DOI QR코드

DOI QR Code

PEP-1-FK506BP inhibits alkali burn-induced corneal inflammation on the rat model of corneal alkali injury

  • Kim, Dae Won (Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangnung-Wonju National University) ;
  • Lee, Sung Ho (R&D Center, Lumieye Genetics Co., Ltd.) ;
  • Shin, Min Jea (Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University) ;
  • Kim, Kibom (R&D Center, Lumieye Genetics Co., Ltd.) ;
  • Ku, Sae Kwang (Department of Anatomy and Histology, College of Korean Medicine, Daegu Haany University) ;
  • Youn, Jong Kyu (Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University) ;
  • Cho, Su Bin (Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University) ;
  • Park, Jung Hwan (Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University) ;
  • Lee, Chi Hern (Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University) ;
  • Son, Ora (Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University) ;
  • Sohn, Eun Jeong (Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University) ;
  • Cho, Sung-Woo (Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine) ;
  • Park, Jong Hoon (Department of Biological Sciences, Sookmyung Women's University) ;
  • Kim, Hyun Ah (Division of Rheumatology, Department of Internal Medicine, Hallym University Sacred Heart Hospital) ;
  • Han, Kyu Hyung (Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University) ;
  • Park, Jinseu (Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University) ;
  • Eum, Won Sik (Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University) ;
  • Choi, Soo Young (Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University)
  • Received : 2015.03.04
  • Accepted : 2015.03.26
  • Published : 2015.11.30

Abstract

FK506 binding protein 12 (FK506BP) is a small peptide with a single FK506BP domain that is involved in suppression of immune response and reactive oxygen species. FK506BP has emerged as a potential drug target for several inflammatory diseases. Here, we examined the protective effects of directly applied cell permeable FK506BP (PEP-1-FK506BP) on corneal alkali burn injury (CAI). In the cornea, there was a significant decrease in the number of cells expressing pro-inflammation, apoptotic, and angiogenic factors such as TNF-α, COX-2, and VEGF. Both corneal opacity and corneal neovascularization (CNV) were significantly decreased in the PEP-1-FK506BP treated group. Our results showed that PEP-1-FK506BP can significantly inhibit alkali burn-induced corneal inflammation in rats, possibly by accelerating corneal wound healing and by reducing the production of angiogenic factors and inflammatory cytokines. These results suggest that PEP-1-FK506BP may be a potential therapeutic agent for CAI.

Keywords

INTRODUCTION

Alkali injuries of the eye represent one of the most serious forms of eye trauma and may cause extensive damage to the ocular surface epithelium, cornea, and anterior segment, resulting in irreversible vision loss (1-3). Corneal alkali burn injury (CAI) occurs rarely, but is accompanied by a poor prognosis for rehabilitation. Acute inflammation of CAI is characterized by rapid infiltration of neutrophils into the cornea; the chronic inflammation involves migration and recruitment of inflammatory cells over extended periods into the cornea with resultant damage to the normal tissue. This damage is induced by secretion of proteolytic enzymes and/or oxidative derivatives into the extracellular matrix (4). In an effort to preserve epithelial integrity and prevent stromal ulceration, a number of therapeutic strategies have been tried, including treatment with fibronectin, retinoic acid, sodium hyaluronate, as well as conjunctival transplant (5-8). In this report, we tested the anti-inflammatory effect of PEP-1-FK506BP and found it to be an effective anti-inflammatory agent in a CAI rat model. The rat model of CAI has been used as a valuable animal model for developing ocular drugs (9-11).

FK506-binding proteins (FK506BPs) belong to a family of immunophilins named for their ability to bind to immunosuppressive drugs. FK506BP is a small peptide (12 kDa) with a single FK506BP domain. FK506BP is involved in multiple biological processes, including binding to rapamycin and the formation of a complex of rapamycin-mediated PI3K/Akt (12, 13). It has been demonstrated that protein transduction domains (PTDs) can deliver various exogenous molecules into living cells and tissues. In a previous study, we showed that topical application of PEP-1-FK506BP to mice ears significantly inhibited 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced inflammation (14-18).

