1
|
Orive Bañuelos A, Sacristán Egüén C, Arce Soto A, Santamaría Carro A, Feijóo Lera R, Andollo Victoriano N, Etxebarria Ecenarro J. Boston Type II Keratoprosthesis, 7 Years of Experience and Outcomes From a Spanish Tertiary Eye Care Hospital. Am J Ophthalmol 2024; 269:327-338. [PMID: 39214393 DOI: 10.1016/j.ajo.2024.08.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 08/06/2024] [Accepted: 08/09/2024] [Indexed: 09/04/2024]
Abstract
PURPOSE To report midterm outcomes of Boston Keratoprosthesis type II at a reference center in Spain. DESIGN Retrospective case series. METHODS This observational study included medical records of 9 patients who underwent Boston Keratoprosthesis type II surgery at Cruces University Hospital from May 2016 through May 2023. A total of 9 eyes received this device during the study period. One patient who had undergone a modification of the standard procedure was excluded. Preoperative and postoperative parameters were studied. The main outcomes analyzed were visual acuity, device retention, and complications and their management. RESULTS The most common indication for implantation was severe Sjögren syndrome (33.3%) followed by graft-versus-host disease (22.2%). The mean follow-up was 3.89 years (±2.08). Visual acuity improved to better than or equal to 20/40 in 8 eyes (88.8%) and 20/25 in 6 eyes (66.6%), while best-corrected visual acuity of 20/20 was achieved in 4 eyes (44.4%). At the last visit, maximum visual acuity was maintained in 4 patients. The most common postoperative complication was retroprosthetic membrane formation (77.7%). New-onset glaucoma was detected in 4 patients (44.4%). Device extrusion occurred in 1 eye and 2 more patients required replacement surgery. All but 1 of the patients are on postoperative treatment with systemic immunosuppressants. CONCLUSIONS The Boston type II Keratoprosthesis is a useful option for visual rehabilitation in end-stage ocular surface diseases. Multidisciplinary management of complications is of vital importance for the maintenance of vision and the device. Immunosuppressive treatment helps control the inflammation that leads to most associated complications.
Collapse
Affiliation(s)
- Ana Orive Bañuelos
- From the Department of Ophthalmology (A.O.B., C.S.E., A.S.C., R.F.L., N.A.V., J.E.E.), BioCruces Bizkaia Health Research Institute, University Hospital of Cruces, Barakaldo, Spain.
| | - Cristina Sacristán Egüén
- From the Department of Ophthalmology (A.O.B., C.S.E., A.S.C., R.F.L., N.A.V., J.E.E.), BioCruces Bizkaia Health Research Institute, University Hospital of Cruces, Barakaldo, Spain
| | - Ana Arce Soto
- Department of Ophthalmology (A.A.S.), Hospital of Manacor, Spain
| | - Alaitz Santamaría Carro
- From the Department of Ophthalmology (A.O.B., C.S.E., A.S.C., R.F.L., N.A.V., J.E.E.), BioCruces Bizkaia Health Research Institute, University Hospital of Cruces, Barakaldo, Spain
| | - Raquel Feijóo Lera
- From the Department of Ophthalmology (A.O.B., C.S.E., A.S.C., R.F.L., N.A.V., J.E.E.), BioCruces Bizkaia Health Research Institute, University Hospital of Cruces, Barakaldo, Spain
| | - Noelia Andollo Victoriano
- From the Department of Ophthalmology (A.O.B., C.S.E., A.S.C., R.F.L., N.A.V., J.E.E.), BioCruces Bizkaia Health Research Institute, University Hospital of Cruces, Barakaldo, Spain; Department of Cell Biology and Histology (N.A.V., J.E.E.), School of Medicine and Nursing, University of the Basque Country, Leioa, Spain
| | - Jaime Etxebarria Ecenarro
- From the Department of Ophthalmology (A.O.B., C.S.E., A.S.C., R.F.L., N.A.V., J.E.E.), BioCruces Bizkaia Health Research Institute, University Hospital of Cruces, Barakaldo, Spain; Department of Cell Biology and Histology (N.A.V., J.E.E.), School of Medicine and Nursing, University of the Basque Country, Leioa, Spain
| |
Collapse
|
2
|
Yu F, Zhao X, Wang Q, Fang PH, Liu L, Du X, Li W, He D, Zhang T, Bai Y, Liu L, Li S, Yuan J. Engineered Mesenchymal Stromal Cell Exosomes-Loaded Microneedles Improve Corneal Healing after Chemical Injury. ACS NANO 2024. [PMID: 39047084 DOI: 10.1021/acsnano.4c00423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/27/2024]
Abstract
Corneal alkali burns represent a prevalent ophthalmic emergency with the potential to induce blindness. The main contributing mechanisms include excessive inflammation and delayed wound healing. Existing clinical therapies have limitations, promoting the exploration of alternative methods that offer improved efficacy and reduced side effects. Adipose-derived stem cell-exosome (ADSC-Exo) has the potential to sustain immune homeostasis and facilitate tissue regeneration. Nevertheless, natural ADSC-Exo lacks disease specificity and exhibits limited bioavailability on the ocular surface. In this study, we conjugated antitumor necrosis factor-α antibodies (aT) to the surface of ADSC-Exo using matrix metalloproteinase-cleavable peptide chains to create engineered aT-Exo with synergistic effects. In both in vivo and in vitro assessments, aT-Exo demonstrated superior efficacy in mitigating corneal injuries compared to aT alone, unmodified exosomes, or aT simply mixed with exosomes. The cleavable conjugation of aT-Exo notably enhanced wound healing and alleviated inflammation more effectively. Simultaneously, we developed poly(vinyl alcohol) microneedles (MNs) for precise and sustained exosome delivery. The in vivo results showcased the superior therapeutic efficiency of MNs compared with conventional topical administration and subconjunctival injection. Therefore, the bioactive nanodrugs-loaded MNs treatment presents a promising strategy for addressing ocular surface diseases.
Collapse
Affiliation(s)
- Fei Yu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510623, China
| | - Xuan Zhao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510623, China
| | - Qian Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510623, China
| | - Po-Han Fang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510623, China
| | - Liu Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510623, China
| | - Xinyue Du
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510623, China
| | - Weihua Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510623, China
| | - Dalian He
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510623, China
| | - Tingting Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510623, China
| | - Ying Bai
- Guangdong Engineering Technology Research Centre for Functional Biomaterials, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510006, China
| | - Lu Liu
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR 999077, China
| | - Saiqun Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510623, China
| | - Jin Yuan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510623, China
| |
Collapse
|
3
|
Yao Q, Wu H, Ren H, Cao J, Shao Y, Liu G, Lu P. Inhibition of Experimental Corneal Neovascularization by the Tight Junction Protein ZO-1. J Ocul Pharmacol Ther 2024; 40:379-388. [PMID: 39172123 DOI: 10.1089/jop.2023.0162] [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] [Indexed: 08/23/2024] Open
Abstract
Purpose: To explore the effects of the tight junction protein zonula occludens 1 (ZO-1) on experimental corneal neovascularization (CNV). Methods: CNV models were established in the left eyes of BALB/c mice using NaOH. Anti-ZO-1 neutralizing antibody was topically applied to the burnt corneas after modeling thrice a day for 1 week. CD31 expression was analyzed to calculate the ratio of CNV number to area using a corneal whole-mount fluorescent immunohistochemical assay. Messenger ribonucleic acid (mRNA) and protein expression levels of ZO-1, vascular endothelial growth factor (VEGF), interleukin (IL)-1β, IL-6, IL-8, IL-18, monocyte chemoattractant protein-1 (MCP-1), tumor necrosis factor alpha (TNF-α), phosphorylated protein kinase C (pPKC), and clusterin in burned corneas were detected by reverse transcriptase polymerase chain reaction (PCR) and western blot analyses. Infiltration of neutrophils, macrophages, and progenitor cells was examined by flow cytometry. Results: CNV was obviously greater in 45 s than in 15 s alkali injury group. In another experiment, CNV was obviously greater in the ZO-1 antibody group than in the vehicle-treated group. Corneal mRNA and protein expression levels of VEGF, IL-1β, IL-6, IL-8, IL-18, and MCP-1 were significantly higher in the ZO-1 antibody group than in the control group. Infiltration of neutrophils, macrophages, and progenitor cells was significantly greater in the ZO-1 antibody group than in the control group. TNF-α expression was much higher in 45 s than in 15 s alkali injury group. However, protein expression of pPKC and clusterin was much lower in 45 s than in 15 s alkali injury group. Conclusions: Anti-ZO-1 neutralizing antibody-treated mice exhibited enhanced alkali-induced CNV through enhanced intracorneal infiltration of progenitor and inflammatory cells.
