CM, Lewin AS. The NLRP3 Inflammasome and its Role in Age-Related Macular Degeneration. Adv Exp Med Biol. 2016;854:59–65. pmid:26427394
  61. 61. Elliott EI, Sutterwala FS. Initiation and perpetuation of NLRP3 inflammasome activation and assembly. Immunol Rev. 2015;265(1):35–52. pmid:25879282
  62. 62. Jager LD, Dabelic R, Waiboci LW, Lau K, Haider MS, Ahmed CM, et al. The kinase inhibitory region of SOCS-1 is sufficient to inhibit T-helper 17 and other immune functions in experimental allergic encephalomyelitis. J Neuroimmunol. 2011;232(1–2):108–18. pmid:21131060
  63. 63. Ahmed CM, Larkin J, Johnson HM. SOCS1 Mimetics and Antagonists: A Complementary Approach to Positive and Negative Regulation of Immune Function. Front Immunol. 2015;6:183. pmid:25954276
  64. 64. Guo B. IL-10 Modulates Th17 Pathogenicity during Autoimmune Diseases. J Clin Cell Immunol. 2016;7(2).
  65. 65. Steinman L. A brief history of T(H)17, the first major revision in the T(H)1/T(H)2 hypothesis of T cell-mediated tissue damage. Nat Med. 2007;13(2):139–45. pmid:17290272
  66. 66. Park H, Li Z, Yang XO, Chang SH, Nurieva R, Wang YH, et al. A distinct lineage of CD4 T cells regulates tissue inflammation by producing interleukin 17. Nat Immunol. 2005;6(11):1133–41. pmid:16200068
  67. 67. Rizzo LV, Xu H, Chan CC, Wiggert B, Caspi RR. IL-10 has a protective role in experimental autoimmune uveoretinitis. Int Immunol. 1998;10(6):807–14. pmid:9678762
  68. 68. Horai R, Caspi RR. Cytokines in autoimmune uveitis. J Interferon Cytokine Res. 2011;31(10):733–44. pmid:21787221
  69. 69. Chong WP, Horai R, Mattapallil MJ, Silver PB, Chen J, Zhou R, et al. IL-27p28 inhibits central nervous system autoimmunity by concurrently antagonizing Th1 and Th17 responses. J Autoimmun. 2014;50:12–22. pmid:24021664
  70. 70. Kristensen M, Birch D, Mørck Nielsen H. Applications and Challenges for Use of Cell-Penetrating Peptides as Delivery Vectors for Peptide and Protein Cargos. Int J Mol Sci. 2016;17(2).
  71. 71. He C, Yu CR, Mattapallil MJ, Sun L, Larkin Iii J, Egwuagu CE. SOCS1 Mimetic Peptide Suppresses Chronic Intraocular Inflammatory Disease (Uveitis). Mediators Inflamm. 2016;2016:2939370. pmid:27703302
  72. 72. He C, Yu CR, Sun L, Mahdi RM, Larkin J 3rd, Egwuagu CE. Topical administration of a suppressor of cytokine signaling-1 (SOCS1) mimetic peptide inhibits ocular inflammation and mitigates ocular pathology during mouse uveitis. J Autoimmun. 2015;62:31–8. pmid:26094775
  73. 73. Gao W, Zhao Z, Yu G, Zhou Z, Zhou Y, Hu T, et al. VEGI attenuates the inflammatory injury and disruption of blood-brain barrier partly by suppressing the TLR4/NF-kappaB signaling pathway in experimental traumatic brain injury. Brain Res. 2015;1622:230–9. pmid:26080076
  74. 74. Liu Y, Leo LF, McGregor C, Grivitishvili A, Barnstable CJ, Tombran-Tink J. Pigment epithelium-derived factor (PEDF) peptide eye drops reduce inflammation, cell death and vascular leakage in diabetic retinopathy in Ins2(Akita) mice. Mol Med. 2012;18:1387–401. pmid:23019073
  75. 75. Vigneswara V, Esmaeili M, Deer L, Berry M, Logan A, Ahmed Z. Eye drop delivery of pigment epithelium-derived factor-34 promotes retinal ganglion cell neuroprotection and axon regeneration. Mol Cell Neurosci. 2015;68:212–21. pmid:26260110
  76. 76. Zhang X, Li Y, Cheng Y, Tan H, Li Z, Qu Y, et al. Tat PTD-endostatin: A novel anti-angiogenesis protein with ocular barrier permeability via eye-drops. Biochim Biophys Acta. 2015;1850(6):1140–9. pmid:25662072
  77. 77. Ahmed CM, Dabelic R, Martin JP, Jager LD, Haider SM, Johnson HM. Enhancement of antiviral immunity by small molecule antagonist of suppressor of cytokine signaling. J Immunol. 2010;185(2):1103–13. pmid:20543109
  78. 78. Ahmed CM, Biswal MR, Li H, Han P, Ildefonso CJ, Lewin AS. Repurposing an orally available drug for the treatment of geographic atrophy. Mol Vis. 2016;22:294–310. pmid:27110092
  79. 79. Biswal MR, Ahmed CM, Ildefonso CJ, Han P, Li H, Jivanji H, et al. Systemic treatment with a 5HT1a agonist induces anti-oxidant protection and preserves the retina from mitochondrial oxidative stress. Exp Eye Res. 2015;140:94–105. pmid:26315784