Inflammatory enzymes such as cyclooxygenase-2 (COX-2) as well as cytokines, IL-1β, and TNF- α contribute to the pathogenesis of several inflammatory diseases (19, 20). COX-2 is an inducible isoform generated by a number of pro-inflammatory stimuli, lipopolysaccharide (LPS) and tumor promoters including TPA (21, 22). Additionally, it is well known that COX-2, Caspase-3, and vascular endothelial growth factor (VEGF) expression levels are increased in conjunction with CAI (23-25). It has been shown that treatment with PEP-1-FK506BP decreased elevated levels of pro-inflammatory cytokines in dry eye syndrome and ameliorated atopic dermatitis. In this report, we investigated the protective effects of PEP-1-FK506BP in a rat CAI model. The results suggest that PEP-1-FK506BP may be a potential drug target for CAI, although the precise action of FK506BP needs to be further elucidated.

 

RESULTS AND DISCUSSION

Changes on the epithelial wound healing

Topical application of PEP-1-FK506BP to CAI-induced rats significantly inhibited corneal damage and infiltration of mononuclear cells which express apoptosis markers, pro-inflammatory cytokines, angiogeneic factors, and inflammatory factors. This study demonstrated that a PEP-1-FK506BP protein can be directly transduced into mouse corneal and conjunctival tissue where it effectively rescued mice from Botulinum toxin A-induced dry eye (26). The protective mechanism of PEP-1-FK506BP needs further exploration, but it has also been shown that PEP-1-FK506BP protected several cells from oxidative stress-induced cell toxicity and could be a potential therapeutic agent against atopic dermatitis because of its anti-inflammatory effects (26-28).

The potential consequence of PEP-1-FK506BP on wound healing of the cornea was examined using a corneal alkali injury (CAI) rat model. Corneal reepithelialization after CAI plays a crucial role in the cessation of eye damage (29). Significant (P < 0.01) increases in corneal damage were detected in the CAI control group throughout experimental periods compared with intact control and correspond well with previous studies (30, 31). 0.1% sodium hyaluronate was used as a positive control to test the in vivo efficacy of PEP-1-FK506BP on CAI. Corneal damage was significantly decreased in the 0.1% sodium hyaluronate treated control group. Topically applied PEP-1-FK506BP effectively decreased corneal damage from 7-14 days after initiation of treatment (CAI to one group and 0.1% sodium hyaluronate to another group) (Fig. 1). The extent of damage in the PEP-1-FK506BP treated group was similar to that of the 0.1% sodium hyaluronate treated control group.

Fig. 1.Changes in corneal damage during topical application of test materials (5 μl/eye; twice a day for 14 days) in CAI-induced rats. 5 mm of filter paper was soaked with 1N NaOH and was placed onto the center of cornea for 60 seconds to induce CAI. After rinsing with sterile saline (10 ml) as previously described, corneal damage was monitored for 14 days (20). Saline soaked filter papers were used for the intact control in place of alkali soaked filter paper. For each treated group, 5 μl of PEP-1-FK506BP or 0.1% sodium hyaluronate (Samil Pharm. Co., Korea) was topically administered to eyes, twice a day for 14 days.

CAI induced histopathologic changes on eye

Acute corneal inflammation is characterized by rapid infiltration of neutrophils into the cornea. Chronic inflammation involves recruitment and migration of inflammatory cells over extended periods into the cornea with resultant damage to the normal tissue due to release of proteolytic enzymes and/or oxidative derivatives into the extracellular matrix (4, 32).