Collapse
Affiliation(s)
- Qingying Yao
- Department of Ophthalmology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Hongya Wu
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Hang Ren
- Department of Ophthalmology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jiufa Cao
- Department of Ophthalmology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Ying Shao
- Department of Ophthalmology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Gaoqin Liu
- Department of Ophthalmology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Peirong Lu
- Department of Ophthalmology, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
4
|
Paschalis EI, Zhou C, Sharma J, Dohlman TH, Kim S, Lei F, Chodosh J, Vavvas D, Urtti A, Papaliodis G, Dohlman CH. The prophylactic value of TNF-α inhibitors against retinal cell apoptosis and optic nerve axon loss after corneal surgery or trauma. Acta Ophthalmol 2024; 102:e381-e394. [PMID: 37803488 PMCID: PMC10997738 DOI: 10.1111/aos.15786] [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: 03/13/2023] [Revised: 09/13/2023] [Accepted: 09/18/2023] [Indexed: 10/08/2023]
Abstract
BACKGROUND AND PURPOSE Late secondary glaucoma is an often-severe complication after acute events like anterior segment surgery, trauma and infection. TNF-α is a major mediator that is rapidly upregulated, diffusing also to the retina and causes apoptosis of the ganglion cells and degeneration of their optic nerve axons (mediating steps to glaucomatous damage). Anti-TNF-α antibodies are in animals very effective in protecting the retinal cells and the optic nerve-and might therefore be useful prophylactically against secondary glaucoma in future such patients. Here we evaluate (1) toxicity and (2) efficacy of two TNF-α inhibitors (adalimumab and infliximab), in rabbits by subconjunctival administration. METHODS For drug toxicity, animals with normal, unburned corneas were injected with adalimumab (0.4, 4, or 40 mg), or infliximab (1, 10, or 100 mg). For drug efficacy, other animals were subjected to alkali burn before such injection, or steroids (for control). The rabbits were evaluated clinically with slit lamp and photography, electroretinography, optical coherence tomography, and intraocular pressure manometry. A sub-set of eyes were stained ex vivo after 3 days for retinal cell apoptosis (TUNEL). In other experiments the optic nerves were evaluated by paraphenylenediamine staining after 50 or 90 days. Loss of retinal cells and optic nerve degeneration were quantified. RESULTS Subconjunctival administration of 0.4 mg or 4.0 mg adalimumab were well tolerated, whereas 40.0 mg was toxic to the retina. 1, 10, or 100 mg infliximab were also well tolerated. Analysis of the optic nerve axons after 50 days confirmed the safety of 4.0 mg adalimumab and of 100 mg infliximab. For efficacy, 4.0 mg adalimumab subconjunctivally in 0.08 mL provided practically full protection against retinal cell apoptosis 3 days following alkali burn, and infliximab 100 mg only slightly less. At 90 days following burn injury, control optic nerves showed about 50% axon loss as compared to 8% in the adalimumab treatment group. CONCLUSIONS Subconjunctival injection of 4.0 mg adalimumab in rabbits shows no eye toxicity and provides excellent neuroprotection, both short (3 days) and long-term (90 days). Our total. accumulated data from several of our studies, combined with the present paper, suggest that corneal injuries, including surgery, might benefit from routine administration of anti-TNF-α biologics to reduce inflammation and future secondary glaucoma.
Collapse
Affiliation(s)
- Eleftherios I. Paschalis
- Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, USA
- Schepens Eye Research Institute, Boston Keratoprosthesis Laboratory/Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, USA
- Disruptive Technology Laboratory, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, USA
| | - Chengxin Zhou
- Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, USA
- Schepens Eye Research Institute, Boston Keratoprosthesis Laboratory/Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, USA
- Disruptive Technology Laboratory, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, USA
| | - Jyoti Sharma
- Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, USA
- Schepens Eye Research Institute, Boston Keratoprosthesis Laboratory/Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, USA
- Disruptive Technology Laboratory, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, USA
| | - Thomas H. Dohlman
- Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, USA
- Schepens Eye Research Institute, Boston Keratoprosthesis Laboratory/Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, USA
| | - Sarah Kim
- Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, USA
- Schepens Eye Research Institute, Boston Keratoprosthesis Laboratory/Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, USA
| | - Fengyang Lei
- Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, USA
- Schepens Eye Research Institute, Boston Keratoprosthesis Laboratory/Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, USA
- Disruptive Technology Laboratory, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, USA
| | - James Chodosh
- Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, USA
- Schepens Eye Research Institute, Boston Keratoprosthesis Laboratory/Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, USA
- Disruptive Technology Laboratory, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, USA
| | - Demetrios Vavvas
- Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, USA
- Angiogenesis Laboratory, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, USA
| | - Arto Urtti
- Division of Pharmaceutical Biosciences, University of Helsinki, Finland and School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - George Papaliodis
- Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, USA
| | - Claes H. Dohlman
- Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, USA
- Schepens Eye Research Institute, Boston Keratoprosthesis Laboratory/Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
5
|
Orive Bañuelos A, Fideliz de la Paz M, Feijóo Lera R, Santamaría Carro A, Martínez Grau A, Andollo Victoriano N, Etxebarria Ecenarro J. Transmucosal Boston Keratoprosthesis Type II in a Case of Severe Bilateral Chemical Burn. Cornea 2024; 43:261-264. [PMID: 37906002 DOI: 10.1097/ico.0000000000003426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 10/08/2023] [Indexed: 11/02/2023]
Abstract
PURPOSE The purpose of this report was to describe a new alternative surgical technique combining oral mucosa and the Boston Keratoprosthesis type II (BKPro II) in a patient with severe chemical burn. METHODS We present a case of a 37-year-old man who suffered a severe bilateral chemical burn. Visual acuity was light perception and projection in both eyes. After many surgeries, including eyelid reconstruction, eyelashes electrolysis, and amniotic membrane transplants, he presented a corneal thinning in his right eye that was managed with an autologous oral mucosa graft. Considering the limited function of the eyelids, mild symblepharon, and dry ocular surface, we proposed the implantation of a BKPro II. The patient voiced his desire of avoiding complete tarsorrhaphy for better cosmesis. Osteo-odonto-keratoprosthesis or tibial bone keratoprothesis was also discarded at the patient's express wish. A year and a half later, the oral mucosa was transplanted, and once the ocular surface was stabilized, we lift the mucosa 270 degrees and performed the implantation of the BKPro II covering this device with the mucosa trephined avoiding the complete tarsorrhaphy and theraby modifying the standard procedure. RESULTS After 34 months of follow-up, the patient did not develop any complications, his visual acuity is stable (0.3 decimal), and funduscopic examination showed a pale optic disk, vascular tortuosity, and an epiretinal membrane that remains stable up to this period. CONCLUSIONS Transmucosal BKPro II may be considered as an alternative surgical technique for implant support in BKPro II carriers who want to improve their cosmesis avoiding, avoid complete tarsorrhaphy, and do not want to be exposed to dental or bone surgeries.
Collapse
Affiliation(s)
- Ana Orive Bañuelos
- Department of Ophthalmology, BioCruces Bizkaia Health Research Institute, University Hospital of Cruces, Barakaldo, Spain
| | - María Fideliz de la Paz
- Centro de Oftalmología Barraquer, Universitat Internacional de Catalunya, Barcelona, Spain
- Institut Universitari Barraquer, Universitat Autónoma de Barcelona, Barcelona, Spain; and
| | - Raquel Feijóo Lera
- Department of Ophthalmology, BioCruces Bizkaia Health Research Institute, University Hospital of Cruces, Barakaldo, Spain
| | - Alaitz Santamaría Carro
- Department of Ophthalmology, BioCruces Bizkaia Health Research Institute, University Hospital of Cruces, Barakaldo, Spain
| | - Ainhoa Martínez Grau
- Centro de Oftalmología Barraquer, Universitat Internacional de Catalunya, Barcelona, Spain
- Institut Universitari Barraquer, Universitat Autónoma de Barcelona, Barcelona, Spain; and
| | - Noelia Andollo Victoriano
- Department of Ophthalmology, BioCruces Bizkaia Health Research Institute, University Hospital of Cruces, Barakaldo, Spain
- Department of Cell Biology and Histology, School of Medicine and Nursing, University of the Basque Country (UPV/ EHU), Leioa, Spain
| | - Jaime Etxebarria Ecenarro
- Department of Ophthalmology, BioCruces Bizkaia Health Research Institute, University Hospital of Cruces, Barakaldo, Spain
- Department of Cell Biology and Histology, School of Medicine and Nursing, University of the Basque Country (UPV/ EHU), Leioa, Spain
| |
Collapse
|
6
|
Zhou C, Lei F, Mittermaier M, Ksander B, Dana R, Dohlman CH, Vavvas DG, Chodosh J, Paschalis EI. Opposing Roles of Blood-Borne Monocytes and Tissue-Resident Macrophages in Limbal Stem Cell Damage after Ocular Injury. Cells 2023; 12:2089. [PMID: 37626899 PMCID: PMC10453077 DOI: 10.3390/cells12162089] [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: 07/04/2023] [Revised: 08/14/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023] Open
Abstract
Limbal stem cell (LSC) deficiency is a frequent and severe complication after chemical injury to the eye. Previous studies have assumed this is mediated directly by the caustic agent. Here we show that LSC damage occurs through immune cell mediators, even without direct injury to LSCs. In particular, pH elevation in the anterior chamber (AC) causes acute uveal stress, the release of inflammatory cytokines at the basal limbal tissue, and subsequent LSC damage and death. Peripheral C-C chemokine receptor type 2 positive/CX3C motif chemokine receptor 1 negative (CCR2+ CX3CR1-) monocytes are the key mediators of LSC damage through the upregulation of tumor necrosis factor-alpha (TNF-α) at the limbus. In contrast to peripherally derived monocytes, CX3CR1+ CCR2- tissue-resident macrophages have a protective role, and their depletion prior to injury exacerbates LSC loss and increases LSC vulnerability to TNF-α-mediated apoptosis independently of CCR2+ cell infiltration into the tissue. Consistently, repopulation of the tissue by new resident macrophages not only restores the protective M2-like phenotype of macrophages but also suppresses LSC loss after exposure to inflammatory signals. These findings may have clinical implications in patients with LSC loss after chemical burns or due to other inflammatory conditions.
