1
|
Suanno G, Genna VG, Maurizi E, Dieh AA, Griffith M, Ferrari G. Cell therapy in the cornea: The emerging role of microenvironment. Prog Retin Eye Res 2024; 102:101275. [PMID: 38797320 DOI: 10.1016/j.preteyeres.2024.101275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 05/22/2024] [Accepted: 05/23/2024] [Indexed: 05/29/2024]
Abstract
The cornea is an ideal testing field for cell therapies. Its highly ordered structure, where specific cell populations are sequestered in different layers, together with its accessibility, has allowed the development of the first stem cell-based therapy approved by the European Medicine Agency. Today, different techniques have been proposed for autologous and allogeneic limbal and non-limbal cell transplantation. Cell replacement has also been attempted in cases of endothelial cell decompensation as it occurs in Fuchs dystrophy: injection of cultivated allogeneic endothelial cells is now in advanced phases of clinical development. Recently, stromal substitutes have been developed with excellent integration capability and transparency. Finally, cell-derived products, such as exosomes obtained from different sources, have been investigated for the treatment of severe corneal diseases with encouraging results. Optimization of the success rate of cell therapies obviously requires high-quality cultured cells/products, but the role of the surrounding microenvironment is equally important to allow engraftment of transplanted cells, to preserve their functions and, ultimately, lead to restoration of tissue integrity and transparency of the cornea.
Collapse
Affiliation(s)
- Giuseppe Suanno
- Vita-Salute San Raffaele University, Milan, Italy; Eye Repair Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Eleonora Maurizi
- Centre for Regenerative Medicine ''S. Ferrari'', University of Modena and Reggio Emilia, Modena, Italy
| | - Anas Abu Dieh
- Maisonneuve-Rosemont Hospital Research Centre, Montreal, Quebec, Canada
| | - May Griffith
- Maisonneuve-Rosemont Hospital Research Centre, Montreal, Quebec, Canada.
| | - Giulio Ferrari
- Vita-Salute San Raffaele University, Milan, Italy; Eye Repair Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy; Ophthalmology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
2
|
Okoyeocha EOM, Tewari-Singh N. Chloropicrin induced ocular injury: Biomarkers, potential mechanisms, and treatments. Toxicol Lett 2024; 396:70-80. [PMID: 38677567 DOI: 10.1016/j.toxlet.2024.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 04/09/2024] [Accepted: 04/17/2024] [Indexed: 04/29/2024]
Abstract
Ocular tissue, especially the cornea, is overly sensitive to chemical exposures. The availability and adoption of chemical threat agent chloropicrin (CP) is growing in the United States as a pesticide and fumigant; thereby increasing the risk of its use in warfare, terrorist attacks and non-intentional exposure. Exposure to CP results in immediate ocular, respiratory, and dermal injury; however, we lack knowledge on its mechanism of toxicity as well as of its breakdown products like chlorine and phosgene, and effective therapies are elusive. Herein, we have reviewed the recent findings on exposure route, toxicity and likely mechanisms of CP induced ocular toxicity based on other vesicating chemical warfare agents that cause ocular injury. We have focused on the implication of their toxicity and mechanistic outcomes in the ocular tissue, especially the cornea, which could be useful in the development of broad-spectrum effective therapeutic options. We have discussed on the potential countermeasures, overall hallmarks and challenges involved in studying ocular injuries from chemical threat agent exposures. Finally, we reviewed useful available technologies and methods that can assist in the identification of effective medical countermeasures for chemical threat agents related ocular injuries.
Collapse
Affiliation(s)
- Ebenezar O M Okoyeocha
- Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, 48824, USA
| | - Neera Tewari-Singh
- Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, 48824, USA.
| |
Collapse
|
3
|
Blohm KO, Nell B. Adjunctive bevacizumab therapy in an equine corneal stromal invasive squamous cell carcinoma with a 53-months follow-up. Tierarztl Prax Ausg G Grosstiere Nutztiere 2024; 52:108-114. [PMID: 38701802 PMCID: PMC11068419 DOI: 10.1055/a-2253-8103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 11/28/2023] [Indexed: 05/05/2024]
Abstract
A 17-year-old Appaloosa mare was referred for evaluation of presumed refractory keratitis of the left eye. Gross examination revealed ocular discomfort and corneal neovascularization with a nasal focal opacification affecting approximately 40% of the corneal surface. On ophthalmic examination, extensive subepithelial to mid-stromal vascular branching accompanied by a homogeneous white, dense opacification, which affected up to 80% of the total corneal thickness, were apparent. Signs of concurrent uveitis were absent. Deep-stromal lamellar keratectomy with a conjunctival pedicle graft was performed under general anesthesia. Histopathology confirmed a poorly differentiated corneal stromal invasive squamous cell carcinoma (SI-SCC) with neoplastic cell extension to the surgical margins. Postoperatively, 4 topical mitomycin C 0.04% chemotherapy cycles combined with oral firocoxib therapy were initiated. Seven months after surgery, regrowth of the SI-SCC was clinically suspected. A total volume of 1 ml bevacizumab 2.5% was administered in the standing sedated horse via 3 mid-stromal corneal injections. Four weeks later, intrastromal bevacizumab injections (ISBIs) were repeated, however, this time the solution was injected directly into the main corneal vessel branches.Seven weeks after the second ISBIs, the left eye was comfortable and significant remission of corneal vascularization and opacity was recognized. No recurrence has been noted for a follow-up period of more than 53 months.Equine SI-SCC usually has a very poor prognosis for globe maintenance. To the authors' knowledge this is the first report of well-tolerated intrastromal antivascular endothelial growth factor adjunctive therapy with bevazicumab 2.5% and SI-SCC resolution after a multimodal treatment approach.
Collapse
Affiliation(s)
- Klaas-Ole Blohm
- Tierklinik Lüsche im SANAKENA, Appen-Etz, Germany
- Tierarztpraxis Schönhoff, Stendal, Germany
- Department for Companion Animals and Horses, Ophthalmology Unit, University of Veterinary Medicine, Vienna, Austria
| | - Barbara Nell
- Department for Companion Animals and Horses, Ophthalmology Unit, University of Veterinary Medicine, Vienna, Austria
| |
Collapse
|
4
|
Atalay E, Altuğ B, Çalışkan ME, Ceylan S, Özler ZS, Figueiredo G, Lako M, Figueiredo F. Animal Models for Limbal Stem Cell Deficiency: A Critical Narrative Literature Review. Ophthalmol Ther 2024; 13:671-696. [PMID: 38280103 PMCID: PMC10853161 DOI: 10.1007/s40123-023-00880-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 12/19/2023] [Indexed: 01/29/2024] Open
Abstract
This literature review will provide a critical narrative overview of the highlights and potential pitfalls of the reported animal models for limbal stem cell deficiency (LSCD) and will identify the neglected aspects of this research area. There exists significant heterogeneity in the literature regarding the methodology used to create the model and the predefined duration after the insult when the model is supposedly fully fit for evaluations and/or for testing various therapeutic interventions. The literature is also replete with examples wherein the implementation of a specific model varies significantly across different studies. For example, the concentration of the chemical, as well as its duration and technique of exposure in a chemically induced LSCD model, has a great impact not only on the validity of the model but also on the severity of the complications. Furthermore, while some models induce a full-blown clinical picture of total LSCD, some are hindered by their ability to yield only partial LSCD. Another aspect to consider is the nature of the damage induced by a specific method. As thermal methods cause more stromal scarring, they may be better suited for assessing the anti-fibrotic properties of a particular treatment. On the other hand, since chemical burns cause more neovascularisation, they provide the opportunity to tap into the potential treatments for anti-neovascularisation. The animal species (i.e., rats, mice, rabbits, etc.) is also a crucial factor in the validity of the model and its potential for clinical translation, with each animal having its unique set of advantages and disadvantages. This review will also elaborate on other overlooked aspects, such as the anaesthetic(s) used during experiments, the gender of the animals, care after LSCD induction, and model validation. The review will conclude by providing future perspectives and suggestions for further developments in this rather important area of research.
Collapse
Affiliation(s)
- Eray Atalay
- Department of Ophthalmology, Eskişehir Osmangazi University Medical School, Eskişehir, Turkey
| | - Burcugül Altuğ
- Cellular Therapy and Stem Cell Production Application, Research Centre (ESTEM), Eskişehir Osmangazi University, Eskişehir, Turkey
| | | | - Semih Ceylan
- Eskişehir Osmangazi University Medical School, Eskişehir, Turkey
| | | | | | - Majlinda Lako
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Francisco Figueiredo
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK.
- Department of Ophthalmology, Royal Victoria Infirmary, Newcastle University, Newcastle upon Tyne, NE1 4LP, UK.
| |
Collapse
|
5
|
Mishra N, Kant R, Kandhari K, Ammar DA, Tewari-Singh N, Pantcheva MB, Petrash JM, Agarwal C, Agarwal R. Nitrogen Mustard-Induced Ex Vivo Human Cornea Injury Model and Therapeutic Intervention by Dexamethasone. J Pharmacol Exp Ther 2024; 388:484-494. [PMID: 37474260 PMCID: PMC10801761 DOI: 10.1124/jpet.123.001760] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/26/2023] [Accepted: 06/30/2023] [Indexed: 07/22/2023] Open
Abstract
Sulfur mustard (SM), a vesicating agent first used during World War I, remains a potent threat as a chemical weapon to cause intentional/accidental chemical emergencies. Eyes are extremely susceptible to SM toxicity. Nitrogen mustard (NM), a bifunctional alkylating agent and potent analog of SM, is used in laboratories to study mustard vesicant-induced ocular toxicity. Previously, we showed that SM-/NM-induced injuries (in vivo and ex vivo rabbit corneas) are reversed upon treatment with dexamethasone (DEX), a US Food and Drug Administration-approved, steroidal anti-inflammatory drug. Here, we optimized NM injuries in ex vivo human corneas and assessed DEX efficacy. For injury optimization, one cornea (randomly selected from paired eyes) was exposed to NM: 100 nmoles for 2 hours or 4 hours, and 200 nmoles for 2 hours, and the other cornea served as a control. Injuries were assessed 24 hours post NM-exposure. NM 100 nmoles exposure for 2 hours was found to cause optimal corneal injury (epithelial thinning [∼69%]; epithelial-stromal separation [6-fold increase]). In protein arrays studies, 24 proteins displayed ≥40% change in their expression in NM exposed corneas compared with controls. DEX administration initiated 2 hours post NM exposure and every 8 hours thereafter until 24 hours post-exposure reversed NM-induced corneal epithelial-stromal separation [2-fold decrease]). Of the 24 proteins dysregulated upon NM exposure, six proteins (delta-like canonical Notch ligand 1, FGFbasic, CD54, CCL7, endostatin, receptor tyrosine-protein kinase erbB-4) associated with angiogenesis, immune/inflammatory responses, and cell differentiation/proliferation, showed significant reversal upon DEX treatment (Student's t test; P ≤ 0.05). Complementing our animal model studies, DEX was shown to mitigate vesicant-induced toxicities in ex vivo human corneas. SIGNIFICANCE STATEMENT: Nitrogen mustard (NM) exposure-induced injuries were optimized in an ex vivo human cornea culture model and studies were carried out at 24 h post 100 nmoles NM exposure. Dexamethasone (DEX) administration (started 2 h post NM exposure and every 8 h thereafter) reversed NM-induced corneal injuries. Molecular mediators of DEX action were associated with angiogenesis, immune/inflammatory responses, and cell differentiation/proliferation, indicating DEX aids wound healing via reversing vesicant-induced neovascularization (delta-like canonical Notch ligand 1 and FGF basic) and leukocyte infiltration (CD54 and CCL7).