To evaluate the degree of corneal damage to rats, inflammatory cells were detected on the cornea, anterior chambers, and lens by H&E staining (Fig. 2). Note that infiltration of inflammatory cells was detected in CAI induced rats. Numerous mononuclear inflammatory cells infiltrated into stroma, cornea, anterior chambers, and lens in the CAI control group (Figs. 2D-F). Damage to surrounding tissue and stroma neovascularization (arrows) in corneal epithelia was also dramatically increased in the CAI control group (Figs. 2D-F). This corneal damage was significantly inhibited in both the hyaluronate and PEP-1-FK506BP treated groups (Fig. 2G-L) relative to the CAI control. Not only were the number of infiltrated inflammatory cells in the cornea, but also various histopathologic changes including thicknesses of cornea, anterior chambers, and neovasculatures decreased to the same degree in both of PEP-1-FK506BP and sodium hyaluronate treated groups.

Fig. 2.Anti-inflammatory effects of PEP-1-FK506BP in corneal tissue. Images of H&E staining of corneal tissues of CAI induced rats after PEP-1-FK506BP or 0.1% sodium hyaluronate treatment as well as intact control and CAI controls. H&E staining of cornea to anterior chambers in the intact control (A-C), CAI control (D-F), 0.1% sodium hyaluronate (G-I), and PEP-1-FK506BP (J-L) treated groups. Scale bars = 200 μm. Original magnification ×40 and ×200 (EP, epithelium; ST, stroma; AC, anterior chambers; LE, lens).

CAI induced infiltration of immunoreactive cells

To test the protective effects of PEP-1-FK506BP on eye inflammation, we evaluated the number of infiltrated immunoreactive mononuclear cells after corneal alkali injury. Infiltration of inflammatory cells, mainly polymorphoneutrophils (PMNs) on the cornea and anterior chambers, between the inner limiting membranes of the cornea and the lens are common histopathological features of CAI. Infiltration of PMNs induce corneal neovascularization, fibrosis, and increases corneal thicknesses (33). The number of cells which express apoptotic markers (caspase-3 and PARP), pro-inflammatory cytokines (TNF-α and IL-1β), and angiogenic or inflammatory factors (VEGF, COX-2, and iNOS) were counted by immunohistochemistry. VEGF, COX-2, IL-1β, and iNOS immunoreactive cells were remarkably increased in corneal epithelium (Fig. 3). Statistically significant (P < 0.01) decreases in the numbers of caspase-3, PARP, TNF-α, VEGF, COX-2, IL-1β, and iNOS-immunoreactive cells were observed in the corneal epithelium in response to PEP-1-FK506BP treatment. Significant (P < 0.01) decreases of caspase-3, PARP, TNF-α, COX-2, and iNOS were detected while VEGF and IL-1β-immunoreactive cells decreased non-significantly in 0.1% sodium hyaluronate treated rats compared with CAI control rats. The overall inhibitory efficacy of test materials on the eye demonstrated that PEP-1-FK506BP may provide a therapeutic agent for immunoreactives of apoptotic markers, pro-inflammatory cytokines, and angiogenic factors.

Fig. 3.Changes in the number of immunoreactive cells in cornea epithelium. After CAI inducement, for each group of rats 5 μl/rat of PEP-1-FK506BP or 0.1% sodium hyaluronate was tropically applied twice per day for 14 days. At day 14, anti-inflammatory, anti-angiogeneic, and anti-apoptotic effects of PEP-1- FK506BP in corneal epithelia were evaluated by IHC of corneal tissue. IHC images after CAI were digitized and each immune reactive cell was counted. (A) TNF-α, (B) IL-1β, (C) COX-2, (D) VEGF, (E) iNOS, (F) Caspase-3, an d (G) PARP immunoreactive cells were counted in cornea epithelium. Values are expressed as Mean ± SD of ten rats. *P < 0.01 as compared with intact control by LSD test. #P < 0.01 as compared with CAI control by LSD test.