Collapse
Affiliation(s)
- Chengxin Zhou
- Department of Ophthalmology, Massachusetts Eye and Ear and Schepens Eye Research Institute, Mass General Brigham, Harvard Medical School, Boston, MA 02114, USA; (C.Z.); (F.L.); (M.M.); (B.K.); (R.D.); (C.H.D.); (J.C.)
- Boston Keratoprosthesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear, Mass General Brigham, Harvard Medical School, Boston, MA 02114, USA
- Disruptive Technology Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Fengyang Lei
- Department of Ophthalmology, Massachusetts Eye and Ear and Schepens Eye Research Institute, Mass General Brigham, Harvard Medical School, Boston, MA 02114, USA; (C.Z.); (F.L.); (M.M.); (B.K.); (R.D.); (C.H.D.); (J.C.)
- Boston Keratoprosthesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear, Mass General Brigham, Harvard Medical School, Boston, MA 02114, USA
- Disruptive Technology Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Mirja Mittermaier
- Department of Ophthalmology, Massachusetts Eye and Ear and Schepens Eye Research Institute, Mass General Brigham, Harvard Medical School, Boston, MA 02114, USA; (C.Z.); (F.L.); (M.M.); (B.K.); (R.D.); (C.H.D.); (J.C.)
| | - Bruce Ksander
- Department of Ophthalmology, Massachusetts Eye and Ear and Schepens Eye Research Institute, Mass General Brigham, Harvard Medical School, Boston, MA 02114, USA; (C.Z.); (F.L.); (M.M.); (B.K.); (R.D.); (C.H.D.); (J.C.)
| | - Reza Dana
- Department of Ophthalmology, Massachusetts Eye and Ear and Schepens Eye Research Institute, Mass General Brigham, Harvard Medical School, Boston, MA 02114, USA; (C.Z.); (F.L.); (M.M.); (B.K.); (R.D.); (C.H.D.); (J.C.)
| | - Claes H. Dohlman
- Department of Ophthalmology, Massachusetts Eye and Ear and Schepens Eye Research Institute, Mass General Brigham, Harvard Medical School, Boston, MA 02114, USA; (C.Z.); (F.L.); (M.M.); (B.K.); (R.D.); (C.H.D.); (J.C.)
- Boston Keratoprosthesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear, Mass General Brigham, Harvard Medical School, Boston, MA 02114, USA
| | - Demetrios G. Vavvas
- Retina Service, Department of Ophthalmology, Massachusetts Eye and Ear, Mass General Brigham, Harvard Medical School, Boston, MA 02114, USA;
| | - James Chodosh
- Department of Ophthalmology, Massachusetts Eye and Ear and Schepens Eye Research Institute, Mass General Brigham, Harvard Medical School, Boston, MA 02114, USA; (C.Z.); (F.L.); (M.M.); (B.K.); (R.D.); (C.H.D.); (J.C.)
- Boston Keratoprosthesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear, Mass General Brigham, Harvard Medical School, Boston, MA 02114, USA
- Disruptive Technology Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
- Department of Ophthalmology and Visual Sciences, University of New Mexico School of Medicine, Albuquerque, NM 87108, USA
| | - Eleftherios I. Paschalis
- Department of Ophthalmology, Massachusetts Eye and Ear and Schepens Eye Research Institute, Mass General Brigham, Harvard Medical School, Boston, MA 02114, USA; (C.Z.); (F.L.); (M.M.); (B.K.); (R.D.); (C.H.D.); (J.C.)
- Boston Keratoprosthesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear, Mass General Brigham, Harvard Medical School, Boston, MA 02114, USA
- Disruptive Technology Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
7
|
Zhou C, Lei F, Sharma J, Hui PC, Wolkow N, Dohlman CH, Vavvas DG, Chodosh J, Paschalis EI. Sustained Inhibition of VEGF and TNF-α Achieves Multi-Ocular Protection and Prevents Formation of Blood Vessels after Severe Ocular Trauma. Pharmaceutics 2023; 15:2059. [PMID: 37631272 PMCID: PMC10458495 DOI: 10.3390/pharmaceutics15082059] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 07/25/2023] [Accepted: 07/27/2023] [Indexed: 08/27/2023] Open
Abstract
PURPOSE This study aimed to develop a clinically feasible and practical therapy for multi-ocular protection following ocular injury by using a thermosensitive drug delivery system (DDS) for sustained delivery of TNF-α and VEGF inhibitors to the eye. METHODS A thermosensitive, biodegradable hydrogel DDS (PLGA-PEG-PLGA triblock polymer) loaded with 0.7 mg of adalimumab and 1.4 mg of aflibercept was injected subconjunctivally into Dutch-belted pigmented rabbits after corneal alkali injury. Control rabbits received 2 mg of IgG-loaded DDS or 1.4 mg of aflibercept-loaded DDS. Animals were followed for 3 months and assessed for tolerability and prevention of corneal neovascularization (NV), improvement of corneal re-epithelialization, inhibition of retinal ganglion cell (RGC) and optic nerve axon loss, and inhibition of immune cell infiltration into the cornea. Drug-release kinetics was assessed in vivo using an aqueous humor protein analysis. RESULTS A single subconjunctival administration of dual anti-TNF-α/anti-VEGF DDS achieved a sustained 3-month delivery of antibodies to the anterior chamber, iris, ciliary body, and retina. Administration after corneal alkali burn suppressed CD45+ immune cell infiltration into the cornea, completely inhibited cornea NV for 3 months, accelerated corneal re-epithelialization and wound healing, and prevented RGC and optic nerve axon loss at 3 months. In contrast, anti-VEGF alone or IgG DDS treatment led to persistent corneal epithelial defect (combined: <1%; anti-VEGF: 15%; IgG: 10%, of cornea area), increased infiltration of CD45+ immune cells into the cornea (combined: 28 ± 20; anti-VEGF: 730 ± 178; anti-IgG: 360 ± 186, cells/section), and significant loss of RGCs (combined: 2.7%; anti-VEGF: 63%; IgG: 45%) and optic nerve axons at 3 months. The aqueous humor protein analysis showed first-order release kinetics without adverse effects at the injection site. CONCLUSIONS Concomitant inhibition of TNF-α and VEGF prevents corneal neovascularization and ameliorates subsequent irreversible damage to the retina and optic nerve after severe ocular injury. A single subconjunctival administration of this therapy, using a biodegradable, slow-release thermosensitive DDS, achieved the sustained elution of therapeutic levels of antibodies to all ocular tissues for 3 months. This therapeutic approach has the potential to dramatically improve the outcomes of severe ocular injuries in patients and improve the therapeutic outcomes in patients with retinal vascular diseases.
Collapse
Affiliation(s)
- Chengxin Zhou
- Department of Ophthalmology, Harvard Medical School, Boston, MA 02115, USA; (C.Z.); (F.L.); (P.-C.H.); (N.W.); (C.H.D.); (D.G.V.)
- Boston Keratoprosthesis Laboratory, Massachusetts Eye and Ear, Boston, MA 02114, USA
| | - Fengyang Lei
- Department of Ophthalmology, Harvard Medical School, Boston, MA 02115, USA; (C.Z.); (F.L.); (P.-C.H.); (N.W.); (C.H.D.); (D.G.V.)
- Boston Keratoprosthesis Laboratory, Massachusetts Eye and Ear, Boston, MA 02114, USA
| | - Jyoti Sharma
- Department of Ophthalmology, Harvard Medical School, Boston, MA 02115, USA; (C.Z.); (F.L.); (P.-C.H.); (N.W.); (C.H.D.); (D.G.V.)
- Boston Keratoprosthesis Laboratory, Massachusetts Eye and Ear, Boston, MA 02114, USA
| | - Pui-Chuen Hui
- Department of Ophthalmology, Harvard Medical School, Boston, MA 02115, USA; (C.Z.); (F.L.); (P.-C.H.); (N.W.); (C.H.D.); (D.G.V.)