Collapse
Affiliation(s)
- Neha Mishra
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences (N.M., R.K., K.K., C.A., R.A.) and Department of Ophthalmology, School of Medicine (M.B.P., J.M.P.) University of Colorado-Anschutz Medical Campus, Aurora, Colorado; Lions Eye Institute for Transplant and Research, Tampa, Florida (D.A.A.); and Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, Michigan (N.T.-S.)
| | - Rama Kant
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences (N.M., R.K., K.K., C.A., R.A.) and Department of Ophthalmology, School of Medicine (M.B.P., J.M.P.) University of Colorado-Anschutz Medical Campus, Aurora, Colorado; Lions Eye Institute for Transplant and Research, Tampa, Florida (D.A.A.); and Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, Michigan (N.T.-S.)
| | - Kushal Kandhari
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences (N.M., R.K., K.K., C.A., R.A.) and Department of Ophthalmology, School of Medicine (M.B.P., J.M.P.) University of Colorado-Anschutz Medical Campus, Aurora, Colorado; Lions Eye Institute for Transplant and Research, Tampa, Florida (D.A.A.); and Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, Michigan (N.T.-S.)
| | - David A Ammar
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences (N.M., R.K., K.K., C.A., R.A.) and Department of Ophthalmology, School of Medicine (M.B.P., J.M.P.) University of Colorado-Anschutz Medical Campus, Aurora, Colorado; Lions Eye Institute for Transplant and Research, Tampa, Florida (D.A.A.); and Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, Michigan (N.T.-S.)
| | - Neera Tewari-Singh
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences (N.M., R.K., K.K., C.A., R.A.) and Department of Ophthalmology, School of Medicine (M.B.P., J.M.P.) University of Colorado-Anschutz Medical Campus, Aurora, Colorado; Lions Eye Institute for Transplant and Research, Tampa, Florida (D.A.A.); and Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, Michigan (N.T.-S.)
| | - Mina B Pantcheva
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences (N.M., R.K., K.K., C.A., R.A.) and Department of Ophthalmology, School of Medicine (M.B.P., J.M.P.) University of Colorado-Anschutz Medical Campus, Aurora, Colorado; Lions Eye Institute for Transplant and Research, Tampa, Florida (D.A.A.); and Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, Michigan (N.T.-S.)
| | - J Mark Petrash
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences (N.M., R.K., K.K., C.A., R.A.) and Department of Ophthalmology, School of Medicine (M.B.P., J.M.P.) University of Colorado-Anschutz Medical Campus, Aurora, Colorado; Lions Eye Institute for Transplant and Research, Tampa, Florida (D.A.A.); and Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, Michigan (N.T.-S.)
| | - Chapla Agarwal
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences (N.M., R.K., K.K., C.A., R.A.) and Department of Ophthalmology, School of Medicine (M.B.P., J.M.P.) University of Colorado-Anschutz Medical Campus, Aurora, Colorado; Lions Eye Institute for Transplant and Research, Tampa, Florida (D.A.A.); and Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, Michigan (N.T.-S.)
| | - Rajesh Agarwal
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences (N.M., R.K., K.K., C.A., R.A.) and Department of Ophthalmology, School of Medicine (M.B.P., J.M.P.) University of Colorado-Anschutz Medical Campus, Aurora, Colorado; Lions Eye Institute for Transplant and Research, Tampa, Florida (D.A.A.); and Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, Michigan (N.T.-S.)
| |
Collapse
|
6
|
Zhylkibayev A, Ung TT, Mobley J, Athar M, Gorbatyuk M. The Involvement of Unfolded Protein Response in the Mechanism of Nitrogen Mustard-Induced Ocular Toxicity. J Pharmacol Exp Ther 2024; 388:518-525. [PMID: 37914413 PMCID: PMC10801749 DOI: 10.1124/jpet.123.001814] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 09/25/2023] [Accepted: 09/26/2023] [Indexed: 11/03/2023] Open
Abstract
Nitrogen mustard (NM) is a known surrogate of sulfur mustard, a chemical-warfare agent that causes a wide range of ocular symptoms, from a permanent reduction in visual acuity to blindness upon exposure. Although it has been proposed that the two blistering agents have a similar mechanism of toxicity, the mode of NM-induced cell death in ocular tissue has not been fully explored. Therefore, we hypothesized that direct ocular exposure to NM in mice leads to retinal tissue injury through chronic activation of the unfolded protein response (UPR) PERK arm in corneal cells and VEGF secretion, eventually causing cell death. We topically applied NM directly to mice to analyze ocular and retinal tissues at 2 weeks postexposure. A dramatic decline in retinal function, measured by scotopic and photopic electroretinogram responses, was detected in the mice. This decline was associated with enhanced TUNEL staining in both corneal and retinal tissues. In addition, exposure of corneal cells to NM revealed 228 differentially and exclusively expressed proteins primarily associated with the UPR, ferroptosis, and necroptosis. Moreover, these cells exhibited activation of the UPR PERK arm and an increase in VEGF secretion. Enhancement of VEGF staining was later observed in the corneas of the exposed mice. Therefore, our data indicated that the mechanism of NM-induced ocular toxicity should be carefully examined and that future research should identify a signaling molecule transmitted via a prodeath pathway from the cornea to the retina. SIGNIFICANCE STATEMENT: This study demonstrated that NM topical exposure in mice results in dramatic decline in retinal function associated with enhanced TUNEL staining in both corneal and retinal tissues. We also found that the NM treatment of corneal cells resulted in 228 differentially and exclusively expressed proteins primarily associated with ferroptosis. Moreover, these cells manifest the UPR PERK activation and an increase in VEGF secretion. The latter was also found in the corneas of the cexposed mice.
Collapse
Affiliation(s)
- Assylbek Zhylkibayev
- School of Optometry, Department of Optometry and Vision Science (A.Z., T.T.U., M.G.), School of Medicine, Departments of Anesthesiology and Perioperative Medicine (J.M.), and Department of Dermatology (M.A.), University of Alabama at Birmingham, Birmingham, Alabama
| | - Trong Thuan Ung
- School of Optometry, Department of Optometry and Vision Science (A.Z., T.T.U., M.G.), School of Medicine, Departments of Anesthesiology and Perioperative Medicine (J.M.), and Department of Dermatology (M.A.), University of Alabama at Birmingham, Birmingham, Alabama
| | - James Mobley
- School of Optometry, Department of Optometry and Vision Science (A.Z., T.T.U., M.G.), School of Medicine, Departments of Anesthesiology and Perioperative Medicine (J.M.), and Department of Dermatology (M.A.), University of Alabama at Birmingham, Birmingham, Alabama
| | - Mohammad Athar
- School of Optometry, Department of Optometry and Vision Science (A.Z., T.T.U., M.G.), School of Medicine, Departments of Anesthesiology and Perioperative Medicine (J.M.), and Department of Dermatology (M.A.), University of Alabama at Birmingham, Birmingham, Alabama
| | - Marina Gorbatyuk
- School of Optometry, Department of Optometry and Vision Science (A.Z., T.T.U., M.G.), School of Medicine, Departments of Anesthesiology and Perioperative Medicine (J.M.), and Department of Dermatology (M.A.), University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
7
|
Mishra N, Kant R, Kandhari K, Tewari-Singh N, Anantharam P, Croutch CR, Pantcheva MB, Petrash JM, Araj H, Agarwal C, Agarwal R. Establishing a Dexamethasone Treatment Regimen To Alleviate Sulfur Mustard-Induced Corneal Injuries in a Rabbit Model. J Pharmacol Exp Ther 2024; 388:469-483. [PMID: 37316330 PMCID: PMC10801779 DOI: 10.1124/jpet.123.001680] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/10/2023] [Accepted: 05/18/2023] [Indexed: 06/16/2023] Open
Abstract
Sulfur mustard (SM) is an ominous chemical warfare agent. Eyes are extremely susceptible to SM toxicity; injuries include inflammation, fibrosis, neovascularization (NV), and vision impairment/blindness, depending on the exposure dosage. Effective countermeasures against ocular SM toxicity remain elusive and are warranted during conflicts/terrorist activities and accidental exposures. We previously determined that dexamethasone (DEX) effectively counters corneal nitrogen mustard toxicity and that the 2-hour postexposure therapeutic window is most beneficial. Here, the efficacy of two DEX dosing frequencies [i.e., every 8 or 12 hours (initiated, as previously established, 2 hours after exposure)] until 28 days after SM exposure was assessed. Furthermore, sustained effects of DEX treatments were observed up to day 56 after SM exposure. Corneal clinical assessments (thickness, opacity, ulceration, and NV) were performed at the day 14, 28, 42, and 56 post-SM exposure time points. Histopathological assessments of corneal injuries (corneal thickness, epithelial degradation, epithelial-stromal separation, inflammatory cell, and blood vessel counts) using H&E staining and molecular assessments (COX-2, MMP-9, VEGF, and SPARC expressions) were performed at days 28, 42, and 56 after SM exposure. Statistical significance was assessed using two-way ANOVA, with Holm-Sidak post hoc pairwise multiple comparisons; significance was established if P < 0.05 (data represented as the mean ± S.E.M.). DEX administration every 8 hours was more potent than every 12 hours in reversing ocular SM injury, with the most pronounced effects observed at days 28 and 42 after SM exposure. These comprehensive results are novel and provide a comprehensive DEX treatment regimen (therapeutic-window and dosing-frequency) for counteracting SM-induced corneal injuries. SIGNIFICANCE STATEMENT: The study aims to establish a dexamethasone (DEX) treatment regimen by comparing the efficacy of DEX administration at 12 versus 8 hours initiated 2 hours after exposure. DEX administration every 8 hours was more effective in reversing sulfur mustard (SM)-induced corneal injuries. SM injury reversal during DEX administration (initial 28 days after exposure) and sustained [further 28 days after cessation of DEX administration (i.e., up to 56 days after exposure)] effects were assessed using clinical, pathophysiological, and molecular biomarkers.
Collapse
Affiliation(s)
- Neha Mishra
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences (N.M., R.K., K.K., N.T.-S., C.A., R.A.) and Department of Ophthalmology, School of Medicine (M.B.P., J.M.P.), University of Colorado-Anschutz Medical Campus, Aurora, Colorado; MRIGlobal, Kansas City, Missouri (P.A., C.R.C.); and Department of Health and Human Services, National Institutes of Health National Eye Institute, Bethesda, Maryland (H.A.)
| | - Rama Kant
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences (N.M., R.K., K.K., N.T.-S., C.A., R.A.) and Department of Ophthalmology, School of Medicine (M.B.P., J.M.P.), University of Colorado-Anschutz Medical Campus, Aurora, Colorado; MRIGlobal, Kansas City, Missouri (P.A., C.R.C.); and Department of Health and Human Services, National Institutes of Health National Eye Institute, Bethesda, Maryland (H.A.)
| | - Kushal Kandhari
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences (N.M., R.K., K.K., N.T.-S., C.A., R.A.) and Department of Ophthalmology, School of Medicine (M.B.P., J.M.P.), University of Colorado-Anschutz Medical Campus, Aurora, Colorado; MRIGlobal, Kansas City, Missouri (P.A., C.R.C.); and Department of Health and Human Services, National Institutes of Health National Eye Institute, Bethesda, Maryland (H.A.)
| | - Neera Tewari-Singh
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences (N.M., R.K., K.K., N.T.-S., C.A., R.A.) and Department of Ophthalmology, School of Medicine (M.B.P., J.M.P.), University of Colorado-Anschutz Medical Campus, Aurora, Colorado; MRIGlobal, Kansas City, Missouri (P.A., C.R.C.); and Department of Health and Human Services, National Institutes of Health National Eye Institute, Bethesda, Maryland (H.A.)
| | - Poojya Anantharam
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences (N.M., R.K., K.K., N.T.-S., C.A., R.A.) and Department of Ophthalmology, School of Medicine (M.B.P., J.M.P.), University of Colorado-Anschutz Medical Campus, Aurora, Colorado; MRIGlobal, Kansas City, Missouri (P.A., C.R.C.); and Department of Health and Human Services, National Institutes of Health National Eye Institute, Bethesda, Maryland (H.A.)
| | - Claire R Croutch
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences (N.M., R.K., K.K., N.T.-S., C.A., R.A.) and Department of Ophthalmology, School of Medicine (M.B.P., J.M.P.), University of Colorado-Anschutz Medical Campus, Aurora, Colorado; MRIGlobal, Kansas City, Missouri (P.A., C.R.C.); and Department of Health and Human Services, National Institutes of Health National Eye Institute, Bethesda, Maryland (H.A.)
| | - Mina B Pantcheva
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences (N.M., R.K., K.K., N.T.-S., C.A., R.A.) and Department of Ophthalmology, School of Medicine (M.B.P., J.M.P.), University of Colorado-Anschutz Medical Campus, Aurora, Colorado; MRIGlobal, Kansas City, Missouri (P.A., C.R.C.); and Department of Health and Human Services, National Institutes of Health National Eye Institute, Bethesda, Maryland (H.A.)
| | - J Mark Petrash
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences (N.M., R.K., K.K., N.T.-S., C.A., R.A.) and Department of Ophthalmology, School of Medicine (M.B.P., J.M.P.), University of Colorado-Anschutz Medical Campus, Aurora, Colorado; MRIGlobal, Kansas City, Missouri (P.A., C.R.C.); and Department of Health and Human Services, National Institutes of Health National Eye Institute, Bethesda, Maryland (H.A.)
| | - Houmam Araj
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences (N.M., R.K., K.K., N.T.-S., C.A., R.A.) and Department of Ophthalmology, School of Medicine (M.B.P., J.M.P.), University of Colorado-Anschutz Medical Campus, Aurora, Colorado; MRIGlobal, Kansas City, Missouri (P.A., C.R.C.); and Department of Health and Human Services, National Institutes of Health National Eye Institute, Bethesda, Maryland (H.A.)