PEP-1-FK506BP reduced CAI induced corneal opacity and angiogenesis

Cornea opacification and CNV have been used as a predictable index for detecting the efficacy of various test materials in CAI models (34, 35). In this system, cornea opacification was evaluated using a corneal opacity scoring system of subcorneal architectures including the pupil, iris vessels, or anterior chambers (36). In the present study, marked increases in corneal opacification grades (level 5: the highest level) were detected in the CAI control group after 1N NaOH exposure. PEP-1- FK506BP significant protected against the effect on the cornea from alkali injury. Sodium hyaluronate had a statistically significant positive influence on epithelial re-surfacing, especially during the late healing phase (37, 38), and is used as a reference drug in CAI experiments (39) because of its favorable anti-inflammatory and anti-oxidative effects (40, 41). Corneal opacity scores were significantly increased in the CAI control group immediately after CAI induction and throughout the experimental period compared with the intact control group. However, these increases in corneal opacity scores were significantly inhibited by PEP-1-FK506BP treatment or 0.1% sodium hyaluronate from day 2 and 5 after initiation of topical application, respectively. The protective effects of PEP-1-FK506BP were greater than that of 0.1% sodium hyaluronate (Fig. 4A).

Fig. 4.Effect of PEP-1-FK506BP on corneal opacity and neovascularization after alkali injury. (A) Corneal opacity scores and (B) CNV scores were evaluated for 2 weeks following the CAI challenge. Values are expressed as Mean ± SD for 10 rats in each group. *P < 0.05, between PEP-1-FK506BP treated rats and intact control rats by LSD test.

The protective effect of PEP-1-FK506BP on CNV has been studied in this report since neovascularization is directly related to vision loss (42-44). CNV was also evaluated using neovascularization scores for 2 weeks from the CAI induction (Fig. 4B). There were significant (P < 0.01) increases in corneal neovascularization scores in the CAI control group from day 1 after CAI induction and throughout the experimental period. These increases in corneal neovascularization scores were significantly (P < 0.01 or P < 0.05) inhibited by treatment with 0.1% sodium hyaluronate and PEP-1-FK506BP from day 6 and day 5, respectively. The cornea neovascularization scores at the end point were −10.22 and −21.78% in the 0.1% sodium hyaluronate and PEP-1-FK506BP treated groups, respectively. This indicates that PEP-1-FK506BP might be a more effective therapeutic agent for corneal alkali burn induced neovascularization and related vision losses than 0.1% sodium hyaluronate. From these results, we suggest PEP-1-FK506BP is a promising therapeutic lead molecule for corneal reepithelialization after alkali injury.

 

MATERIALS AND METHODS

Animals

All procedures used in this study were in accordance with the principles of the Association for Research in Vision and Ophthalmology (ARVO) Statement for the Use of Animals in Ophthalmic and Vision Research. All experimental procedures involving animals and their care were in accordance with the Guide for the Care and Use of Laboratory Animals of the National Veterinary Research and Quarantine Service of Korea, and were approved by the Hallym Medical Center Institutional Animal Care and Use Committee. 6-week-old male Sprague-Dawley rats (SLC, JAPAN) were used after acclimatization for 7 days.

Induction of CAI

The rats (n = 10 per group) were anesthetized by 25 mg/kg intraperitoneal injection of Zoletile 50 (Virbac Lab., France). To induce alkali injuries, 5 mm in diameter filter paper soaked with 1N NaOH was placed on the central cornea for 60 seconds. The eyes were then rinsed with sterile saline (10 ml) as previously described (33). For the intact control, saline soaked filter papers were placed on the cornea, in place of the alkali soaked paper. For each treated group, 5 μl of PEP-1- FK506BP or 0.1% sodium hyaluronate (Samil Pharm. Co., Korea) was topically administered to eyes, twice a day for 14 days. In intact and CAI control groups, saline was applied instead of test materials. Changes in corneal opacity, neovascularization, and cornea epithelial wound healings were monitored via histopathological observation for 14 days.