- Boston Keratoprosthesis Laboratory, Massachusetts Eye and Ear, Boston, MA 02114, USA
| | - Natalie Wolkow
- Department of Ophthalmology, Harvard Medical School, Boston, MA 02115, USA; (C.Z.); (F.L.); (P.-C.H.); (N.W.); (C.H.D.); (D.G.V.)
- David G. Cogan Laboratory of Eye Pathology and Ophthalmic Plastic Surgery Service, Massachusetts Eye and Ear, Boston, MA 02114, USA
| | - Claes H. Dohlman
- Department of Ophthalmology, Harvard Medical School, Boston, MA 02115, USA; (C.Z.); (F.L.); (P.-C.H.); (N.W.); (C.H.D.); (D.G.V.)
- Boston Keratoprosthesis Laboratory, Massachusetts Eye and Ear, Boston, MA 02114, USA
| | - Demetrios G. Vavvas
- Department of Ophthalmology, Harvard Medical School, Boston, MA 02115, USA; (C.Z.); (F.L.); (P.-C.H.); (N.W.); (C.H.D.); (D.G.V.)
- Angiogenesis Laboratory, Massachusetts Eye and Ear, Boston, MA 02114, USA
| | - James Chodosh
- Department of Ophthalmology, Harvard Medical School, Boston, MA 02115, USA; (C.Z.); (F.L.); (P.-C.H.); (N.W.); (C.H.D.); (D.G.V.)
- Boston Keratoprosthesis Laboratory, Massachusetts Eye and Ear, Boston, MA 02114, USA
- Disruptive Technology Laboratory, Massachusetts Eye and Ear, Boston, MA 02114, USA
- Department of Ophthalmology and Visual Sciences, University of New Mexico School of Medicine, Albuquerque, NM 87108, USA
| | - Eleftherios I. Paschalis
- Department of Ophthalmology, Harvard Medical School, Boston, MA 02115, USA; (C.Z.); (F.L.); (P.-C.H.); (N.W.); (C.H.D.); (D.G.V.)
- Boston Keratoprosthesis Laboratory, Massachusetts Eye and Ear, Boston, MA 02114, USA
- Disruptive Technology Laboratory, Massachusetts Eye and Ear, Boston, MA 02114, USA
| |
Collapse
|
8
|
Asuku M, Shupp JW. Burn wound conversion: clinical implications for the treatment of severe burns. J Wound Care 2023; 32:S11-S20. [PMID: 37121662 DOI: 10.12968/jowc.2023.32.sup5.s11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
The identification of novel treatments for severe burn wounds relies on accurate clinical assessments of the extent of injury. However, evaluation of burn wound depth can be challenging due to the tendency for burn wounds to progress over time in a little-understood process known as 'burn wound conversion'. Local factors affecting the burn wound, such as inflammation, oxidative stress-induced tissue damage, vasostasis and bacterial infections, lead to increased cell death by apoptosis or oncosis, while systemic events may promote burn wound conversion. Acute shock, metabolic derangements, age or immunomodulation can modify cytokine secretion, lower immune responses, decrease blood flow or cause bacterial infection at the burn wound site. Therefore, therapeutic approaches targeting specific mechanisms that reduce cell death, improve wound reperfusion and promote tissue regrowth should favourably enhance burn wound healing, and long-term functional and aesthetic outcomes. Our current understanding of these mechanisms mostly comes from animal studies, underscoring the need for extensive research in humans. A streamlined approach would be to investigate the parallels in other disease states that exhibit ischaemia and potential reperfusion, such as ischaemic stroke and myocardial infarction. Moreover, in view of the limited knowledge available on the subject, the need exists for further clinical research into burn wound conversion and novel target pathways to ameliorate its effects. This review describes events that affect the viability of cells at the burn wound site resulting in burn wound conversion, and identifies potential targets for clinical interventions that may diminish burn wound conversion.
Collapse
Affiliation(s)
| | - Jeffrey W Shupp
- Department of Surgery, Biochemistry and Molecular & Cellular Biology, Georgetown University School of Medicine, MedStar Washington Hospital Center, Washington, DC, US
| |
Collapse
|
9
|
Özkaya D, Karaca U, Usta Sofu G, Savran M, Özgöçmen M, Ertuğrul A. Effect of adalimumab on experimental corneal neovascularization model. Int Ophthalmol 2023; 43:2119-2128. [PMID: 37012439 DOI: 10.1007/s10792-023-02695-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 03/12/2023] [Indexed: 04/05/2023]
Abstract
PURPOSE To evaluate the efficacy of adalimumab (ADA) on inhibition of experimental corneal neovascularization (CNV) and compare the outcomes with bevacizumab (BEVA). METHODS Twenty-four female Winstar rats (48 eyes) were used. Silver/Potassium Nitrate sticks were used for creating CNV. Forty-eight eyes of the rats were separated into 6 groups. The eyes which only NaCl was injected subconjunctivally (SC) formed Group-1. The eyes which CNV was created and NaCl, BEVA (2.5 mg/0.05 mL), ADA (2.5 mg/0.05 mL), respectively, were injected SC formed group-2, 3 and 4. The eyes which only BEVA and ADA, respectively, were injected SC formed group-5 and 6. Five days later the animals were sacrificed. Hematoxylin and eosin staining, Masson trichrome staining, Vascular endothelial growth factor (VEGF), and Platelet-derived growth factor (PDGF) antibodies were performed. RESULTS Histochemical results showed that there was no histopathological finding in group-1, 5, and 6. Collagen fiber irregularity was observed in group-2 and there was a significant improvement in collagen fiber irregularity in group-3 and 4. Collagen fiber proliferation was higher in group-2 than in group-3 and 4. VEGF and PDGF stainings were not observed in group-1, 5, and 6. VEGF and PDGF stainings were observed in group-2 and significantly decreased in group-3 and 4 compared to group-2. ADA was found to be superior to BEVA in terms of decreasing VEGF staining. CONCLUSION Both BEVA and ADA were effective in inhibiting CNV. Subconjunctival ADA seems to be more effective than BEVA in terms of inhibiting VEGF expression. Further experimental studies about ADA and BEVA are needed.
Collapse
Affiliation(s)
- Dilek Özkaya
- Department of Ophthalmology, Faculty of Medicine, Süleyman Demirel University, 32260, Isparta, Turkey.
| | - Umut Karaca
- Department of Ophthalmology, Faculty of Medicine, Süleyman Demirel University, 32260, Isparta, Turkey
| | - Gülşah Usta Sofu
- Department of Ophthalmology, Faculty of Medicine, Süleyman Demirel University, 32260, Isparta, Turkey
| | - Mehtap Savran
- Department of Pharmacology, Faculty of Medicine, Süleyman Demirel University, Isparta, Turkey
| | - Meltem Özgöçmen
- Department of Histology, Faculty of Medicine, Süleyman Demirel University, Isparta, Turkey
| | - Alper Ertuğrul
- Department of Ophthalmology, Faculty of Medicine, Süleyman Demirel University, 32260, Isparta, Turkey
| |
Collapse
|
10
|
Bai Y, Jiao X, Hu J, Xue W, Zhou Z, Wang W. WTAP promotes macrophage recruitment and increases VEGF secretion via N6-methyladenosine modification in corneal neovascularization. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166708. [PMID: 37019244 DOI: 10.1016/j.bbadis.2023.166708] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 03/03/2023] [Accepted: 03/28/2023] [Indexed: 04/05/2023]
Abstract
BACKGROUND Corneal neovascularization (CNV) can be caused by chemical burns. Macrophages are involved in angiogenesis and lymphangiogenesis during CNV. The aim of this study was to investigate whether Wilms' tumor 1-associated protein (WTAP) is involved in macrophage recruitment and VEGF secretion via N6-methyladenosine (m6A) modification. METHODS A CNV mouse model was established by corneal alkali burn. Tumor necrosis factor alpha (TNF-α) was used to stimulate vascular endothelial cells. m6A immunoprecipitation qPCR was used to determine the enrichment of m6A levels in mRNAs. The H3K9me3 enrichment in the promoter region of CC motif chemokine ligand 2 (CCL2) was detected by chromatin immunoprecipitation assay. The WTAP inhibition in vivo was performed using the adeno-associated virus. RESULTS In the alkali burn corneal tissues, angiogenesis and lymphangiogenesis were promoted as CD31 and LYVE-1 expressions were elevated, and the number of macrophages as well as WTAP expression were increased. Under the TNF-α stimulation, WTAP promoted the recruitment of endothelial cells to macrophages by promoting CCL2 secretion. Mechanistically, WTAP affected the enrichment of H3K9me3 at the CCL2 promoter by regulating the m6A level of SUV39H1 mRNA. The in vivo experiment showed that VEGFA/C/D secretion of macrophages was reduced after WTAP interference. Mechanistically, WTAP regulated the translational efficiency of HIF-1α via m6A modification. CONCLUSION WTAP affected macrophage recruitment to endothelial cells via regulation of H3K9me3-mediated CCL2 transcription. WTAP also affected macrophage secretion of VEGFA/C/D via m6A-mediated translation regulation of HIF-1α. Both pathways were involved in the WTAP regulation of angiogenesis and lymphangiogenesis during CNV.