| | - Chapla Agarwal
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences (N.M., R.K., K.K., N.T.-S., C.A., R.A.) and Department of Ophthalmology, School of Medicine (M.B.P., J.M.P.), University of Colorado-Anschutz Medical Campus, Aurora, Colorado; MRIGlobal, Kansas City, Missouri (P.A., C.R.C.); and Department of Health and Human Services, National Institutes of Health National Eye Institute, Bethesda, Maryland (H.A.)
| | - Rajesh Agarwal
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences (N.M., R.K., K.K., N.T.-S., C.A., R.A.) and Department of Ophthalmology, School of Medicine (M.B.P., J.M.P.), University of Colorado-Anschutz Medical Campus, Aurora, Colorado; MRIGlobal, Kansas City, Missouri (P.A., C.R.C.); and Department of Health and Human Services, National Institutes of Health National Eye Institute, Bethesda, Maryland (H.A.)
| |
Collapse
|
8
|
Horwitz V, Cohen M, Gore A, Gez R, Gutman H, Kadar T, Dachir S, Kendler S. Predicting clinical outcome of sulfur mustard induced ocular injury using machine learning model. Exp Eye Res 2023; 236:109671. [PMID: 37776992 DOI: 10.1016/j.exer.2023.109671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 09/23/2023] [Accepted: 09/27/2023] [Indexed: 10/02/2023]
Abstract
The sight-threatening sulfur mustard (SM) induced ocular injury presents specific symptoms in each clinical stage. The acute injury develops in all exposed eyes and may heal or deteriorate into chronic late pathology. Early detection of eyes at risk of developing late pathology may assist in providing unique monitoring and specific treatments only to relevant cases. In this study, we evaluated a machine-learning (ML) model for predicting the development of SM-induced late pathology based on clinical data of the acute phase in the rabbit model. Clinical data from 166 rabbit eyes exposed to SM vapor was used retrospectively. The data included a comprehensive clinical evaluation of the cornea, eyelids and conjunctiva using a semi-quantitative clinical score. A random forest classifier ML model, was trained to predict the development of corneal neovascularization four weeks post-ocular exposure to SM vapor using clinical scores recorded three weeks earlier. The overall accuracy in predicting the clinical outcome of SM-induced ocular injury was 73%. The accuracy in identifying eyes at risk of developing corneal neovascularization and future healed eyes was 75% and 59%, respectively. The most important parameters for accurate prediction were conjunctival secretion and corneal opacity at 1w and corneal erosions at 72 h post-exposure. Predicting the clinical outcome of SM-induced ocular injury based on the acute injury parameters using ML is demonstrated for the first time. Although the prediction accuracy was limited, probably due to the small dataset, it pointed out towards various parameters during the acute injury that are important for predicting SM-induced late pathology and revealing possible pathological mechanisms.
Collapse
Affiliation(s)
- Vered Horwitz
- Department of Pharmacology, Israel Institute for Biological Research, Ness Ziona, 74100, Israel.
| | - Maayan Cohen
- Department of Pharmacology, Israel Institute for Biological Research, Ness Ziona, 74100, Israel
| | - Ariel Gore
- Department of Pharmacology, Israel Institute for Biological Research, Ness Ziona, 74100, Israel
| | - Rellie Gez
- Department of Pharmacology, Israel Institute for Biological Research, Ness Ziona, 74100, Israel
| | - Hila Gutman
- Department of Pharmacology, Israel Institute for Biological Research, Ness Ziona, 74100, Israel
| | - Tamar Kadar
- Department of Pharmacology, Israel Institute for Biological Research, Ness Ziona, 74100, Israel
| | - Shlomit Dachir
- Department of Pharmacology, Israel Institute for Biological Research, Ness Ziona, 74100, Israel
| | - Shai Kendler
- Department of Environmental Physics, Israel Institute for Biological Research, Ness Ziona, 74100, Israel; Faculty of Civil & Environmental Engineering, Technion-Israeli Institute of Technology, Haifa, 320000, Israel
| |
Collapse
|
9
|
Moshirfar M, Masud M, Harvey DH, Payne C, Bruce E, Ronquillo YC, Hoopes PC. The Multifold Etiologies of Limbal Stem Cell Deficiency: A Comprehensive Review on the Etiologies and Additional Treatment Options for Limbal Stem Cell Deficiency. J Clin Med 2023; 12:4418. [PMID: 37445454 DOI: 10.3390/jcm12134418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 06/16/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
Given the various ocular manifestations of limbal stem cell insufficiency, an awareness of the genetic, acquired, and immunological causes and associated additional treatments of limbal stem cell deficiency (LSCD) is essential for providers. We performed a comprehensive review of the literature on the various etiologies and specific therapies for LSCD. The resources utilized in this review included Medline (PubMed), Embase, and Google Scholar. All English-language articles and case reports published from November 1986 through to October 2022 were reviewed in this study. There were collectively 99 articles on these topics. No other exclusion criteria were applied. Depending on the etiology, ocular manifestations of limbal stem cell deficiency range from dry eye syndrome and redness to more severe outcomes, including corneal ulceration, ocular surface failure, and vision loss. Identifying the source of damage for LSCD is critical in the treatment process, given that therapy may extend beyond the scope of the standard protocol, including artificial tears, refractive surgery, and allogeneic stem cell transplants. This comprehensive review of the literature demonstrates the various genetic, acquired, and immunological causes of LSCD and the spectrum of supplemental therapies available.
Collapse
Affiliation(s)
- Majid Moshirfar
- Hoopes Vision Research Center, Hoopes Vision, Draper, UT 84020, USA
- John A. Moran Eye Center, School of Medicine, University of Utah, Salt Lake City, UT 84132, USA
- Utah Lions Eye Bank, Murray, UT 84107, USA
| | - Maliha Masud
- School of Medicine, University of Nevada Reno, Reno, NV 89557, USA
| | - Devon Hori Harvey
- College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Carter Payne
- Hoopes Vision Research Center, Hoopes Vision, Draper, UT 84020, USA
| | - Elayna Bruce
- McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | | | - Philip C Hoopes
- Hoopes Vision Research Center, Hoopes Vision, Draper, UT 84020, USA
| |
Collapse
|
10
|
Soleimani M, Momenaei B, Baradaran-Rafii A, Cheraqpour K, An S, Ashraf MJ, Abedi F, Javadi MA, Djalilian AR. Mustard Gas-Induced Ocular Surface Disorders: An Update on the Pathogenesis, Clinical Manifestations, and Management. Cornea 2023; 42:776-786. [PMID: 36729713 PMCID: PMC10164045 DOI: 10.1097/ico.0000000000003182] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 09/12/2022] [Indexed: 02/03/2023]
Abstract
PURPOSE Mustard gas (MG) is a potent blistering and alkylating agent that has been used for military and terrorism purposes. Ocular surface injuries are common after exposure to MG. This review provides an update on the pathophysiology, ocular surface complications, and treatment options for MG-related ocular injuries. METHODS Required information was obtained by reviewing various databases such as Cochrane Library, Google Scholar, and PubMed until March 2022. Data were collected by using keywords: "mustard gas" OR "sulfur mustard" AND "eye" OR "cornea" OR "ocular complication" OR "keratitis" OR "keratopathy" OR "limbal stem cell deficiency" OR "dry eye." RESULTS Chronic intracellular toxicity, inflammation, and ischemia have been shown to play an essential role in the pathogenesis of MG injury. Ocular surface injuries can have acute, chronic, and most distinctly a delayed-onset presentation leading to various degrees of limbal stem cell deficiency. To date, no treatment has been agreed on as the standard treatment for chronic/delayed-onset MG keratopathy. Based on the authors' experience, we propose a management algorithm for MG-related ocular surface injuries involving optimization of ocular health, anti-inflammatory therapy, and if needed surgical interventions. The management of chronic and delayed-onset presentation remains challenging. CONCLUSIONS MG keratopathy is a unique form of chemical injury which can lead to a range of ocular surface pathologies. Long-term anti-inflammatory therapy even in patients with seemingly mild disease may potentially reduce the likelihood of the development of more severe delayed-onset disease.
Collapse
Affiliation(s)
- Mohammad Soleimani
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, Illinois, USA
- Eye Research Center, Farabi Eye Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Bita Momenaei
- Eye Research Center, Farabi Eye Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Baradaran-Rafii
- Ophthalmic Research Center, Research Institute for Ophthalmology and Vision Science, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of ophthalmology, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Kasra Cheraqpour
- Eye Research Center, Farabi Eye Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Seungwon An
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Mohammad Javad Ashraf
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Farshad Abedi
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Mohammad Ali Javadi
- Ophthalmic Research Center, Research Institute for Ophthalmology and Vision Science, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali R. Djalilian
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, Illinois, USA
| |
Collapse
|
11
|
Ebenezar OO, Roney A, Goswami DG, Petrash JM, Sledge D, Komáromy AM, Liby KT, Tewari-Singh N. Ocular injury progression and cornea histopathology from chloropicrin vapor exposure: Relevant clinical biomarkers in mice. Exp Eye Res 2023; 230:109440. [PMID: 36933694 DOI: 10.1016/j.exer.2023.109440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 03/13/2023] [Accepted: 03/16/2023] [Indexed: 03/18/2023]
Abstract
Ocular tissue is highly sensitive to chemical exposures. Chloropicrin (CP), a choking agent employed during World War I and currently a popular pesticide and fumigating agent, is a potential chemical threat agent. Accidental, occupational, or intentional exposure to CP results in severe ocular injury, especially to the cornea; however, studies on ocular injury progression and underlying mechanisms in a relevant in vivo animal model are lacking. This has impaired the development of effective therapies to treat the acute and long-term ocular toxicity of CP. To study the in vivo clinical and biological effects of CP ocular exposure, we tested different CP exposure doses and durations in mice. These exposures will aid in the study of acute ocular injury and its progression as well as identify a moderate dose to develop a relevant rodent ocular injury model with CP. The left eyes of male BALB/c mice were exposed to CP (20% CP for 0.5 or 1 min or 10% CP for 1 min) using a vapor cap, with the right eyes serving as controls. Injury progression was evaluated for 25 days post-exposure. CP-exposure caused a significant corneal ulceration and eyelid swelling which resolved by day 14 post exposure. In addition, CP-exposure caused significant corneal opacity and neovascularization. Development of hydrops (severe corneal edema with corneal bullae) and hyphema (blood accumulation in the anterior chamber) was observed as advanced CP effects. Mice were euthanized at day 25 post-CP-exposure, and the eyes were harvested to further study the corneal injury. Histopathological analyses showed a significant CP-induced decrease in corneal epithelial thickness and increased stromal thickness with more pronounced damage, including stromal fibrosis, edema, neovascularization, trapped epithelial cells, anterior and posterior synechiae, and infiltration of inflammatory cells. Loss of the corneal endothelial cells and Descemet's membrane could be associated with the CP-induced corneal edema and hydrops which could lead to long term term pathological conditions. Although exposure to 20% CP for 1 min caused more eyelid swelling, ulceration, and hyphema, similar effects were observed with all CP exposures. These novel findings following CP ocular exposure in a mouse model outline the corneal histopathologic changes that associate with the continuing ocular clinical effects. The data are useful in designing further studies to identify and correlate the clinical and biological markers of CP ocular injury progression with acute and long-term toxic effects on cornea and other ocular tissues. We take a crucial step towards CP ocular injury model development and in pathophysiological studies to identify molecular targets for therapeutic interventions.