Histomorphometry

The proportion of damaged corneal epithelium region, (%/5 mm of corneal epithelial lining), total thickness of cornea (μm/crossly trimmed corneal tissues), numbers of inflammatory cells infiltrated into the cornea (cells/mm2 of cornea stroma), and anterior chambers (cells/mm2 of anterior chambers, between cornea and lens), and numbers of neovasculatures in the cornea (vessels/mm2 of cornea stroma) were measured using a digital image analyzer (DMI-300, DMI, KOREA). The cells, which were stained by each antiserum including caspase- 3, PARP, TNF-#945;, VEGF, COX-2, IL-1#946;, and iNOS were regarded as immunoreactive (45). The percentages of caspase-3 and PARP-immunoreactive cells in cornea epithelium were measured by the digital image analyzer. The numbers of TNF-α, VEGF, COX-2, IL-1β, and iNOS-positive cells per mm2 of anterior chambers were measured, as well as caspase-3 and PARP.

Corneal opacity scoring

A previously described scoring system (36) was used to measure the degree of opacification (between 0-5); Briefly, 0 = clear and compact cornea, 1 = minimal superficial opacity, 2 = mild deep (stromal) opacity with pupil margin and iris vessels visible, 3 = moderate stromal opacity with only pupil margin visible, 4 = intense stromal opacity with anterior chamber visible, 5 = maximal corneal opacity with total obscuration of the anterior chamber.

Neovascularization scoring

Neovascularization was graded between 0 and 3 per corneal quadrant, with increments of 0.5, by a grid system based on the centripetal extent of neovascular branch outgrowth from the corneoscleral limbus (46). Neovascularization was generally graded by the extent of vessel invasion toward the central cornea regions, and was subdivided into 12 grades by the length of invaded vessels. Dilated limbal vessels not penetrating the corneal stroma were not considered to represent corneal neovascularization. All grading was done in a masked fashion. Scores for each quadrant were then summed to derive the cornea neovascularization index (range, 0-12) for each eye.

Statistical analysis

The obtained data were analyzed by one way ANOVA test followed by least-significant differences (LSD) multi comparison test. P < 0.05 was significantly different.