Collapse
|
11
|
Volkova MV, Shen N, Polyanskaya A, Qi X, Boyarintsev VV, Kovaleva EV, Trofimenko AV, Filkov GI, Mezentsev AV, Rybalkin SP, Durymanov MO. Tissue-Oxygen-Adaptation of Bone Marrow-Derived Mesenchymal Stromal Cells Enhances Their Immunomodulatory and Pro-Angiogenic Capacity, Resulting in Accelerated Healing of Chemical Burns. Int J Mol Sci 2023; 24:4102. [PMID: 36835513 PMCID: PMC9963537 DOI: 10.3390/ijms24044102] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/08/2023] [Accepted: 02/13/2023] [Indexed: 02/22/2023] Open
Abstract
Transplantation of mesenchymal stromal cells (MSCs) provides a powerful tool for the management of multiple tissue injuries. However, poor survival of exogenous cells at the site of injury is a major complication that impairs MSC therapeutic efficacy. It has been found that tissue-oxygen adaptation or hypoxic pre-conditioning of MSCs could improve the healing process. Here, we investigated the effect of low oxygen tension on the regenerative potential of bone-marrow MSCs. It turned out that incubation of MSCs under a 5% oxygen atmosphere resulted in increased proliferative activity and enhanced expression of multiple cytokines and growth factors. Conditioned growth medium from low-oxygen-adapted MSCs modulated the pro-inflammatory activity of LPS-activated macrophages and stimulated tube formation by endotheliocytes to a much higher extent than conditioned medium from MSCs cultured in a 21% oxygen atmosphere. Moreover, we examined the regenerative potential of tissue-oxygen-adapted and normoxic MSCs in an alkali-burn injury model on mice. It has been revealed that tissue-oxygen adaptation of MSCs accelerated wound re-epithelialization and improved the tissue histology of the healed wounds in comparison with normoxic MSC-treated and non-treated wounds. Overall, this study suggests that MSC adaptation to 'physiological hypoxia' could be a promising approach for facilitating skin injuries, including chemical burns.
Collapse
Affiliation(s)
- Marina V. Volkova
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, National Research University, Dolgoprudny 141701, Russia
| | - Ningfei Shen
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, National Research University, Dolgoprudny 141701, Russia
| | - Anna Polyanskaya
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, National Research University, Dolgoprudny 141701, Russia
| | - Xiaoli Qi
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, National Research University, Dolgoprudny 141701, Russia
| | - Valery V. Boyarintsev
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, National Research University, Dolgoprudny 141701, Russia
| | - Elena V. Kovaleva
- Department of Pathomorphology and Reproductive Toxicology, Research Center of Toxicology and Hygienic Regulation of Biopreparations, NRC Institute of Immunology FMBA of Russia, Ul. Lenina 102A, Serpukhov 142253, Russia
| | - Alexander V. Trofimenko
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, National Research University, Dolgoprudny 141701, Russia
| | - Gleb I. Filkov
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, National Research University, Dolgoprudny 141701, Russia
| | - Alexandre V. Mezentsev
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, National Research University, Dolgoprudny 141701, Russia
| | - Sergey P. Rybalkin
- Department of Pathomorphology and Reproductive Toxicology, Research Center of Toxicology and Hygienic Regulation of Biopreparations, NRC Institute of Immunology FMBA of Russia, Ul. Lenina 102A, Serpukhov 142253, Russia
| | - Mikhail O. Durymanov
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, National Research University, Dolgoprudny 141701, Russia
| |
Collapse
|
12
|
Effect of Tauroursodeoxycholic Acid on Inflammation after Ocular Alkali Burn. Int J Mol Sci 2022; 23:ijms231911717. [PMID: 36233018 PMCID: PMC9570278 DOI: 10.3390/ijms231911717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/26/2022] [Accepted: 09/27/2022] [Indexed: 11/25/2022] Open
Abstract
Inflammation is the main cause of corneal and retinal damage in an ocular alkali burn (OAB). The aim of this study was to investigate the effect of tauroursodeoxycholic acid (TUDCA) on ocular inflammation in a mouse model of an OAB. An OAB was induced in C57BL/6j mouse corneas by using 1 M NaOH. TUDCA (400 mg/kg) or PBS was injected intraperitoneally (IP) once a day for 3 days prior to establishing the OAB model. A single injection of Infliximab (6.25 mg/kg) was administered IP immediately after the OAB. The TUDCA suppressed the infiltration of the CD45-positive cells and decreased the mRNA and protein levels of the upregulated TNF-α and IL-1β in the cornea and retina of the OAB. Furthermore, the TUDCA treatment inhibited the retinal glial activation after an OAB. The TUDCA treatment not only ameliorated CNV and promoted corneal re-epithelization but also attenuated the RGC apoptosis and preserved the retinal structure after the OAB. Finally, the TUDCA reduced the expression of the endoplasmic reticulum (ER) stress molecules, IRE1, GRP78 and CHOP, in the retinal tissues of the OAB mice. The present study demonstrated that the TUDCA inhibits ocular inflammation and protects the cornea and retina from injury in an OAB mouse model. These results provide a potential therapeutic intervention for the treatment of an OAB.
Collapse
|
13
|
Saini C, Chen TC, Young LH, Vavvas DG, Vangel M, Papaliodis GN, Mukai S, Turalba AV, Rhee DJ, Wu DM, Eliott D, Miller JB, Song BJ, Shen LQ, Pasquale LR, Chodosh J. Restoration of Vision in Severe, Cicatricial, Ocular Surface Disease with the Boston Keratoprosthesis Type II. Am J Ophthalmol 2022; 243:42-54. [PMID: 35850253 DOI: 10.1016/j.ajo.2022.06.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 06/27/2022] [Accepted: 06/30/2022] [Indexed: 11/01/2022]
Abstract
PURPOSE To assess clinical outcomes of patients with severe, cicatricial ocular surface disease (OSD) implanted with the currently marketed design of the Boston keratoprosthesis type II (BK2). DESIGN Retrospective cohort study. METHODS Records of consecutive patients undergoing BK2 implantation from June 2009 to March 2021 were assessed for postoperative visual acuity, postoperative complications, device replacement, and additional surgeries. RESULTS Fifty-six eyes of 53 patients with a mean follow-up of 45.8 months (range: 0.2-134.7 months) were included. Stevens-Johnson syndrome/toxic epidermal necrolysis was the most common indication (49.1%), followed by mucous membrane pemphigoid (39.6%) and other OSD (11.3%). Visual acuity improved from LogMAR 2.2±0.5 preoperatively to 1.5±1.2 at final follow-up. Fifty of 56 eyes saw ≥20/200 at some point postoperatively. Of the eyes with a follow-up of more than 5 years, 50.0% retained a visual acuity of ≥20/200 at their final follow-up. The most common complications, over the entire postoperative course (mean ∼4 years), were de-novo or worsening glaucoma (41.1%), choroidal effusions (30.3%), retinal detachment (25.0%) and end-stage glaucoma (25.0%). In a univariate analysis, patients who experienced irreversible loss of ≥20/200 visual acuity were more likely to have been previously implanted with an older design of BK2, less likely to be on preoperative systemic immunosuppressive therapy, and less likely to have undergone concurrent glaucoma tube implantation, compared to patients who retained ≥20/200 acuity (p<0.04 for all). CONCLUSIONS Advances in device design and postoperative care have made implantation of BK2 a viable option for corneal blindness in the setting of severe cicatricial OSD.