Collapse
Affiliation(s)
- Okoyeocha Om Ebenezar
- Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, 48824, USA
| | - Andrew Roney
- Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, 48824, USA
| | - Dinesh G Goswami
- Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, 48824, USA
| | - J Mark Petrash
- Department of Ophthalmology, School of Medicine, University of Colorado-Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Dodd Sledge
- Michigan State University Veterinary Diagnostic Laboratory, East Lansing, MI, 48824, USA
| | - András M Komáromy
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, MI, 48824, USA
| | - Karen T Liby
- Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, 48824, USA
| | - Neera Tewari-Singh
- Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, 48824, USA.
| |
Collapse
|
12
|
Gore A, Kadar T, Cohen M, Gutman H, Gez R, Dachir S, Horwitz V. The use of aflibercept (VEGF trap) in mitigating sulfur mustard-induced corneal neovascularization in a rabbit model. Toxicol Rep 2023; 10:206-215. [PMID: 36825253 PMCID: PMC9941054 DOI: 10.1016/j.toxrep.2023.01.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 01/24/2023] [Accepted: 01/25/2023] [Indexed: 01/29/2023] Open
Abstract
Sulfur mustard (SM)-induced ocular injury is characterized by an acute inflammatory response that may become chronic or enter a latent phase with delayed pathology. This study aimed to evaluate the efficacy of ziv-aflibercept and aflibercept in preventing and ameliorating corneal neovascularization (NV), respectively, following chemical eye exposure to SM vapor in a rabbit model. Chemical SM ocular insult was induced in the right eye of rabbits. A single application of ziv-aflibercept was administered 2 h or 9 days post-exposure. A single subconjunctival aflibercept treatment in an ocular formulation was administered 4 weeks after SM vapor exposure and subsequent to an initial 1-week treatment with 0.1 % dexamethasone. Clinical monitoring was performed 5-12 weeks post-exposure, and digital corneal pictures were taken to assess the extent of NV. The rabbits were euthanized and the corneas were processed for histological assessment. Treatment with ziv-aflibercept 2 h and 9 days post-exposure moderately reduced insult severity and partially delayed or prevented corneal NV. Aflibercept application 4 weeks post-exposure significantly reduced the extent of NV for 8 weeks. The substantial decrease in existing corneal NV in this group was confirmed by histology. These results reveal the powerful anti-angiogenic efficacy of the VEGF-trap for ameliorating existing NV as opposed to preventing NV development, revealing the ability of this treatment to mitigate corneal NV.
Collapse
|
13
|
Zhylkibayev A, Srivastava R, Anantharam P, Crotch C, Athar M, Gorbatyuk M. Retinal injury mouse model and pathophysiological assessment of the effect of arsenical vesicants. Exp Eye Res 2023; 226:109354. [PMID: 36539053 PMCID: PMC10448564 DOI: 10.1016/j.exer.2022.109354] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 12/01/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022]
Abstract
The eye is ten times more vulnerable to chemical warfare agents than other organs. Consistently, exposure to vesicant arsenical lewisite (LEW) manifests significant corneal damage leading to chronic inflammation, corneal opacity, vascularization, and edema, culminating in corneal cell death. However, despite the progress has made in the research field investigating arsenical-induced pathogenesis of the anterior chamber of the eye, the retinal damage resulted from exposure to arsenicals has not been identified yet. Therefore, we investigated the effects of direct ocular exposure (DOE) to LEW and phenylarsine oxide (PAO) on the retina. DOE to arsenicals was conducted using the vapor cap method at the MRIGlobal facility or an eye patch soaked in solutions with different PAO concentrations at UAB. Animals were assessed at 1, 3, 14, and 28 days postexposure. Results of the study demonstrated that both arsenicals cause severe retinal damage, activating proinflammatory programs and launching apoptotic cell death. Moreover, the DOE to PAO resulted in diminishing ERG amplitudes in a dose-dependent manner, indicating severe retinal damage. The current study established a prototype mouse model of arsenical-induced ocular damage that can be widely used to identify the key cellular signaling pathways involved in retinal damage pathobiology and to validate medical countermeasures against the progression of ocular damage.
Collapse
Affiliation(s)
- Assylbek Zhylkibayev
- Department of Optometry and Vision Science, School of Optometry, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ritesh Srivastava
- Research Center of Excellence in Arsenicals and Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | | | - Mohammad Athar
- Research Center of Excellence in Arsenicals and Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Marina Gorbatyuk
- Department of Optometry and Vision Science, School of Optometry, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
14
|
Masood F, Chang JH, Akbar A, Song A, Hu WY, Azar DT, Rosenblatt MI. Therapeutic Strategies for Restoring Perturbed Corneal Epithelial Homeostasis in Limbal Stem Cell Deficiency: Current Trends and Future Directions. Cells 2022; 11:3247. [PMID: 36291115 PMCID: PMC9600167 DOI: 10.3390/cells11203247] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/11/2022] [Accepted: 10/12/2022] [Indexed: 02/03/2023] Open
Abstract
Limbal stem cells constitute an important cell population required for regeneration of the corneal epithelium. If insults to limbal stem cells or their niche are sufficiently severe, a disease known as limbal stem cell deficiency occurs. In the absence of functioning limbal stem cells, vision-compromising conjunctivalization of the corneal epithelium occurs, leading to opacification, inflammation, neovascularization, and chronic scarring. Limbal stem cell transplantation is the standard treatment for unilateral cases of limbal stem cell deficiency, but bilateral cases require allogeneic transplantation. Herein we review the current therapeutic utilization of limbal stem cells. We also describe several limbal stem cell markers that impact their phenotype and function and discuss the possibility of modulating limbal stem cells and other sources of stem cells to facilitate the development of novel therapeutic interventions. We finally consider several hurdles for widespread adoption of these proposed methodologies and discuss how they can be overcome to realize vision-restoring interventions.
Collapse
Affiliation(s)
- Faisal Masood
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Jin-Hong Chang
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Anosh Akbar
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Amy Song
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Wen-Yang Hu
- Department of Urology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Dimitri T. Azar
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Mark I. Rosenblatt
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
15
|
Joseph LB, Gordon MK, Zhou P, Hahn RA, Lababidi H, Croutch CR, Sinko PJ, Heck DE, Laskin DL, Laskin JD. Sulfur mustard corneal injury is associated with alterations in the epithelial basement membrane and stromal extracellular matrix. Exp Mol Pathol 2022; 128:104807. [PMID: 35798063 PMCID: PMC10044521 DOI: 10.1016/j.yexmp.2022.104807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 05/20/2022] [Accepted: 06/29/2022] [Indexed: 12/15/2022]
Abstract
Sulfur mustard (SM; bis(2-chloroethyl) sulfide) is a highly reactive bifunctional alkylating agent synthesized for chemical warfare. The eyes are particularly sensitive to SM where it causes irritation, pain, photophobia, and blepharitis, depending on the dose and duration of exposure. In these studies, we examined the effects of SM vapor on the corneas of New Zealand white male rabbits. Edema and hazing of the cornea, signs of acute injury, were observed within one day of exposure to SM, followed by neovascularization, a sign of chronic or late phase pathology, which persisted for at least 28 days. Significant epithelial-stromal separation ranging from ~8-17% of the epithelial surface was observed. In the stroma, there was a marked increase in CD45+ leukocytes and a decrease of keratocytes, along with areas of disorganization of collagen fibers. SM also disrupted the corneal basement membrane and altered the expression of perlecan, a heparan sulfate proteoglycan, and cellular fibronectin, an extracellular matrix glycoprotein. This was associated with an increase in basement membrane matrix metalloproteinases including ADAM17, which is important in remodeling of the basement membrane during wound healing. Tenascin-C, an extracellular matrix glycoprotein, was also upregulated in the stroma 14-28 d post SM, a finding consistent with its role in organizing structural components of the stroma necessary for corneal transparency. These data demonstrate that SM vapor causes persistent alterations in structural components of the cornea. Further characterization of SM-induced injury in rabbit cornea will be useful for the identification of targets for the development of ocular countermeasures.
Collapse
Affiliation(s)
- Laurie B Joseph
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, United States of America.
| | - Marion K Gordon
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, United States of America
| | - Peihong Zhou
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, United States of America
| | - Rita A Hahn
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, United States of America
| | - Hamdi Lababidi
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, United States of America
| | | | - Patrick J Sinko
- Department of Pharmaceutical Science, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, United States of America
| | - Diane E Heck
- Department of Public Health, New York Medical College, Valhalla, NY 10595, United States of America
| | - Debra L Laskin
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, United States of America
| | - Jeffrey D Laskin
- Department of Environmental and Occupational Health and Justice, Rutgers University School of Public Health, Piscataway, NJ 08854, United States of America
| |
Collapse
|
16
|
Mishra N, Agarwal R. Research models of sulfur mustard- and nitrogen mustard-induced ocular injuries and potential therapeutics. Exp Eye Res 2022; 223:109209. [PMID: 35961426 DOI: 10.1016/j.exer.2022.109209] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 07/27/2022] [Accepted: 08/02/2022] [Indexed: 11/20/2022]
Abstract
Sulfur mustard (SM) is a notorious, bifunctional alkylating vesicant that was first used in warfare during World War I in 1917 and since then has been deployed in numerous skirmishes with its most recent documented use being during the Middle Eastern conflicts. Apart from its use in combat and terrorist activities, continual threat of accidental exposure from old stockpiles and improperly discarded munitions is ever present, especially to the innocent and unassuming civilian populations. SM can cause devastating injuries, depending on the dosage of SM exposure, route of exposure, as well as the physiological conditions of the individuals exposed. The most common routes of exposure are ocular, dermal, and exposure to the lungs and respiratory tissues through inhalation. Eyes are the most susceptible organ to SM-induced toxicities owing to their high moisture content and rapidly dividing cells. Additionally, ocular injury causes the most expeditious disablement of individuals even upon whole-body exposures. Therefore, it is imperative to understand the mechanisms underlying SM-induced ocular toxicity and design therapeutic interventions to prevent/mitigate ocular injuries. Ocular SM exposure may cause a wide range of symptoms such as inflammation, lacrimation, itching, dryness, photophobia, edema of the cornea/sclera/retina/iris, conjunctivitis, degradation of the corneal layer, fusion of two or more ocular layers, neovascularization, fibrosis, and temporary or permanent structural damage to one or more ocular layers. These symptoms may lead to vision impairments, resulting in partial or complete blindness that may be permanent. The highly toxic and exceedingly notorious nature of SM makes it a highly regulated chemical, requiring very expensive licensing, security, and safety requirements; thus, the more easily accessible analogue, nitrogen mustard (NM) that mimics SM-induced toxicity and injuries is employed in plethora of studies conducted in different animal models and culture systems. This review provides a comprehensive account of the injuries and symptoms that occur upon ocular SM exposures in human patients as well as studies in animal (in vivo, ex vivo) and cell (in vitro) models of SM and NM ocular exposures. Special emphasis has been laid on highlighting the strengths and lacunae in the research as well as the possible unexplored avenues of mechanisms underlying mustard-induced ocular injury that can be explored in future research endeavors. Furthermore, development of therapeutic interventions and targets of interest in the ocular system exposed to SM and NM, based on studies in human patients as well as in vivo, ex vivo, and in vitro models has been discussed in great depth, providing a valuable knowledge database to delineate pathways associated with vesicant-induced toxicity, and strategies/diagnostic tools against SM-induced toxicity.
Collapse
Affiliation(s)
- Neha Mishra
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Rajesh Agarwal
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA.
| |
Collapse
|
17
|
Goswami DG, Mishra N, Kant R, Agarwal C, Croutch CR, Enzenauer RW, Petrash MJ, Tewari-Singh N, Agarwal R. Pathophysiology and inflammatory biomarkers of sulfur mustard-induced corneal injury in rabbits. PLoS One 2021; 16:e0258503. [PMID: 34637469 PMCID: PMC8509852 DOI: 10.1371/journal.pone.0258503] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 09/28/2021] [Indexed: 01/01/2023] Open
Abstract
Sulfur mustard (SM) is a cytotoxic, vesicating, chemical warfare agent, first used in 1917; corneas are particularly vulnerable to SM exposure. They may develop inflammation, ulceration, neovascularization (NV), impaired vision, and partial/complete blindness depending upon the concentration of SM, exposure duration, and bio-physiological conditions of the eyes. Comprehensive in vivo studies have established ocular structural alterations, opacity, NV, and inflammation upon short durations (<4 min) of SM exposure. In this study, detailed analyses of histopathological alterations in corneal structure, keratocytes, inflammatory cells, blood vessels, and expressions of cyclooxygenase (COX)-2, matrix metalloproteinase (MMP)-9, vascular endothelial growth factor (VEGF), and cytokines were performed in New Zealand white rabbits, in a time-dependent manner till 28 days, post longer durations (5 and 7 min) of ocular SM exposure to establish quantifiable endpoints of injury and healing. Results indicated that SM exposure led to duration-dependent increases in corneal thickness, opacity, ulceration, epithelial-stromal separation, and epithelial degradation. Significant increases in NV, keratocyte death, blood vessels, and inflammatory markers (COX-2, MMP-9, VEGF, and interleukin-8) were also observed for both exposure durations compared to the controls. Collectively, these findings would benefit in temporal delineation of mechanisms underlying SM-induced corneal toxicity and provide models for testing therapeutic interventions.