References

  1. Ormerod LD, Abelson MB and Kenyon KR (1989) Standard models of corneal injury using alkali-immersed filter discs. Invest Ophthalmol Vis Sci 30, 2148-2153
  2. Wagoner MD (1997) Chemical injuries of the eye: current concepts in pathophysiology and therapy. Surv Ophthalmol 41, 275-313 https://doi.org/10.1016/S0039-6257(96)00007-0
  3. Liu X, Lin Z, Zhou T et al (2011) Anti-angiogenic and anti-inflammatory effects of SERPINA3K on corneal injury. PLoS One 6, e16712 https://doi.org/10.1371/journal.pone.0016712
  4. Kao WW, Ebert J, Kao CW, Covington H and Cintron C (1986) Development of monoclonal antibodies recognizing collagenase from rabbit PMN; the presence of this enzyme in ulcerating corneas. Curr Eye Res 5, 801-815 https://doi.org/10.3109/02713688609029231
  5. Phan TM, Foster CS, Shaw CD, Zagachin LM and Colvin RB (1991) Topical fibronectin in an alkali burn model of corneal ulceration in rabbits. Arch Ophthalmol 109, 414-419 https://doi.org/10.1001/archopht.1991.01080030116051
  6. Tseng SC (1985) Topical retinoid treatment for dry eye disorders. Trans Ophthalmol Soc UK 104, 489-495
  7. Reim M (1990) A new treatment concept for severe caustic and thermal burns of the eyes. Klin Monbl Augenheilkd 196, 1-5 https://doi.org/10.1055/s-2008-1046118
  8. Thoft RA (1977) Conjunctival transplantation. Arch Ophthalmol 95, 1425-1427 https://doi.org/10.1001/archopht.1977.04450080135017
  9. Sekundo W, Augustin AJ and Strempel I (2002) Topical allopurinol or corticosteroids and acetylcysteine in the early treatment of experimental corneal alkali burns: a pilot study. Eur J Ophthalmol 12, 366-372 https://doi.org/10.1177/112067210201200504
  10. Dan L, Shi-long Y, Miao-li L et al (2008) Inhibitory effect of oral doxycycline on neovascularization in a rat corneal alkali burn model of angiogenesis. Curr Eye Res 33, 653-660 https://doi.org/10.1080/02713680802245772
  11. Guler M, Yilmaz T, Ozercan I and Elkiran T (2009) The inhibitory effects of trastuzumab on corneal neovascularization. Am J Ophthalmol 147, 703-708 https://doi.org/10.1016/j.ajo.2008.09.022
  12. Snyder SH and Sabatini DM (1995) Immunophilins and the nervous system. Nat Med 1, 32-37 https://doi.org/10.1038/nm0195-32
  13. Schiene-Fischer C and Yu C (2001) Receptor accessory folding helper enzymes: the functional role of peptidyl prolyl cis/trans isomerases. FEBS Lett 495, 1-6 https://doi.org/10.1016/S0014-5793(01)02326-2
  14. Dietz GP (2010) Cell-penetrating peptide technology to deliver chaperones and associated factors in diseases and basic research. Curr Pharm Biotechol 11, 167-174 https://doi.org/10.2174/138920110790909731
  15. Liu L, Yu R, Shi Y et al (2014) Transduced protein transduction domain linked HSP27 protected LECs against UVB radiation-induced damage. Exp Eye Res 120, 36-42 https://doi.org/10.1016/j.exer.2013.12.016
  16. Kim SY, Jeong HJ, Kim DW et al (2011) Transduced PEP-1-FK506BP inhibits the inflammatory response in the Raw 264.7 cells and mouse models. Immunobiol 216, 771-781 https://doi.org/10.1016/j.imbio.2010.12.008
  17. Wadia JS and Dowdy SF (2002) Protein transduction technology. Curr Opin Biotechnol 13, 52-56 https://doi.org/10.1016/S0958-1669(02)00284-7
  18. Youn JK, Kim DW, Kim ST et al (2014) PEP-1-HO-1 prevents MPTP-induced degeneration of dopaminergic neurons in a Parkinson’s disease mouse model. BMB Rep 47, 569-574 https://doi.org/10.5483/BMBRep.2014.47.10.286
  19. Bertolini A, Ottani A and Sandrini M (2002) Selective COX-2 inhibitors and dual acting anti-inflammatory drugs; critical remarks. Curr Med Chem 9, 1033-1043 https://doi.org/10.2174/0929867024606650
  20. Firestein GS (2003) Evolving concepts of rheumatoid arthritis. Nature 423, 356-361 https://doi.org/10.1038/nature01661
  21. Uto T, Fujii M and Hou DX (2007) Effects of 6-(methylsulfinyl) hexyl isothio cyanate on cyclooxygenase-2 expression induced by lipopolysaccharide, interferon-gamma and 12- O-tetradecanoylphorbol-13-acetate. Oncol Rep 17, 233-238
  22. Duan Y, Chen F, Zhang et al (2014) Aspirin inhibits lipopolysaccharide-induced COX-2 expression and PGE2 production in porcine alveolar macrophages by modulating protein kinase C and protein tyrosine phosphatase activity. BMB Rep 47, 45-50 https://doi.org/10.5483/BMBRep.2014.47.1.089
  23. Kawamura A, Tatsuguchi A, Ishizaki M, Takahashi H and Fukuda Y (2008) Expression of microsomal prostaglandin e synthase-1 in fibroblasts of rabbit alkali-burned corneas. Cornea 27, 1156-1163 https://doi.org/10.1097/ICO.0b013e318180e53e
  24. Cejkova J, Trosan P, Cejka C et al (2013) Suppression of alkali-induced oxidative injury in the cornea by mesenchymal stem cells growing on nanofiber scaffolds and transferred onto the damaged corneal surface. Exp Eye Res 116, 312-323 https://doi.org/10.1016/j.exer.2013.10.002