Collapse
Affiliation(s)
- Chhavi Saini
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Teresa C Chen
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Lucy H Young
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Demetrios G Vavvas
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Mark Vangel
- Biostatistics Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - George N Papaliodis
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Shizuo Mukai
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Angela V Turalba
- Ophthalmology and Visual Services, Atrius Health, Boston, Massachusetts, USA
| | - Douglas J Rhee
- University Hospitals, Case Western Reserve University, Cleveland, Ohio, USA
| | - David M Wu
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Dean Eliott
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - John B Miller
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Brian J Song
- Department of Ophthalmology, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Lucy Q Shen
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Louis R Pasquale
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, USA
| | - James Chodosh
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
14
|
Abstract
Millions of people worldwide are bilaterally blind due to corneal diseases including infectious etiologies, trauma, and chemical injuries. While corneal transplantation can successfully restore sight in many, corneal graft survival decreases in eyes with chronic inflammation and corneal vascularization. Additionally, the availability of donor cornea material can be limited, especially in underdeveloped countries where corneal blindness may also be highly prevalent. Development of methods to create and implant an artificial cornea (keratoprosthesis)may be the only option for patients whose eye disease is not suitable for corneal transplantation or who live in regions where corneal transplantation is not possible. The Boston keratoprosthesis (B-KPro) is the most commonly implanted keratoprosthesis worldwide, having restored vision in thousands of patients. This article describes the initial design of the B-KPro and the modifications that have been made over many years. Additionally, some of the complications of surgical implantation and long-term care challenges, particularly complicating inflammation and glaucoma, are discussed. Expected final online publication date for the Annual Review of Vision Science, Volume 8 is September 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Claes Dohlman
- Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, USA;
| |
Collapse
|
15
|
Lambuk L, Ahmad S, Sadikan MZ, Nordin NA, Kadir R, Nasir NAA, Chen X, Boer J, Plebanski M, Mohamud R. Targeting Differential Roles of Tumor Necrosis Factor Receptors as a Therapeutic Strategy for Glaucoma. Front Immunol 2022; 13:857812. [PMID: 35651608 PMCID: PMC9149562 DOI: 10.3389/fimmu.2022.857812] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 04/19/2022] [Indexed: 11/13/2022] Open
Abstract
Glaucoma is an irreversible sight-threatening disorder primarily due to elevated intraocular pressure (IOP), leading to retinal ganglion cell (RGC) death by apoptosis with subsequent loss of optic nerve fibers. A considerable amount of empirical evidence has shown the significant association between tumor necrosis factor cytokine (TNF; TNFα) and glaucoma; however, the exact role of TNF in glaucoma progression remains unclear. Total inhibition of TNF against its receptors can cause side effects, although this is not the case when using selective inhibitors. In addition, TNF exerts its antithetic roles via stimulation of two receptors, TNF receptor I (TNFR1) and TNF receptor II (TNFR2). The pro-inflammatory responses and proapoptotic signaling pathways predominantly mediated through TNFR1, while neuroprotective and anti-apoptotic signals induced by TNFR2. In this review, we attempt to discuss the involvement of TNF receptors (TNFRs) and their signaling pathway in ocular tissues with focus on RGC and glial cells in glaucoma. This review also outlines the potential application TNFRs agonist and/or antagonists as neuroprotective strategy from a therapeutic standpoint. Taken together, a better understanding of the function of TNFRs may lead to the development of a treatment for glaucoma.
Collapse
Affiliation(s)
- Lidawani Lambuk
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu, Malaysia
| | - Suhana Ahmad
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu, Malaysia
| | - Muhammad Zulfiqah Sadikan
- Centre for Neuroscience Research (NeuRon), Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh, Malaysia
| | - Nor Asyikin Nordin
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu, Malaysia
| | - Ramlah Kadir
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu, Malaysia
| | - Nurul Alimah Abdul Nasir
- Centre for Neuroscience Research (NeuRon), Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh, Malaysia
| | - Xin Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China
| | - Jennifer Boer
- School of Health and Biomedical Sciences, Royal Melbourne Institute Technology (RMIT) University, Bundoora, VIC, Australia
| | - Magdalena Plebanski
- School of Health and Biomedical Sciences, Royal Melbourne Institute Technology (RMIT) University, Bundoora, VIC, Australia
| | - Rohimah Mohamud
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu, Malaysia
| |
Collapse
|
16
|
Huang Y, Yuan M, Duan F, Yang Y, Lou B, Lin X. Inhibition of endoplasmic reticulum stress by 4-phenylbutyrate alleviates retinal inflammation and the apoptosis of retinal ganglion cells after ocular alkali burn in mice. Inflamm Res 2022; 71:577-590. [PMID: 35415762 DOI: 10.1007/s00011-022-01565-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 02/28/2022] [Accepted: 03/19/2022] [Indexed: 12/29/2022] Open
Abstract
OBJECTIVE Retinal ganglion cell (RGC) apoptosis is one of the most severe complications that causes permanent visual impairment following ocular alkali burn (OAB). Currently, very few treatment options exist for this condition. This study was conducted to determine the effect of 4-phenylbutyric acid (4-PBA) on endoplasmic reticulum (ER) stress after OAB using a well-established OAB mouse model. METHODS Ocular alkali burn was induced in C57BL/6 mouse corneas using 1 M NaOH. 4-PBA (10 mg/kg; 250 μL per injection) or saline (250 μL per injection) was injected intraperitoneally once per day for 3 days before the establishment of the OAB model. The apoptosis of retinal ganglion cells (RGCs) was assessed by terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) assay, and the histological damage was examined by hematoxylin and eosin and immunofluorescence assay on retinal flat mounts. The key inflammatory response and the expression of ER stress-related markers in the retinal tissues were assessed by real-time PCR, western blotting and histologic analyses. RESULTS 4-PBA significantly alleviated the apoptosis of RGCs and prevented the structural damage of the retina, as determined by the evaluation of RGC density and retinal thickness. Inhibition of ER stress by 4-PBA decreased the expression of vital proinflammatory cytokines, tumor necrosis factor alpha, and interleukin-1 beta; and suppressed the activation of retinal microglial cells and nuclear factor-kappa B (NF-κB). 4-PBA reduced the expression of the ER stress molecules, glucose-regulated protein 78, activated transcription factor 6, inositol-requiring enzyme-1 (IRE1), X-box-binding protein 1 splicing, and CCAAT/enhancer-binding protein homologous protein, in the retinal tissues and RGCs of OAB mice. CONCLUSIONS The present study demonstrated that the inhibition of ER stress by 4-PBA alleviates the inflammatory response via the IRE1/NF-κB signaling pathway and protects the retina and RGCs from injury in an OAB mouse model. Such findings further suggest that 4-PBA might have potential therapeutic implications for OAB treatment.
Collapse
Affiliation(s)
- Yanqiao Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Miner Yuan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Fang Duan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Yao Yang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Bingsheng Lou
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Xiaofeng Lin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, 510060, China.
| |
Collapse
|
17
|
Intermediate-term Visual Outcomes and Complications of Type 1 Boston Keratoprosthesis With and Without Glaucoma Surgery. Cornea 2022; 41:950-957. [PMID: 35184127 DOI: 10.1097/ico.0000000000002962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Accepted: 11/12/2021] [Indexed: 11/25/2022]
Abstract
PURPOSE Glaucoma is a cause of comorbidity in patients receiving the Boston keratoprosthesis (KPro). The aim of this study was to report the outcomes of the Boston KPro with or without glaucoma surgery. METHODS This was a retrospective single-center cohort study. Patients who underwent Boston KPro from March 2009 to February 2019 were included. One eye per patient (the first surgery) was included in this study. Patients were classified into 2 groups: KPro only (group 1) and KPro with any form of glaucoma procedure (group 2). Main outcome measures were Best-corrected visual acuity (BCVA), functional success (BCVA 20/200 or better), anatomical success (retention of KPro at the last follow-up), and complications. RESULTS Seventy-one eyes were included: 27 eyes (38%) in group 1 and 44 (62%) in group 2. There was no statistically significant difference in BCVA between groups 1 and 2 at each time point. Of the eyes in group 1, 11% lost light perception vision and 4.5% in group 2 (P = 0.293). There was no difference in anatomical success with 70% in group 1 and 77% in group 2 (P = 0.703) at the last follow-up, with a median failure time of 18 months. The functional success was 48% for group 1 and 50% for group 2 (P = 0.541). CONCLUSIONS Eyes undergoing KPro with glaucoma surgery before or at the same time carry a similar functional and anatomical success to eyes without glaucoma surgery.
Collapse
|
18
|
Khoshdel AR, Emami Aleagha O, Shahriary A, Aghamollaei H, Najjar Asiabani F. Topical Effects of N-Acetyl Cysteine and Doxycycline on Inflammatory and Angiogenic Factors in the Rat Model of Alkali-Burned Cornea. J Interferon Cytokine Res 2022; 42:82-89. [PMID: 35029525 DOI: 10.1089/jir.2021.0150] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The aim of this study was to analyze the single and combined effects of N-acetyl cysteine (NAC) and doxycycline (Dox) on the inflammatory and angiogenic factors in the rat model of alkali-burned cornea. Rats were treated with a single and combined 0.5% NAC and 12.5 μg/mL Dox eye drops and evaluated on days 3, 7, and 28. In the corneas of various groups, the activity of Catalase (CAT), superoxide dismutase (SOD), and glutathione peroxidase (GPx) enzymes was assessed. The expression of inflammatory factors (TNF-α, Rel-a, and CXCL-1) and angiogenic factors (VEGF-a, MMP2, and MMP9) was measured using real-time polymerase chain reaction. The antioxidant enzyme activities decreased substantially 3 days after injury with sodium hydroxide (NaOH). NAC and combined NAC+ Dox topical treatments increased the SOD enzyme activity on day 28 (P < 0.05). The expression of TNF-α and Rel-a genes following single and combined treatment of NAC and Dox decreased significantly on days 7 and 28 (P < 0.05). The mRNA level of angiogenic factors and corneal neovascularization (CNV) level declined in NaOH-injured rats treated with Dox (P < 0.05). The topical treatment of Dox could attenuate inflammation and CNV complications. However, NAC treatment may not reduce the expression of angiogenic genes.