Collapse
Affiliation(s)
- Dinesh G. Goswami
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Neha Mishra
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Rama Kant
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Chapla Agarwal
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Claire R. Croutch
- Medical Countermeasures Division, MRIGlobal, Kansas City, Missouri, United States of America
| | - Robert W. Enzenauer
- Department of Ophthalmology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Mark J. Petrash
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
- Department of Ophthalmology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Neera Tewari-Singh
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Rajesh Agarwal
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
- * E-mail:
| |
Collapse
|
18
|
Sabzevare M, Yazdani F, Karami A, Haddadi M, Aghamollaei H, Shahriary A. The effect of N-acetyl cysteine and doxycycline on TNF-α-Rel-a inflammatory pathway and downstream angiogenesis factors in the cornea of rats injured by 2-chloroethyl-ethyl sulfide. Immunopharmacol Immunotoxicol 2021; 43:452-460. [PMID: 34167418 DOI: 10.1080/08923973.2021.1939370] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
BACKGROUND Cornea injury of sulfur mustard (SM) is considered as the most devastating injuries to the eye. This study aimed to evaluate the single and combined effects of N-acetyl cysteine (NAC) and doxycycline on the inflammatory pathway and cornea neovascularization (CNV) in the rat model of SM-injured cornea. MATERIALS AND METHODS The right cornea of male Sprague-Dawley rats was subjected to 2-chloroethyl-ethyl sulfide (CEES). Rats were topically treated with a single and combined of 0.5% NAC and 12.5 μg/ml doxycycline and examined at 3rd, 15th, and 21st days. The activity of three antioxidant enzymes was analyzed in the cornea of different groups. Real-time PCR was performed to measure gene expression of inflammatory factors (tnf-α, rel-a & cxcl-1) and angiogenesis factors (vegf-a, mmp2,9) in the cornea lysates. The histological and opacity assessments were also carried out. RESULTS The activity of antioxidant enzymes significantly declined 3 days after the CEES damage. NAC eye drop recovered the enzyme activity on the 21st day of treatment (p-value < .05). The expression of tnf-α and rel-a genes significantly increased after CEES cornea exposure, while NAC declined their expression on the 7th and 21st days. The CNV score and angiogenesis factor expression were decreased in the long term by single and combined treatments (p-value < .05), but the infiltration of inflammatory cells was not completely amended. CONCLUSION NAC and doxycycline eye drop could improve the CNV complication. Also, NAC was an effective treatment against the inflammatory pathway involved in CEES-injured cornea.
Collapse
Affiliation(s)
- Melad Sabzevare
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Farshad Yazdani
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Ali Karami
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mohaddese Haddadi
- Department of Biology, Islamic Azad University of Branch Parand, Student of M.S of Molecular Genetics, Tehran, Iran
| | - Hossein Aghamollaei
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Alireza Shahriary
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
19
|
Fuchs A, Giuliano EA, Sinha NR, Mohan RR. Ocular toxicity of mustard gas: A concise review. Toxicol Lett 2021; 343:21-27. [PMID: 33600921 DOI: 10.1016/j.toxlet.2021.02.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 01/21/2021] [Accepted: 02/12/2021] [Indexed: 01/08/2023]
Abstract
Sulfur mustard (SM) is a chemical warfare agent that has been used throughout recent history and remains a threat today. Exposed soldiers and civilians experience a variety of symptoms primarily in the respiratory system, skin, and eyes. The ocular tissues are highly sensitive to damage by SM and undergo unique manifestations of acute, chronic, and delayed complications that can persist for months and years after exposure. The mechanisms of this unique mustard gas keratopathy are still not fully understood and animal models for the study of this disease are discussed. Recent advances in mechanisms of injury are included in this review. Ophthalmic manifestations of SM injury including persistent epithelial defects, limbal stem cell deficiency, corneal neovascularization, dry eye, and corneal opacification have been reported. A wide variety of medical and surgical therapies have been studied and are reviewed here along with potential future therapies.
Collapse
Affiliation(s)
- Allison Fuchs
- One-Health Vision Research Program, Departments of Veterinary Medicine & Surgery and Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, United States
| | - Elizabeth A Giuliano
- Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, United States; One-Health Vision Research Program, Departments of Veterinary Medicine & Surgery and Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, United States
| | - Nishant R Sinha
- Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, United States; One-Health Vision Research Program, Departments of Veterinary Medicine & Surgery and Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, United States
| | - Rajiv R Mohan
- Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, United States; One-Health Vision Research Program, Departments of Veterinary Medicine & Surgery and Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, United States; Mason Eye Institute, School of Medicine, University of Missouri, Columbia, MO, United States.
| |
Collapse
|
20
|
Gore A, Kadar T, Dachir S, Horwitz V. Therapeutic measures for sulfur mustard-induced ocular injury. Toxicol Lett 2021; 340:58-66. [PMID: 33440228 DOI: 10.1016/j.toxlet.2021.01.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 12/28/2020] [Accepted: 01/04/2021] [Indexed: 02/07/2023]
Abstract
The use of sulfur mustard (SM) in global terrorism is still a relevant threat to both civilian population and military personnel. Casualties exposed to SM may present mild, moderate or severe acute ocular lesions followed by a complete ocular resolution, chronic lesions or re-emerged ocular pathologies after a latent period. Current treatment for SM-induced ocular injury is based mainly on the clinical manifestation at the different stages of the injury and includes pharmaceutical and surgical interventions. These therapeutic measures are beneficial but not sufficient, and the ocular injury remains a continuous challenge for medical professionals. This review focuses on treatment experience carried out in humans and studied in animal models, for both SM-induced ocular acute injury and late pathology. In general, therapeutic measures are based on clinical features of the ocular injury or on the involvement of specific factors during the ocular injury that point out towards potential treatments. Anti-inflammatory treatments and limbal stem cell transplantation techniques were developed based on the clinical manifestation of the ocular injury. Optional therapies for impaired corneal innervation and endothelium are suggested for future research. Additionally, studies on potential treatments with anti-matrix metalloproteinase (MMP), anti-vascular endothelial growth factor (VEGF) and anti-IL-6 agents are discussed. Consequently, future studies may reveal the potential of additional pharmacological and biological treatments or advanced cellular and molecular biology methods to serve as novel therapeutic measures and techniques for this complicated ocular injury.
Collapse
Affiliation(s)
- Ariel Gore
- Department of Pharmacology, Israel Institute for Biological Research, Ness Ziona, 74100, Israel.
| | - Tamar Kadar
- Department of Pharmacology, Israel Institute for Biological Research, Ness Ziona, 74100, Israel
| | - Shlomit Dachir
- Department of Pharmacology, Israel Institute for Biological Research, Ness Ziona, 74100, Israel
| | - Vered Horwitz
- Department of Pharmacology, Israel Institute for Biological Research, Ness Ziona, 74100, Israel.
| |
Collapse
|
21
|
Araj H, Tumminia SJ, Yeung DT. Ocular Surface - Merging Challenges and Opportunities. Transl Vis Sci Technol 2020; 9:3. [PMID: 33200045 PMCID: PMC7645223 DOI: 10.1167/tvst.9.12.3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 07/08/2020] [Indexed: 12/14/2022] Open
Abstract
Dichotomies are double-edged: they can simplify and enlighten as well as exaggerate and entangle. Seeing the eye as anterior segment vs. posterior segment simplifies the formidable task of dissecting the function of the eye. Yet this view creates artificial divisions in a coherent whole. Clearly, vision requires the convergence of the light refractive function of the front of the eye with the light sensing function of the back of the eye. The National Eye Institute has long aimed to foster research across the visual pathway. Finding the right balance is a constant work in progress. A recently held scientific meeting which we co-organized with the United States Army Medical Research Institute of Chemical Defense, offered an opportunity to take stock of what the anterior segment in general, and the ocular surface in particular, bring to our understanding of biology and disease of the eye. Multiple dichotomies surfaced: acute vs. chronic disease; epithelial vs. endothelial damage; fibrotic vs. vascular pathology; inflammation vs. resolution response; chemical exposure vs. countermeasure; monotherapy vs. combination therapy; mechanistic vs. exploratory research; human vs. animal model. Merging some of these dichotomies is the goal of this paper.
Collapse
Affiliation(s)
- Houmam Araj
- Department of Health and Human Services, National Eye Institute/National Institutes of Health, Bethesda, MD, USA
| | - Santa J Tumminia
- Department of Health and Human Services, National Eye Institute/National Institutes of Health, Office of the Director, Bethesda, MD, USA
| | - David T Yeung
- Department of Health and Human Services, National Institute of Allergy and Infectious Diseases/National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
22
|
McGraw MD, Kim SY, White CW, Veress LA. Acute cytotoxicity and increased vascular endothelial growth factor after in vitro nitrogen mustard vapor exposure. Ann N Y Acad Sci 2020; 1479:223-233. [PMID: 32408394 DOI: 10.1111/nyas.14367] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 04/09/2020] [Accepted: 04/20/2020] [Indexed: 12/20/2022]
Abstract
Nitrogen mustard (NM) is a highly toxic alkylating agent. Inhalation exposure can cause acute and chronic lung injury. This study's aims were to develop an in vitro coculture model of mustard-induced airway injury and to identify growth factors contributing to airway pathology. Primary human bronchial epithelial cells cultured with pulmonary endothelial cells were exposed to NM (25, 50, 100, 250, or 500 μM) or PBS (control) for 1 hour. Lactate dehydrogenase (LDH) and transepithelial electrical resistance (TEER) were measured before and 24 h after NM exposure. Fixed cultures were stained for hematoxylin and eosin or live/dead staining. Culture media were analyzed for 11 growth factors. A 1-h vapor exposure to greater than or equal to 50 μM NM increased supernatant LDH, decreased TEER, and caused airway epithelial cell detachment. Endothelial cell death occurred at 500 μM NM. Vascular endothelial growth factor A (VEGF-A) and placental growth factor (PlGF) expression increased in 500 μM NM-exposed cultures compared with PBS-exposed control cultures. NM vapor exposure causes differential cytotoxicity to airway epithelial and endothelial injury in culture. Increased VEGF-A and PlGF expression occurred acutely in airway cocultures. Future studies are required to validate the role of VEGF signaling in mustard-induced airway pathology.
Collapse
Affiliation(s)
- Matthew D McGraw
- Department of Pediatric Pulmonology, University of Rochester Medical Center, Rochester, New York.,Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York
| | - So-Young Kim
- Department of Pediatric Pulmonology, University of Rochester Medical Center, Rochester, New York
| | - Carl W White
- Department of Pediatrics, Pulmonology Section, Pediatric Airway Research Center, University of Colorado Denver, Aurora, Colorado
| | - Livia A Veress
- Department of Pediatrics, Pulmonology Section, Pediatric Airway Research Center, University of Colorado Denver, Aurora, Colorado
| |
Collapse
|
23
|
Mahmoudzadeh R, Heidari-Keshel S, Mehrpour M, Asadi Amoli F, Aghajanpour L, Lashay A. Comparison of Topical Pigment Epithelium-Derived Factor (PEDF) with Topical Bevacizumab for Accelerating the Regression of Corneal Neovascularization in an Experimental Model of Rabbit Corneal Angiogenesis. Ocul Immunol Inflamm 2020; 29:1471-1477. [PMID: 32407187 DOI: 10.1080/09273948.2020.1751211] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Purpose: To evaluate the anti-angiogenic effect of topical administration of Pigment epithelium-derived factor (PEDF) on the reduction of corneal neovascularization (NV) in comparison to topical Bevacizumab.Methods: 18 eyes of 18 New Zealand rabbits were enrolled. Corneal NV was induced by a 7-0 silk suture. After suture removal, rabbits were randomly divided into three groups. In every group, one eye randomly treated with topical bevacizumab or topical PEDF or saline for 14 days. The area and length of neovascularization were measured by Image J. Histological studies were done in three groups.Results: After 14 days, the mean decrease of corneal NV length was 1.84 ± 0.17 mm (P < .001) in PEDF group and 1.6 ± 0.07 mm (P < .001) in bevacizumab group which was significantly more than the saline group (P = .001 and P < .001, respectively). There was no significant difference between PEDF and bevacizumab group in the reduction of corneal NV length (P = .85). The mean decrease of corneal NV area was 4.94 ± 0.55 mm2 (P < .001) in PEDF group and 4.23 ± 0.29 mm2 in the bevacizumab group (P < .001). PEDF and bevacizumab significantly decreased corneal NV area in comparison to the saline group (p = .017, p = .001, respectively). The mean decrease of corneal NV area did not show a significant difference between PEDF and bevacizumab groups (P = .72).Conclusion: Topical PEDF might be an effective and safe treatment option as bevacizumab in a short-term use, indicating that it is as good as the standard. However, long-term effect is required to be investigated.