  25. Singh N, Jani PD, Suthar T, Amin S and Ambati BK (2006) Flt-1 intraceptor induces the unfolded protein response, apoptotic factors, and regression of murine injury-induced corneal neovascularization. Invest Ophthalmol Vis Sci 47, 4787-4793 https://doi.org/10.1167/iovs.06-0419
  26. Kim DW, Lee SH, Ku SK et al (2013) Transduced PEP-1-FK506BP ameliorates corneal injury in Botulinum toxin A-induced dry eye mouse model. BMB Rep 46, 124-129 https://doi.org/10.5483/BMBRep.2013.46.2.272
  27. Kim SY, Sohn EJ, Kim DW et al (2011) Transduced PEP-1-FK506BP ameliorates atopic dermatitis in NC/Nga mice. J Invest Dermatol 131, 1477-1485 https://doi.org/10.1038/jid.2011.49
  28. Song HY, Lee JA, Ju SM et al (2008) Topical transduction of superoxide dismutase mediated by HIV-1 Tat protein transduction domain ameliorates 12-O-tetradecanoyl phorbol- 13-acetate (TPA)-induced inflammation in mice. Biochem Pharmacol 75, 1348-1357 https://doi.org/10.1016/j.bcp.2007.11.015
  29. He J, Bazan NG and Bazan HE (2006) Alkali-induced corneal stromal melting prevention by a novel platelet-activating factor receptor antagonist. Arch Ophthalmol 124, 70-78 https://doi.org/10.1001/archopht.124.1.70
  30. Ma Y, Xu Y, Xiao Z et al (2006) Reconstruction of chemically burned rat corneal surface by bone marrow-derived human mesenchymal stem cells. Stem Cells 24, 315-321 https://doi.org/10.1634/stemcells.2005-0046
  31. Toshida H, Odaka A, Koike D and Murakami A (2008) Effect of retinol palmitate eye drops on experimental keratoconjunctival epithelial damage induced by n-heptanol in rabbit. Curr Eye Res 33, 13-18 https://doi.org/10.1080/02713680701827696
  32. Lim S and Park S (2014) Role of vascular smooth muscle cell in the inflammation of atherosclerosis. BMB Rep 47, 1-7 https://doi.org/10.5483/BMBRep.2014.47.1.285
  33. Yamada J, Dana MR, Sotozono C and Kinoshita S (2003) Local suppression of IL-1 by receptor antagonist in the rat model of corneal alkali injury. Exp Eye Res 76, 161-167 https://doi.org/10.1016/S0014-4835(02)00293-2
  34. Schrage NF, Kompa S, Haller W and Langefeld S (2002) Use of an amphoteric lavage solution for emergency treatment of eye burns. First animal type experimental clinical considerations. Burns 28, 782-786 https://doi.org/10.1016/S0305-4179(02)00194-8
  35. Epstein RJ, Stulting RD, Hendricks RL and Harris DM (1987) Corneal neovascularization: Pathogenesis and inhibition. Cornea 6, 250-257 https://doi.org/10.1097/00003226-198706040-00004
  36. Sonoda Y and Streilein JW (1992) Orthotopic corneal transplantation in mice--evidence that the immunogenetic rules of rejection do not apply. Transplantation 54, 694-704 https://doi.org/10.1097/00007890-199210000-00026
  37. Chung JH, Fagerholm P and Lindstrom B (1989) Hyaluronate in healing of corneal alkali wound in the rabbit. Exp Eye Res 48, 569-576 https://doi.org/10.1016/0014-4835(89)90039-0
  38. Chung J, Park Y, Paek S, Chong Y and Kim W (1999) Effect of Na-hyaluronan on stromal and endothelial healing in experimental corneal alkali wounds. Ophthalmic Res 31, 432-439 https://doi.org/10.1159/000055568
  39. Bae SR, Park C, Choi JC, Poo H, Kim CJ and Sung MH (2010) Effects of ultra high molecular weight poly-gamma-glutamic acid from Bacillus subtilis (chungkookjang) on corneal wound healing. J Microbiol Biotechnol 20, 803-808
  40. Soltes L, Lath D, Mendichi R and Bystricky P (2001) Radical degradation of high molecular weight hyaluronan: inhibition of the reaction by ibuprofen enantiomers. Methods Find Exp Clin Pharmacol 23, 65-71 https://doi.org/10.1358/mf.2001.23.2.627927
  41. Frankel A, Sohn A, Patel RV and Lebwohl M (2011) Bilateral comparison study of pimecrolimus cream 1% and a ceramide-hyaluronic acid emollient foam in the treatment of patients with atopic dermatitis. J Drugs Dermatol 10, 666-672
  42. Mochimaru H, Usui T, Yaguchi T et al (2008) Suppression of alkali burn-induced corneal neovascularization by dendritic cell vaccination targeting VEGF receptor 2. Invest Ophthalmol Vis Sci 49, 2172-2177 https://doi.org/10.1167/iovs.07-1396
  43. Oh JY, Kim MK, Shin MS, Lee HJ, Lee JH and Wee WR (2009) The anti-inflammatory effect of subconjunctival bevacizumab on chemically burned rat corneas. Curr Eye Res 34, 85-91 https://doi.org/10.1080/02713680802607740
  44. Jo H, Jung SH, Kang J, Yim HB and Kang KD (2014) Sulodexide inhibits retinal neovascularization in a mouse model of oxygen-induced retinopathy. BMB Rep 47, 637-642 https://doi.org/10.5483/BMBRep.2014.47.11.009
  45. Lee SJ, Yoo JD, Choi SY and Kwon OS (2014) The expression and secretion of vimentin in the progression of non-alcoholic steatohepatitis. BMB Rep 47, 457-462 https://doi.org/10.5483/BMBRep.2014.47.8.256
  46. Dana MR and Streilein JW (1996) Loss and restoration of immune privilege in eyes with corneal neovascularization. Invest Ophthalmol Vis Sci 37, 2485-2494