Collapse
Affiliation(s)
- Ali Reza Khoshdel
- Modern Epidemiology Research Center, AJA University of Medical Sciences, Tehran, Iran
| | - Omid Emami Aleagha
- Modern Epidemiology Research Center, AJA University of Medical Sciences, Tehran, Iran
| | - Alireza Shahriary
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Hossein Aghamollaei
- Applied Virology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
19
|
Mirarab Razi H, Mosleh N, Shomali T, Tavangar N, Namazi F. Deterioration of wound healing and intense suppression of MMP-9 mRNA expression after short-term administration of different topical glucocorticoids or NSAIDs in an avian model of corneal lesions. IRANIAN JOURNAL OF VETERINARY RESEARCH 2021; 22:188-194. [PMID: 34777518 DOI: 10.22099/ijvr.2021.39864.5783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 06/04/2021] [Accepted: 06/10/2021] [Indexed: 09/30/2022]
Abstract
Background Corneal lesions are considered among important ophthalmic conditions in avian patients. Short-term outcome of using anti-inflammatory agents in corneal lesions of birds are not well-described. Aims The study evaluates effects of different anti-inflammatory agents on healing of alkali burn-induced corneal lesions in layer hens as an avian model. Methods Adult layers were randomly allocated into 7 groups (n=15) as follows: 1. Negative (normal) control (NC), and 2. Positive control (PC) with an experimentally induced-corneal lesion, 3-7. Birds with corneal lesions that were treated with dexamethasone, fluorometholone, prednisolone, ketorolac, or diclofenac eye-drops every 6 hours (QID) for 5 consecutive days. Results At the end of the experiment, proper healing was observed in PC group based on lesion area, while treated groups showed statistically larger lesion sizes as compared to PC birds (P<0.05). Although no significant difference was observed among groups, birds treated with ketorolac, diclofenac or fluorometholone had higher histopathological scores for most of the assayed parameters than other groups. Levels of tumor necrosis factor-α (TNF-α) and vascular endothelial growth factor (VEGF) in corneal tissue of different groups were statistically the same. The mRNA expression of matrix metalloproteinase-9 (MMP-9) was increased 2.5 folds in PC group as compared to NC birds. However, birds treated with anti-inflammatory agents showed no detectable expression of MMP-9 mRNA. Conclusion Five days of topical administration of non-steroidal anti-inflammatory agents (NSAIDs) or glucocorticoids (GCs) is associated with suppression of MMP-9 mRNA expression in corneal tissue and detrimental effects on wound healing in layers with alkali burn-induced corneal ulcers.
Collapse
Affiliation(s)
- H Mirarab Razi
- Resident of Avian Medicine, Department of Clinical Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - N Mosleh
- Department of Clinical Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - T Shomali
- Department of Basic Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - N Tavangar
- Resident of Avian Medicine, Department of Clinical Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - F Namazi
- Department of Pathobiology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| |
Collapse
|
20
|
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
|
21
|
Wang L, He X, Wang Q, Wu T, Liu A, Huang Y. Long-term outcomes of the MICOF keratoprosthesis surgery. Ocul Surf 2021; 21:178-185. [PMID: 34118425 DOI: 10.1016/j.jtos.2021.06.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 06/02/2021] [Accepted: 06/03/2021] [Indexed: 01/17/2023]
Abstract
PURPOSE To evaluate long-term anatomical and functional outcomes of the MICOF keratoprosthesis to treat end-stage corneal blindness. DESIGN Retrospective review of consecutive clinical case series. PARTICIPANTS Between October 2000 and October 2015, at the Department of Ophthalmology of Chinese PLA General Hospital (PLAGH), a total of 132 eyes of 131 patients had undergone a MICOF keratoprosthesis implantation. Of those, 91 eyes of 90 patients were included in this study. METHODS Preoperative information, surgical procedures, and postoperative data were collected for each included eye. MAIN OUTCOME MEASURES Best-corrected visual acuity (BCVA), keratoprosthesis retention, and significant postoperative complications were reported. RESULTS The most common indications for surgery were chemical or thermal burns (68.1%, 62 of 91 eyes) and explosive injury (12.1%, 11 of 91 eyes), followed by Stevens-Johnson Syndrome (10.0%, 9 of 91 eyes), Sjögren's syndrome (4.4%, 4 of 91 eyes), mucous membrane pemphigoid (3.3%, 3 of 91 eyes) and multi-penetrating keratoplasty failure (2.2%, 2 of 91 eyes). The mean follow-up duration was 8.38 ± 3.22 years (range: 5-17.25 years, median: 7.67 years). All patbients had a preoperative visual acuity of hand motions or worse. A MICOF keratoprosthesis significantly improved patients' visual function with bilateral end-stage corneal blindness. Postoperative visual acuity improved to 20/200 or better in 41 eyes (45.1%, of 91 eyes) and to 20/100 or better in 32 eyes (35.2% of 91 eyes) at the last follow-up visit. Preexisting glaucoma was present in 17 (18.7% of 91 eyes). The most common postoperative complications were overgrowth of the surface mucosa (31.9%, 29 of 91 eyes), glaucoma (25.3%, 23 of 91 eyes), retro-prosthetic membrane (15.4%, 14 of 91 eyes), keratoprosthesis device extrusion (15.4%, 14 of 91 eyes), superficial tissue thinning (14.3%, 13 of 91 eyes), endophthalmitis (13.2%, 12 of 91 eyes), titanium frame exposure (13.2%, 12 of 91 eyes), optical cylinder ante-displacement (13.2%, 12 of 91 eyes), cornea melting (7.7%, 7 of 91 eyes), retinal detachment (6.6%, 6 of 91 eyes) and aqueous humour leakage (2.2%, 2 of 91 eyes). 84.6% (77 of 91 eyes) of the eyes retained their initial keratoprosthesis at the latest follow-up. CONCLUSIONS A MICOF keratoprosthesis is a reliable approach to rescue vision in end-stage corneal blinded patients and has better retention than a Boston Kpro TypeⅡ.
Collapse
Affiliation(s)
- Liqiang Wang
- Department of Ophthalmology, Chinese PLA General Hospital, Fuxing Road 28, Beijing, 100853, China.
| | - Xiezhou He
- Department of Ophthalmology, Chinese PLA General Hospital, Fuxing Road 28, Beijing, 100853, China; Medical College, Nankai University, Tianjin, China
| | - Qun Wang
- Department of Ophthalmology, Chinese PLA General Hospital, Fuxing Road 28, Beijing, 100853, China
| | - Tengyun Wu
- Air Force Medical Center of PLA, Beijing, China
| | - Anqi Liu
- Department of Ophthalmology, Chinese PLA General Hospital, Fuxing Road 28, Beijing, 100853, China
| | - Yifei Huang
- Department of Ophthalmology, Chinese PLA General Hospital, Fuxing Road 28, Beijing, 100853, China.
| |
Collapse
|
22
|
Jiang L, He W, Tang F, Tang N, Huang G, Huang W, Wu X, Guan J, Zeng S, Li M, Chen Q, Zhang M, Zhong H, Lan Q, Cui L, Li L, Xu F. Epigenetic Landscape Analysis of the Long Non-Coding RNA and Messenger RNA in a Mouse Model of Corneal Alkali Burns. Invest Ophthalmol Vis Sci 2021; 62:28. [PMID: 33891681 PMCID: PMC8083103 DOI: 10.1167/iovs.62.4.28] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Purpose Corneal alkali burns (CABs) are a common clinical ocular disease, presenting a poor prognosis. Although some long noncoding RNAs (lncRNAs) reportedly play a key role in epigenetic regulation associated with CABs, studies regarding the lncRNA signature in CABs remain rare and elusive. Methods A CAB model was established in C57BL/6J mice and profiling of lncRNA expressions was performed by RNA-Seq. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were conducted to predicate the related pathological pathways and candidate genes. RT-qPCR was used to verify the expression pattern of lncRNAs and related mRNAs, both in vitro and in vivo. Data were statistically analyzed by GraphPad Prism version 6.0. Results In all, 4436 aberrantly expressed lncRNAs were identified in CAB mice when compared with control mice. In the top 13 aberrantly expressed lncRNAs, Bc037156 and 4930511E03Rik were confirmed as the most significantly altered lncRNAs. Pathway analysis revealed that mitogen-activated protein kinase (MAPK) signaling pathway was most enriched. Following 4930511E03Rik siRNA treated, Srgn, IL-1β and Cxcr2 were significant upregulated in corneal epithelial cells, corneal keratocytes, and bone marrow dendritic cells, with NaOH treatment. Moreover, after Bc037156 siRNA treated, expression levels of IL-1β and Srgn were significantly downregulated in the three cell lines. Conclusions Our study suggests that Bc037156 and 4930511E03Rik may be involved in inflammation, immune response, and neovascularization by regulating Srgn, IL-1β, and Cxcr2 expression after CAB. These candidate lncRNAs and mRNAs may be the potential targets for the treatment strategy of the alkali injured cornea.