Collapse
Affiliation(s)
- Raziyeh Mahmoudzadeh
- Eye Research Center, Farabi Eye Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Saeed Heidari-Keshel
- Eye Research Center, Farabi Eye Hospital, Tehran University of Medical Sciences, Tehran, Iran.,Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Mehrpour
- Eye Research Center, Farabi Eye Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Fahimeh Asadi Amoli
- Eye Research Center, Farabi Eye Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Leila Aghajanpour
- Eye Research Center, Farabi Eye Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Lashay
- Eye Research Center, Farabi Eye Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
24
|
Etemad L, Moshiri M, Balali-Mood M. Advances in treatment of acute sulfur mustard poisoning - a critical review. Crit Rev Toxicol 2020; 49:191-214. [PMID: 31576778 DOI: 10.1080/10408444.2019.1579779] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Sulfur mustard (SM) is a blistering chemical warfare agent that was used during the World War I and in the Iraq-Iran conflict. The aim of this paper is to discuss and critically review the published results of experiments on the treatment of SM poisoning based on our clinical and research experience. The victims must remove from the contaminated zone immediately. The best solution for decontamination is large amounts of water, using neutral soap and 0.5% sodium hypochlorite. Severely intoxicated patients should be treated according to advanced life support protocols and intensive care therapy for respiratory disorders and the chemical burn. Sodium thiosulfate infusion (100-500 mg/kg/min) should be started up to 60 min after SM exposure. However, N-acetyle cysteine (NAC) is recommended, none of them acts as specific or effective antidote. The important protective and conservative treatment of SM-induced pulmonary injuries include humidified oxygen, bronchodilators, NAC as muculytic, rehydration, mechanical ventilation, appropriate antibiotics and respiratory physiotherapy as clinically indicated. Treatment of acute SM ocular lesions start with topical antibiotics; preferably sulfacetamide eye drop, continue with lubricants, and artificial tears. Treatment for cutaneous injuries include: moist dressing; preferably with silver sulfadiazine cream, analgesic, anti-pruritic, physically debridement, debridase, Laser debridement, followed by skin autologous split-thickness therapy as clinically indicated. The new suggested medications and therapeutic approaches include: anti-inflammatory agents, Niacinamide, Silibinin, Calmodulin antagonists, Clobetasol, full-thickness skin grafting for skin injuries; Doxycycline; Bevacizumab, and Colchicine for ocular injuries. Recommended compounds based on animal studies include Niacinamide, Aprotinin, des-aspartate-angiotensin-I, Gamma-glutamyltransferase, vitamin E, and vitamin D. In vitro studies revealed that Dimethylthiourea, L-nitroarginine, Methyl-ester, Sodium pyruvate, Butylated hydroxyanisole, ethacrynic acid, and macrolide antibiotics are effective. However, none of them, except macrolide antibiotics have been proved clinically. Avoidance of inappropriate polypharmacy is advisable.
Collapse
Affiliation(s)
- Leila Etemad
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences , Mashhad , Iran
| | - Mohammad Moshiri
- Medical Toxicology Research Center, Faculty of Medicine, Mashhad University of Medical Sciences , Mashhad , Iran
| | - Mahdi Balali-Mood
- Medical Toxicology and Drug Abuse Research Center, Birjand University of Medical Sciences , Birjand , Iran
| |
Collapse
|
25
|
Horwitz V, Cohen-Gihon I, Egoz I, Dachir S, Cohen M, Cohen L, Gutman H, Gez R, Kadar T, Gore A, Beth-Din A, Zvi A, Zaide G, Israeli O. A comprehensive analysis of corneal mRNA levels during sulfur mustard induced ocular late pathology in the rabbit model using RNA sequencing. Exp Eye Res 2019; 184:201-212. [DOI: 10.1016/j.exer.2019.04.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 04/12/2019] [Indexed: 01/21/2023]
|
26
|
Acute corneal injury in rabbits following nitrogen mustard ocular exposure. Exp Mol Pathol 2019; 110:104275. [PMID: 31233733 DOI: 10.1016/j.yexmp.2019.104275] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 05/22/2019] [Accepted: 06/19/2019] [Indexed: 12/17/2022]
Abstract
Sulfur mustard (SM), a potent vesicating chemical warfare agent, and its analog nitrogen mustard (NM), are both strong bi-functional alkylating agents. Eyes, skin, and the respiratory system are the main targets of SM and NM exposure; however, ocular tissue is most sensitive, resulting in severe ocular injury. The mechanism of ocular injury from vesicating agents' exposure is not completely understood. To understand the injury mechanism from exposure to vesicating agents, NM has been previously employed in our toxicity studies on primary human corneal epithelial cells and ex vivo rabbit cornea organ culture model. In the current study, corneal toxicity from NM ocular exposure (1%) was analyzed for up to 28 days post-exposure in New Zealand White male rabbits to develop an acute corneal injury model. NM exposure led to conjunctival and eyelid swelling within a few hours after exposure, in addition to significant corneal opacity and ulceration. An increase in total corneal thickness and epithelial degradation was observed starting at day 3 post-NM exposure, which was maximal at day 14 post-exposure and did not resolve until 28 days post-exposure. There was an NM-induced increase in the number of blood vessels and inflammatory cells, and a decrease in keratocytes in the corneal stroma. NM exposure resulted in increased expression levels of cyclooxygenase-2, Interleukin-8, vascular endothelial growth factor and Matrix Metalloproteinase 9 indicating their involvement in NM-induced corneal injury. These clinical, biological, and molecular markers could be useful for the evaluation of acute corneal injury and to screen for therapies against NM- and SM-induced ocular injury.
Collapse
|
27
|
Eiger-Moscovich M, Livny E, Sella R, Gal-Or O, Nisgav Y, Livnat T, Bahar I. Comparison of Subconjunctival Aflibercept and Betamethasone for the Treatment of Formed Corneal Neovascularization in a Rabbit Model. Ophthalmic Res 2019; 62:116-122. [PMID: 31112967 DOI: 10.1159/000499165] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 02/19/2019] [Indexed: 11/19/2022]
Abstract
AIM To compare the efficacy of aflibercept (Eylea®), a potent antivascular endothelial growth factor (VEGF) agent, with betamethasone (Celestone®) and placebo for the treatment of formed corneal neovascularization in a rabbit model. METHODS A central corneal chemical burn was created in the right eye of 24 New Zealand albino rabbits. Four weeks later, the rabbits were randomly divided into 4 equal groups for subconjunctival injection of aflibercept, betamethasone, aflibercept+ betamethasone, or saline (control). Digital photographs taken at weekly intervals were rated by 2 masked observers for extent, centricity, and density of corneal neovascularization according to a predefined scale. The percentage of corneal surface involved by neovascularization was quantified by image analysis software (Fiji-J). The change in corneal neovascularization from treatment administration (4 weeks after injury) to 4 weeks later (8 weeks after injury) was assessed. The rabbits were then euthanized, and their eyes were enucleated and processed for histopathological and immunofluorescence studies. RESULTS There was no significant difference in the change in corneal neovascularization after treatment among the 4 groups according to the digital images (p > 0.15) or histological evaluation with hematoxylin and eosin (p > 0.08). On immunofluorescence assay, a lower VEGF concentration was observed in all treatment groups compared to the control group. CONCLUSIONS In this rabbit model, corneal neovascularization induced by chemical burn failed to regress with treatment with aflibercept, betamethasone, or their combination.
Collapse
Affiliation(s)
- Maya Eiger-Moscovich
- Department of Ophthalmology, Rabin Medical Center, Beilinson Hospital, Petach Tikva, Israel, .,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel,
| | - Eitan Livny
- Department of Ophthalmology, Rabin Medical Center, Beilinson Hospital, Petach Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ruti Sella
- Department of Ophthalmology, Rabin Medical Center, Beilinson Hospital, Petach Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Orly Gal-Or
- Department of Ophthalmology, Rabin Medical Center, Beilinson Hospital, Petach Tikva, Israel
| | - Yael Nisgav
- Laboratory of Eye Research, Felsenstein Medical Research Center, Beilinson Hospital, Petach Tikva, Israel
| | - Tami Livnat
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Laboratory of Eye Research, Felsenstein Medical Research Center, Beilinson Hospital, Petach Tikva, Israel
| | - Irit Bahar
- Department of Ophthalmology, Rabin Medical Center, Beilinson Hospital, Petach Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
28
|
Horwitz V, Dachir S, Cohen M, Gutman H, Cohen L, Gez R, Buch H, Kadar T, Gore A. Differential expression of corneal and limbal cytokines and chemokines throughout the clinical course of sulfur mustard induced ocular injury in the rabbit model. Exp Eye Res 2018; 177:145-152. [DOI: 10.1016/j.exer.2018.08.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 07/24/2018] [Accepted: 08/13/2018] [Indexed: 12/13/2022]
|
29
|
Goswami DG, Kant R, Tewari-Singh N, Agarwal R. Efficacy of anti-inflammatory, antibiotic and pleiotropic agents in reversing nitrogen mustard-induced injury in ex vivo cultured rabbit cornea. Toxicol Lett 2018; 293:127-132. [PMID: 29174984 DOI: 10.1016/j.toxlet.2017.11.026] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Revised: 11/13/2017] [Accepted: 11/22/2017] [Indexed: 01/01/2023]
Abstract
Vesicating agent, Sulfur mustard (SM), causes devastating eye injury; however, there are no effective antidotes available. Using nitrogen mustard (NM), a bi-functional analog of SM, we have earlier reported that NM-induced corneal injury in ex vivo rabbit cornea organ culture model parallels corneal injury reported with SM. Using this model, we have demonstrated the therapeutic efficacy of dexamethasone (DEX), doxycycline (DOX) and silibinin (SB) in reversing NM (2h exposure)-induced corneal injuries when added immediately after washing NM. In the present study, we further examined the efficacy of similar/higher doses of these agents when added immediately, 2, or 4h after washing NM following its 2h exposure. All three treatment agents caused a reversal in established NM-induced injury biomarkers when added immediately or 2h after washing NM following its 2h exposure; however, when treatments were carried out 4h after washing NM, there was no significant effect. Together, our results further show the beneficial effect of these agents in reversing NM-induced corneal injury and indicate the time window for effective treatment. This could be useful towards future development of targeted therapeutics against vesicant-induced ocular injury.
Collapse
Affiliation(s)
- Dinesh G Goswami
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Rama Kant
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Neera Tewari-Singh
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Rajesh Agarwal
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
30
|
Roshandel D, Eslani M, Baradaran-Rafii A, Cheung AY, Kurji K, Jabbehdari S, Maiz A, Jalali S, Djalilian AR, Holland EJ. Current and emerging therapies for corneal neovascularization. Ocul Surf 2018; 16:398-414. [PMID: 29908870 DOI: 10.1016/j.jtos.2018.06.004] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 06/10/2018] [Accepted: 06/12/2018] [Indexed: 02/08/2023]
Abstract
The cornea is unique because of its complete avascularity. Corneal neovascularization (CNV) can result from a variety of etiologies including contact lens wear; corneal infections; and ocular surface diseases due to inflammation, chemical injury, and limbal stem cell deficiency. Management is focused primarily on the etiology and pathophysiology causing the CNV and involves medical and surgical options. Because inflammation is a key factor in the pathophysiology of CNV, corticosteroids and other anti-inflammatory medications remain the mainstay of treatment. Anti-VEGF therapies are gaining popularity to prevent CNV in a number of etiologies. Surgical options including vessel occlusion and ocular surface reconstruction are other options depending on etiology and response to medical therapy. Future therapies should provide more effective treatment options for the management of CNV.
Collapse
Affiliation(s)
- Danial Roshandel
- Ocular Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Medi Eslani
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, USA; Cincinnati Eye Institute, Edgewood, KY/ University of Cincinnati, Department of Ophthalmology, Cincinnati, OH, USA
| | - Alireza Baradaran-Rafii
- Ocular Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Albert Y Cheung
- Cincinnati Eye Institute, Edgewood, KY/ University of Cincinnati, Department of Ophthalmology, Cincinnati, OH, USA
| | - Khaliq Kurji
- Cincinnati Eye Institute, Edgewood, KY/ University of Cincinnati, Department of Ophthalmology, Cincinnati, OH, USA
| | - Sayena Jabbehdari
- Ocular Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Alejandra Maiz
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Setareh Jalali
- Ocular Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali R Djalilian
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, USA.
| | - Edward J Holland
- Cincinnati Eye Institute, Edgewood, KY/ University of Cincinnati, Department of Ophthalmology, Cincinnati, OH, USA.
| |
Collapse
|
31
|
Giannos SA, Kraft ER, Zhao ZY, Merkley KH, Cai J. Photokinetic Drug Delivery: Near infrared (NIR) Induced Permeation Enhancement of Bevacizumab, Ranibizumab and Aflibercept through Human Sclera. Pharm Res 2018; 35:110. [PMID: 29600470 PMCID: PMC5876416 DOI: 10.1007/s11095-018-2392-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 03/19/2018] [Indexed: 01/04/2023]
Abstract
PURPOSE Permeation studies, with near infrared (NIR) light and anti-aggregation antibody formulation, were used to investigate the in vitro permeation of bevacizumab, ranibizumab and aflibercept through human sclera. METHODS A vertical, spherical Franz cell diffusion apparatus was used for this scleral tissue permeation model. A photokinetic ocular drug delivery (PODD) testing device accommodated the placement of NIR LEDs above the donor chambers. An adjustable LED driver/square wave generator provided electrical energy with a variable pulse rate and pulse width modulation (duty cycle). RESULTS Exposure to non-thermal NIR light had no effect on mAbs with regard to monomer concentration or antibody binding potential, as determined by SE-HPLC and ELISA. The optimal LED wavelength was found to be 950 nm. Duty cycle power of 5% vs 20% showed no difference in permeation. When compared to controls, the combination of non-aggregating antibody formulation and NIR illumination provided an average transscleral drug flux enhancement factor of 3X. CONCLUSION Narrow wavelength incoherent (non-laser) light from an NIR LED source is not harmful to mAbs and can be used to enhance drug permeation through scleral tissue. The topical formulation, combined with pulsed NIR light irradiation, significantly improved scleral permeation of three anti-VEGF antibody drugs.