Cited by

  1. Effects of fermented black ginseng on wound healing mediated by angiogenesis through the mitogen-activated protein kinase pathway in human umbilical vein endothelial cells 2018, https://doi.org/10.1016/j.jgr.2017.07.006
  2. Abietic acid isolated from pine resin (Resina Pini) enhances angiogenesis in HUVECs and accelerates cutaneous wound healing in mice vol.203, 2017, https://doi.org/10.1016/j.jep.2017.03.055
  3. Tat-DJ-1 enhances cell survival by inhibition of oxidative stress, NF-κB and MAPK activation in HepG2 cells vol.39, pp.4, 2017, https://doi.org/10.1007/s10529-017-2286-5
  4. Tat-DJ-1 inhibits oxidative stress-mediated RINm5F cell death through suppression of NF-κB and MAPK activation vol.25, pp.11, 2016, https://doi.org/10.1007/s00044-016-1698-4
  5. Comprehensive Modeling of Corneal Alkali Injury in the Rat Eye 2017, https://doi.org/10.1080/02713683.2017.1317817
  6. Beneficial Effects of Deoxyshikonin on Delayed Wound Healing in Diabetic Mice vol.19, pp.11, 2018, https://doi.org/10.3390/ijms19113660
  7. Inhibited corneal neovascularization in rabbits following corneal alkali burn by double-target interference for VEGF and HIF-1α vol.39, pp.1, 2018, https://doi.org/10.1042/BSR20180552