Collapse
Affiliation(s)
- Steven A Giannos
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, 301 University Blvd, Galveston, Texas, 77555, USA.
| | - Edward R Kraft
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, 301 University Blvd, Galveston, Texas, 77555, USA
| | - Zhen-Yang Zhao
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, 301 University Blvd, Galveston, Texas, 77555, USA
| | - Kevin H Merkley
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, 301 University Blvd, Galveston, Texas, 77555, USA
| | - Jiyang Cai
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, 301 University Blvd, Galveston, Texas, 77555, USA
| |
Collapse
|
32
|
Gore A, Horwitz V, Cohen M, Gutman H, Cohen L, Gez R, Kadar T, Dachir S. Successful single treatment with ziv-aflibercept for existing corneal neovascularization following ocular chemical insult in the rabbit model. Exp Eye Res 2018; 171:183-191. [PMID: 29548928 DOI: 10.1016/j.exer.2018.03.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 03/11/2018] [Accepted: 03/11/2018] [Indexed: 12/27/2022]
Abstract
PURPOSE To evaluate the efficacy of ziv-aflibercept as a treatment for established corneal neovascularization (NV) and to compare its efficacy to that of bevacizumab following ocular chemical insult of sulfur mustard (SM) in the rabbit model. METHODS Chemical SM burn was induced in the right eye of NZW rabbits by vapor exposure. Ziv-aflibercept (2 mg) was applied once to neovascularized eyes by subconjunctival injection while subconjunctival bevacizumab (5 mg) was administered twice a week, for 3 weeks. Non-treated exposed eyes served as a control. A clinical follow-up employed by slit-lamp microscope, was performed up to 12 weeks following exposure and digital photographs of the cornea were taken for measurement of blood vessels length using the image analysis software. Eyes were taken for histological evaluation 2, 4 and 8 weeks following treatment for general morphology and for visualization of NV, using H&E and Masson Trichrome stainings, while conjunctival goblet cell density was determined by PAS staining. RESULTS Corneal NV developed, starting as early as two weeks after exposure. A single subconjunctival treatment of ziv-aflibercept at 4 weeks post exposure, significantly reduced the extent of existing NV already one week following injection, an effect which lasted for at least 8 weeks following treatment, while NV in the non-treated exposed eyes continued to advance. The extensive reduction in corneal NV in the ziv-aflibercept treated group was confirmed by histological evaluation. Bevacizumab multiple treatment showed a benefit in NV reduction, but to a lesser extent compared to the ziv-aflibercept treatment. Finally, ziv-aflibercept increased the density of conjunctival goblet cells as compared to the exposed non-treated group. CONCLUSIONS Subconjunctival ziv-aflibercept single treatment presented a highly efficient long-term therapeutic benefit in reducing existing corneal NV, following ocular sulfur mustard exposure. These findings show the robust anti-angiogenic efficacy of ziv-aflibercept and demonstrate the advantage of this treatment over the other anti-angiogenic therapies in ameliorating corneal NV and protecting the ocular surface.
Collapse
Affiliation(s)
- Ariel Gore
- Department of Pharmacology, Israel Institute for Biological Research, Ness Ziona, 74100, Israel.
| | - Vered Horwitz
- Department of Pharmacology, Israel Institute for Biological Research, Ness Ziona, 74100, Israel
| | - Maayan Cohen
- Department of Pharmacology, Israel Institute for Biological Research, Ness Ziona, 74100, Israel
| | - Hila Gutman
- Department of Pharmacology, Israel Institute for Biological Research, Ness Ziona, 74100, Israel
| | - Liat Cohen
- Department of Pharmacology, Israel Institute for Biological Research, Ness Ziona, 74100, Israel
| | - Rellie Gez
- Department of Pharmacology, Israel Institute for Biological Research, Ness Ziona, 74100, Israel
| | - Tamar Kadar
- Department of Pharmacology, Israel Institute for Biological Research, Ness Ziona, 74100, Israel
| | - Shlomit Dachir
- Department of Pharmacology, Israel Institute for Biological Research, Ness Ziona, 74100, Israel
| |
Collapse
|
33
|
Wu Y, Xue C, Lu Y, Huang Z. The inhibitory effect of different concentrations of KH902 eye drops on corneal neovascularization induced by alkali burn. Indian J Ophthalmol 2017; 65:1127-1132. [PMID: 29133637 PMCID: PMC5700579 DOI: 10.4103/ijo.ijo_339_17] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
PURPOSE The aim of this study was to evaluate the inhibitory effect of different concentrations of KH902 eye drops on rabbit corneal neovascularization (CNV) induced by alkali burn. METHODS Forty-eight adult rabbits were randomized into four groups after alkali burning: Group A (2.5 mg/ml), Group B (5 mg/ml), and Group C (10 mg/ml) by different concentrations of KH902 eye drops and Group D by saline solution as control with three times a day for 2 weeks. At days 7, 14, and 28, the anterior segment photographs, confocal microscopy, and histopathology were performed to evaluate corneal opacity, neovascularization, inflammatory cell density, vessel size, and edema. Immunohistochemistry was applied to analyze the vascular endothelial growth factor (VEGF) level. RESULTS (1) The CNV in the medicine-treated groups showed a reduction without obvious corneal side effects histologically. (2) Compared to the control group, the three medicine-treated groups showed a reduction in the VEGF levels and CNV areas on days 7, 14, and 28 and in the inflammatory cell density on days 14 and 28 (P < 0.01). The difference of inflammatory cell density between the three medicine-treated groups existed on day 14 (P < 0.01). There were differences in the VEGF levels between Groups A, B, and C on days 7, 14, and 28 (P < 0.01), not for Groups B and C on day 28 (P > 0.05). CONCLUSION KH902 eye drops in lower concentration showed an obvious reduction of the CNV growing for rabbit corneal alkali burn without side effects.
Collapse
Affiliation(s)
- Yan Wu
- Department of Ophthalmology, Jinling Medical School of Nanjing Medical University; Department of Ophthalmology, Jinling Hospital, Nanjing 210002, Jiangsu Province, PR China
| | - Chunyan Xue
- Department of Ophthalmology, Jinling Hospital, Nanjing 210002, Jiangsu Province, PR China
| | - Yan Lu
- Department of Ophthalmology, Jinling Hospital, Nanjing 210002, Jiangsu Province, PR China
| | - Zhenping Huang
- Department of Ophthalmology, Jinling Medical School of Nanjing Medical University; Department of Ophthalmology, Jinling Hospital, Nanjing 210002, Jiangsu Province, PR China
| |
Collapse
|
34
|
Tewari-Singh N, Goswami DG, Kant R, Ammar DA, Kumar D, Enzenauer RW, Casillas RP, Croutch CR, Petrash JM, Agarwal R. Histopathological and Molecular Changes in the Rabbit Cornea From Arsenical Vesicant Lewisite Exposure. Toxicol Sci 2017; 160:420-428. [PMID: 28973427 PMCID: PMC5837587 DOI: 10.1093/toxsci/kfx198] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Lewisite (LEW), a potent arsenical vesicating chemical warfare agent, poses a continuous risk of accidental exposure in addition to its feared use as a terrorist weapon. Ocular tissue is exquisitely sensitive to LEW and exposure can cause devastating corneal lesions. However, detailed pathogenesis of corneal injury and related mechanisms from LEW exposure that could help identify targeted therapies are not available. Using an established consistent and efficient exposure system, we evaluated the pathophysiology of the corneal injury in New Zealand white rabbits following LEW vapor exposure (at 0.2 mg/L dose) for 2.5 and 7.5 min, for up to 28 day post-exposure. LEW led to an increase in total corneal thickness starting at day 1 post-exposure and epithelial degradation starting at day 3 post-exposure, with maximal effect at day 7 postexposure followed by recovery at later time points. LEW also led to an increase in the number of blood vessels and inflammatory cells but a decrease in keratocytes with optimal effects at day 7 postexposure. A significant increase in epithelial-stromal separation was observed at days 7 and 14 post 7.5 min LEW exposure. LEW also caused an increase in the expression levels of cyclooxygenase-2, IL-8, vascular endothelial growth factor, and matrix metalloproteinase-9 at all the study time points indicating their involvement in LEW-induced inflammation, vesication, and neovascularization. The outcomes here provide valuable LEW-induced corneal injury endpoints at both lower and higher exposure durations in a relevant model system, which will be helpful to identify and screen therapies against LEW-induced corneal injury.
Collapse
Affiliation(s)
| | | | - Rama Kant
- Department of Pharmaceutical Sciences
| | - David A Ammar
- Department of Ophthalmology, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
| | | | - Robert W Enzenauer
- Department of Ophthalmology, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
| | - Robert P Casillas
- Medical Countermeasures Division, MRIGlobal, Kansas City, Missouri 64110
| | - Claire R Croutch
- Medical Countermeasures Division, MRIGlobal, Kansas City, Missouri 64110
| | - J Mark Petrash
- Department of Ophthalmology, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
| | | |
Collapse
|
35
|
Goswami DG, Agarwal R, Tewari-Singh N. Phosgene oxime: Injury and associated mechanisms compared to vesicating agents sulfur mustard and lewisite. Toxicol Lett 2017; 293:112-119. [PMID: 29141200 DOI: 10.1016/j.toxlet.2017.11.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 11/08/2017] [Accepted: 11/11/2017] [Indexed: 12/24/2022]
Abstract
Phosgene Oxime (CX, Cl2CNOH), a halogenated oxime, is a potent chemical weapon that causes immediate acute injury and systemic effects. CX, grouped together with vesicating agents, is an urticant or nettle agent with highly volatile, reactive, corrosive, and irritating vapor, and has considerably different chemical properties and toxicity compared to other vesicants. CX is absorbed quickly through clothing with faster cutaneous penetration compared to other vesicating agents causing instantaneous and severe damage. For this reason, it could be produced as a weaponized mixture with other chemical warfare agents to enhance their deleterious effects. The immediate devastating effects of CX and easy synthesis makes it a dangerous chemical with both military and terrorist potentials. Although CX is the most potent vesicating agent, it is one of the least studied chemical warfare agents and the pathophysiology as well as long term effects are largely unknown. CX exposure results in immediate pain and inflammation, and it mainly affects skin, eye and respiratory system. There are no antidotes available against CX-induced injury and the treatment is only supportive. This review summarizes existing knowledge regarding exposure, toxicity and the probable underlying mechanisms of CX compared to other important vesicants' exposure.
Collapse
Affiliation(s)
- Dinesh Giri Goswami
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Rajesh Agarwal
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Neera Tewari-Singh
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.
| |
Collapse
|
36
|
Panahi Y, Rajaee SM, Sahebkar A. Ocular Effects of Sulfur Mustard and Therapeutic Approaches. J Cell Biochem 2017; 118:3549-3560. [DOI: 10.1002/jcb.25892] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 01/18/2017] [Indexed: 01/06/2023]
Affiliation(s)
- Yunes Panahi
- Chemical Injuries Research CenterBaqiyatallah University of Medical SciencesTehranIran
| | - Seyyed Mahdi Rajaee
- Chemical Injuries Research CenterBaqiyatallah University of Medical SciencesTehranIran
| | | |
Collapse
|
37
|
Kim JY, Kim SY, Cheon MH, Kim ES, Song IS, Kim MJ, Tchah H. Attenuation of corneal neovascularization by topical low-molecular-weight heparin-taurocholate 7 without bleeding complication. Int J Ophthalmol 2016; 9:1255-9. [PMID: 27672587 DOI: 10.18240/ijo.2016.09.03] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2015] [Accepted: 09/08/2015] [Indexed: 11/23/2022] Open
Abstract
AIM To investigate the antiangiogenic effects and safety of topically administered low-molecular-weight heparin-taurocholate 7 (LHT7) on corneal neovascularization (CoNV). METHODS Twenty-four Sprague-Dawley rats were randomly distributed into four groups of six rats each. The central corneas were cauterized using a silver/potassium nitrate solution. From 2d after cauterization, 12.5 mg/mL (low LHT7 group) or 25 mg/mL (high LHT7 group) LHT7 was topically administered three times daily; 12.5 mg/mL bevacizumab was topically administered as positive control (bevacizumab) group, with normal saline (NS) administered as negative control (NS group). The corneas were digitally photographed to calculate the CoNV percentage from the neovascularized corneal area at 1 and 2wk. RESULTS The 4 study groups did not have different CoNV percentages at 1wk after injury (P>0.05). However, the low LHT, high LHT, and bevacizumab groups had significantly lower CoNV percentages than the NS group at 2wk (all P<0.05). No significant differences in CoNV percentage were found among the low LHT, high LHT, and bevacizumab groups (all P>0.05). All groups except the NS group had lower CoNV percentages at 2wk post-injury than the levels observed at 1wk (all P<0.05). CONCLUSION Topically-administered LHT7 inhibited CoNV without complication after chemical cauterization in the rat.
Collapse
Affiliation(s)
- Jae Yong Kim
- Department of Ophthalmology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea
| | - Soo Yeon Kim
- Department of Ophthalmology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea
| | - Mi Hyun Cheon
- Department of Ophthalmology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea
| | - Eun-Soon Kim
- Department of Ophthalmology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea
| | - In Seok Song
- Department of Ophthalmology, Hanyang University College of Medicine, Seoul 04763, Korea
| | - Myoung Joon Kim
- Department of Ophthalmology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea
| | - Hungwon Tchah
- Department of Ophthalmology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea
| |
Collapse
|
38
|
Nitrogen Mustard-Induced Corneal Injury Involves DNA Damage and Pathways Related to Inflammation, Epithelial-Stromal Separation, and Neovascularization. Cornea 2016; 35:257-66. [PMID: 26555588 DOI: 10.1097/ico.0000000000000685] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE To evaluate the toxic effects and associated mechanisms in corneal tissue exposed to the vesicating agent, nitrogen mustard (NM), a bifunctional alkylating analog of the chemical warfare agent sulfur mustard. METHODS Toxic effects and associated mechanisms were examined in maximally affected corneal tissue using corneal cultures and human corneal epithelial (HCE) cells exposed to NM. RESULTS Analysis of ex vivo rabbit corneas showed that NM exposure increased apoptotic cell death, epithelial thickness, epithelial-stromal separation, and levels of vascular endothelial growth factor, cyclooxygenase 2, and matrix metalloproteinase-9. In HCE cells, NM exposure resulted in a dose-dependent decrease in cell viability and proliferation, which was associated with DNA damage in terms of an increase in p53 ser15, total p53, and H2A.X ser139 levels. NM exposure also induced caspase-3 and poly ADP ribose polymerase cleavage, suggesting their involvement in NM-induced apoptotic death in the rabbit cornea and HCE cells. Similar to rabbit cornea, NM exposure caused an increase in cyclooxygenase 2, matrix metalloproteinase-9, and vascular endothelial growth factor levels in HCE cells, indicating a role of these molecules and related pathways in NM-induced corneal inflammation, epithelial-stromal separation, and neovascularization. NM exposure also induced activation of activator protein 1 transcription factor proteins and upstream signaling pathways including mitogen-activated protein kinases and Akt protein kinase, suggesting that these could be key factors involved in NM-induced corneal injury. CONCLUSIONS Results from this study provide insight into the molecular targets and pathways that could be involved in NM-induced corneal injuries laying the background for further investigation of these pathways in vesicant-induced ocular injuries, which could be helpful in the development of targeted therapies.
Collapse
|
39
|
Goswami DG, Tewari-Singh N, Agarwal R. Corneal toxicity induced by vesicating agents and effective treatment options. Ann N Y Acad Sci 2016; 1374:193-201. [PMID: 27327041 DOI: 10.1111/nyas.13121] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 05/04/2016] [Accepted: 05/10/2016] [Indexed: 12/20/2022]
Abstract
The vesicating agents sulfur mustard (SM) and lewisite (LEW) are potent chemical warfare agents that primarily cause damage to the ocular, skin, and respiratory systems. However, ocular tissue is the most sensitive organ, and vesicant exposure results in a biphasic injury response, including photophobia, corneal lesions, corneal edema, ulceration, and neovascularization, and may cause loss of vision. There are several reports on ocular injury from exposure to SM, which has been frequently used in warfare. However, there are very few reports on ocular injury by LEW, which indicate that injury symptoms appear instantly after exposure and faster than SM. In spite of extensive research efforts, effective therapies for vesicant-induced ocular injuries, mainly to the most affected corneal tissue, are not available. Hence, we have established primary human corneal epithelial cells and rabbit corneal organ culture models with the SM analog nitrogen mustard, which have helped to test the efficacy of potential therapeutic agents. These agents will then be further evaluated against in vivo SM- and LEW-induced corneal injury models, which will assist in the development of potential broad-spectrum therapies against vesicant-induced ocular injuries.
Collapse
Affiliation(s)
- Dinesh G Goswami
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Neera Tewari-Singh
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Rajesh Agarwal
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| |
Collapse
|
40
|
Kılıç Müftüoğlu İ, Aydın Akova Y. Clinical Findings, Follow-up and Treatment Results in Patients with Ocular Rosacea. Turk J Ophthalmol 2016; 46:1-6. [PMID: 27800249 PMCID: PMC5076302 DOI: 10.4274/tjo.48902] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2014] [Accepted: 04/07/2015] [Indexed: 12/02/2022] Open
Abstract
Objectives: To report the clinical features, treatment options and complications in patients with ocular rosacea. Materials and Methods: The records of 48 eyes of 24 patients with ocular rosacea were retrospectively reviewed. Patients’ ocular signs and symptoms were scored between 1 and 4 points according to disease severity; tear film break-up time (BUT) and Schirmer’s test results were recorded before and after the treatment. Preservative-free artificial tears, topical antibiotic eye drops/ointments, short-term topical corticosteroids, topical 0.05% cyclosporine and oral doxycycline treatment were applied as a standard therapy to all patients. Additional treatments were given as needed. Complications were recorded. Results: Twenty-four patients with a mean age of 48.5±35.4 (32-54) years were followed for a mean 15±9.4 (8-36) months. Ocular findings included meibomitis in 100% of cases, anterior blepharitis in 83% (40 eyes), punctate keratopathy in 67% (32 eyes), chalazia in 50% (24 eyes), corneal neovascularization in 50% (24 eyes) and subepithelial infiltrates in 16.6% (8 eyes). Significant improvement of symptoms and clinical findings were achieved in all patients with treatment. The increases in Schirmer’s test and BUT were 3.3±1.5 and 4.5±2.8, respectively (p<0.05). Descemetocele and small corneal perforation occurred in 2 eyes; re-epithelialization was achieved in both eyes with tissue adhesive application (1 eye) and additional amniotic membrane transplantation (1 eye). Four eyes of three patients showed significant regression of corneal neovascularization with topical bevacizumab therapy. Conclusion: Ocular rosacea may present with a variety of ophthalmic signs. It is possible to control the ophthalmic disease with appropriate therapeutic modalities including topical corticosteroids, topical cyclosporine and systemic doxycycline.
Collapse
|
41
|
Gronkiewicz KM, Giuliano EA, Kuroki K, Bunyak F, Sharma A, Teixeira LBC, Hamm CW, Mohan RR. Development of a novel in vivo corneal fibrosis model in the dog. Exp Eye Res 2015; 143:75-88. [PMID: 26450656 DOI: 10.1016/j.exer.2015.09.010] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 09/09/2015] [Accepted: 09/16/2015] [Indexed: 12/11/2022]
Abstract
The aim of this study was to develop a novel in vivo corneal model of fibrosis in dogs utilizing alkali burn and determine the ability of suberanilohydroxamic acid (SAHA) to inhibit corneal fibrosis using this large animal model. To accomplish this, we used seven research Beagle dogs. An axial corneal alkali burn in dogs was created using 1 N NaOH topically. Six dogs were randomly and equally assigned into 2 groups: A) vehicle (DMSO, 2 μL/mL); B) anti-fibrotic treatment (50 μM SAHA). The degree of corneal opacity, ocular health, and anti-fibrotic effects of SAHA were determined utilizing the Fantes grading scale, modified McDonald-Shadduck (mMS) scoring system, optical coherence tomography (OCT), corneal histopathology, immunohistochemistry (IHC), and transmission electron microscopy (TEM). The used alkali burn dose to produce corneal fibrosis was well tolerated as no significant difference in mMS scores between control and treatment groups (p = 0.89) were detected. The corneas of alkali burned dogs showed significantly greater levels of α-smooth muscle actin, the fibrotic marker, than the controls (p = 0.018). Total corneal thickness of all dogs post-burn was significantly greater than baseline OCT images irrespective of treatment (p = 0.004); TEM showed that alkali burned corneas had significantly greater minimum and maximum interfibrillar distances than the controls (p = 0.026, p = 0.018). The tested topical corneal alkali burn dose generated significant opacity and fibrosis in dog corneas without damaging the limbus as evidenced by histopathology, IHC, TEM, and OCT findings, and represents a viable large animal corneal fibrosis in vivo model. Additional in vivo SAHA dosing studies with larger sample size are warranted.
Collapse
Affiliation(s)
- K M Gronkiewicz
- Department of Veterinary Medicine and Surgery, College of Veterinary Medicine, Columbia, MO, USA; Harry S. Truman Memorial Veteran Hospital, Columbia, MO, USA
| | - E A Giuliano
- Department of Veterinary Medicine and Surgery, College of Veterinary Medicine, Columbia, MO, USA; Harry S. Truman Memorial Veteran Hospital, Columbia, MO, USA
| | - K Kuroki
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Columbia, MO, USA
| | - F Bunyak
- Department of Computer Science, University of Missouri, Columbia, MO, USA
| | - A Sharma
- Department of Veterinary Medicine and Surgery, College of Veterinary Medicine, Columbia, MO, USA; Harry S. Truman Memorial Veteran Hospital, Columbia, MO, USA
| | - L B C Teixeira
- Department of Pathological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, WI, USA
| | - C W Hamm
- Mason Eye Institute, University of Missouri, Columbia, MO, USA
| | - R R Mohan
- Department of Veterinary Medicine and Surgery, College of Veterinary Medicine, Columbia, MO, USA; Harry S. Truman Memorial Veteran Hospital, Columbia, MO, USA; Mason Eye Institute, University of Missouri, Columbia, MO, USA.
| |
Collapse
|
42
|
Abbaszadeh M, Aidenloo NS, Nematollahi MK, Motarjemizadeh Q. Investigating the Association between Angiogenic Cytokines and Corneal Neovascularization in Sulfur Mustard Intoxicated Subjects 26 Years after Exposure. Toxicol Int 2015; 21:300-6. [PMID: 25948970 PMCID: PMC4413414 DOI: 10.4103/0971-6580.155375] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
OBJECTIVES This study aimed to evaluate the associations between the concentrations of three major angiogenic cytokines-vascular endothelial growth factor-A165 (VEGF-A165), basic fibroblast growth factor (bFGF), and platelet-derived growth factor-BB (PDGF-BB)-in the tear of sulfur mustard (SM)-exposed subjects and corneal neovascularization (CNV) 26 years after exposure. MATERIALS AND METHODS The concentrations of VEGF-A, bFGF, and PDGF-BB were measured by enzyme-linked immunosorbent assay (ELISA) in reflex tears of (i) SM-injured patients with CNV (positive case group including 18 individuals) and (ii) SM-injured patients without CNV (negative case group including 22 individuals). Then results were compared to corresponding values obtained from tears of 40 healthy control subjects. RESULTS The mean concentrations of all investigated growth factors, VEGF-A165, bFGF, and PDGF-BB, were significantly higher in positive cases than controls (P ≤ 0.001, P = 0.028, and P = 0.041, respectively). Whereas, VEGF-A165 was the only growth factor which displayed significantly elevated concentrations in negative case group compared to the healthy individuals (P = 0.030). Additionally, the mean level of VEGF-A165 was also higher in positive patient group than negative patients (P = 0.022). Subjects with increased concentrations of tear VEGF-A165 were more than 10 times more likely to suffer from CNV than normal individuals (odds ratio (OR) = 10.43, confidence interval (CI): 2.14-38.46, P = 0.001), while elevated levels of bFGF and PDGF-BB increased the risk of CNV by about twofold. CONCLUSION Although all investigated cytokines had increased in tears of positive patients, VEGF-A was the only one which showed a significant correlation with the severity of CNV, and thus played a crucial role in corneal angiogenesis.
Collapse
|