Collapse
Affiliation(s)
- Li Jiang
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, People's Republic of China
| | - Wenjing He
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, People's Republic of China
| | - Fen Tang
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, People's Republic of China
| | - Ningning Tang
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, People's Republic of China
| | - Guangyi Huang
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, People's Republic of China
| | - Wei Huang
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, People's Republic of China
| | - Xiaonian Wu
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, People's Republic of China
| | - Jianpei Guan
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, People's Republic of China
| | - Siming Zeng
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, People's Republic of China
| | - Min Li
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, People's Republic of China
| | - Qi Chen
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, People's Republic of China
| | - Mingyuan Zhang
- Laboratory Animal Center, Guangxi Medical University, Nanning, China
| | - Haibin Zhong
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, People's Republic of China
| | - Qianqian Lan
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, People's Republic of China
| | - Ling Cui
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, People's Republic of China
| | - Lili Li
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, People's Republic of China
| | - Fan Xu
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, People's Republic of China
| |
Collapse
|
23
|
Maier AKB, Reichhart N, Gonnermann J, Kociok N, Riechardt AI, Gundlach E, Strauß O, Joussen AM. Effects of TNFα receptor TNF-Rp55- or TNF-Rp75- deficiency on corneal neovascularization and lymphangiogenesis in the mouse. PLoS One 2021; 16:e0245143. [PMID: 33835999 PMCID: PMC8034740 DOI: 10.1371/journal.pone.0245143] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 12/22/2020] [Indexed: 02/02/2023] Open
Abstract
Tumor necrosis factor (TNF)α is an inflammatory cytokine likely to be involved in the process of corneal inflammation and neovascularization. In the present study we evaluate the role of the two receptors, TNF-receptor (TNF-R)p55 and TNF-Rp75, in the mouse model of suture-induced corneal neovascularization and lymphangiogenesis. Corneal neovascularization and lymphangiogenesis were induced by three 11-0 intrastromal corneal sutures in wild-type (WT) C57BL/6J mice and TNF-Rp55-deficient (TNF-Rp55d) and TNF-Rp75-deficient (TNF-Rp75d) mice. The mRNA expression of VEGF-A, VEGF-C, Lyve-1 and TNFα and its receptors was quantified by qPCR. The area covered with blood- or lymphatic vessels, respectively, was analyzed by immunohistochemistry of corneal flatmounts. Expression and localization of TNFα and its receptors was assessed by immunohistochemistry of sagittal sections and Western Blot. Both receptors are expressed in the murine cornea and are not differentially regulated by the genetic alteration. Both TNF-Rp55d and TNF-Rp75d mice showed a decrease in vascularized area compared to wild-type mice 14 days after suture treatment. After 21 days there were no differences detectable between the groups. The number of VEGF-A-expressing macrophages did not differ when comparing WT to TNF-Rp55d and TNF-Rp75d. The mRNA expression of lymphangiogenic markers VEGF-C or LYVE-1 does not increase after suture in all 3 groups and lymphangiogenesis showed a delayed effect only for TNF-Rp75d. TNFα mRNA and protein expression increased after suture treatment but showed no difference between the three groups. In the suture-induced mouse model, TNFα and its ligands TNF-Rp55 and TNF-Rp75 do not play a significant role in the pathogenesis of neovascularisation and lymphangiogenesis.
Collapse
MESH Headings
- Animals
- Cornea/metabolism
- Cornea/pathology
- Corneal Neovascularization/genetics
- Corneal Neovascularization/pathology
- Gene Deletion
- Humans
- Lymphangiogenesis
- Mice, Inbred C57BL
- RNA, Messenger/genetics
- Receptors, Tumor Necrosis Factor, Type I/analysis
- Receptors, Tumor Necrosis Factor, Type I/genetics
- Receptors, Tumor Necrosis Factor, Type II/analysis
- Receptors, Tumor Necrosis Factor, Type II/genetics
- Mice
Collapse
Affiliation(s)
- Anna-Karina B. Maier
- Department of Ophthalmology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt- Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Nadine Reichhart
- Department of Ophthalmology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt- Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Johannes Gonnermann
- Department of Ophthalmology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt- Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Norbert Kociok
- Department of Ophthalmology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt- Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Aline I. Riechardt
- Department of Ophthalmology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt- Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Enken Gundlach
- Department of Ophthalmology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt- Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Olaf Strauß
- Department of Ophthalmology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt- Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Antonia M. Joussen
- Department of Ophthalmology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt- Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
24
|
Shahriary A, Sabzevari M, Jadidi K, Yazdani F, Aghamollaei H. The Role of Inflammatory Cytokines in Neovascularization of Chemical Ocular Injury. Ocul Immunol Inflamm 2021; 30:1149-1161. [PMID: 33734925 DOI: 10.1080/09273948.2020.1870148] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Aim: Chemical injuries can potentially lead to the necrosis anterior segment of the eye, and cornea in particular. Inflammatory cytokines are the first factors produced after chemical ocular injuries. Inflammation via promoting the angiogenesis factor tries to implement the wound healing mechanism in the epithelial and stromal layer of the cornea. Methods: Narrative review.Results: In our review, we described the patterns of chemical injuries in the cornea and their molecular mechanisms associated with the expression of inflammatory cytokines. Moreover, the effects of inflammation signals on angiogenesis factors and CNV were explained. Conclusion: The contribution of inflammation and angiogenesis causes de novo formation of blood vessels that is known as the corneal neovascularization (CNV). The new vascularity interrupts cornea clarity and visual acuity. Inflammation also depleted the Limbal stem cells (LSCs) in the limbus causing the failure of normal corneal epithelial healing and conjunctivalization of the cornea.
Collapse
Affiliation(s)
- Alireza Shahriary
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Milad Sabzevari
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Khosrow Jadidi
- Vision Health Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Farshad Yazdani
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Hossein Aghamollaei
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
25
|
Dinç E, Dursun Ö, Yilmaz G, Kurt AH, Ayaz L, Vatansever M, Özer Ö, Yilmaz ŞN. Evaluation of Anti-Inflammatory and Antiapoptotic Effects of Bone Marrow and Adipose-Derived Mesenchymal Stem Cells in Acute Alkaline Corneal Burn. J Ocul Pharmacol Ther 2020; 37:24-34. [PMID: 33275515 DOI: 10.1089/jop.2020.0103] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Purpose: The aim of the present study is to comparatively evaluate the anti-inflammatory and antiapoptotic effects of bone marrow and adipose-derived mesenchymal stem cells (MSCs) applied subconjunctivally after alkaline corneal burn. Methods: Thirty-two rats were divided into 4 groups and included in the study (n = 8). While no intervention was made in the control group, a chemical burn was created by applying 4 μL of NaOH soaked in 6 mm filter paper to the right eye of each subject in the other groups under general anesthesia. While only subconjunctival 0.1 mL phosphate-buffered saline (PBS) was injected to in the group 1, 2 × 106 adipose or bone marrow-derived MSC in 0.1 mL PBS was applied subconjunctivally to the subjects in the remaining groups (Group 2 and 3, respectively). Tissue samples were collected for histological analysis on the third day after the burn. Tissue samples were evaluated light microscopically and immunohistochemically stained for interleukin-1 beta (IL-1β), tumor necrosis factor alpha (TNF-α), caspase-3 (Cas-3), and CD68. Results: The IL-1β and TNF-α staining scores and the number of CD68- and Cas-3-positive stained cells were significantly lower in the groups given bone marrow and adipose-derived MSC compared to the alkaline burn group (P < 0.0001, for all parameters). Epithelial IL-1β and TNF-α staining scores were significantly lower in the bone marrow-derived MSC group compared to the adipose-derived MSC group (P < 0.0001, for all parameters). Conclusions: The presented study shows that both bone-marrow and adipose-derived MSCs support wound healing in the corneal tissue and strongly suppress the inflammation occured in the tissue.
Collapse
Affiliation(s)
- Erdem Dinç
- Department of Ophthalmology and Faculty of Medicine, Mersin University, Mersin, Turkey
| | - Özer Dursun
- Department of Ophthalmology and Faculty of Medicine, Mersin University, Mersin, Turkey
| | - Gülsen Yilmaz
- Department of Histology & Embryology, Faculty of Medicine, Mersin University, Mersin, Turkey
| | - A Hakan Kurt
- Department of Pharmacology, Bolu İzzet Baysal University, Bolu, Turkey
| | - Lokman Ayaz
- Department of Biochemistry, Faculty of Pharmacy, Trakya University, Edirne, Turkey
| | - Mustafa Vatansever
- Department of Ophthalmology and Faculty of Medicine, Mersin University, Mersin, Turkey
| | - Ömer Özer
- Department of Ophthalmology and Faculty of Medicine, Mersin University, Mersin, Turkey
| | - Şakir Necat Yilmaz
- Department of Histology & Embryology, Faculty of Medicine, Mersin University, Mersin, Turkey
| |
Collapse
|
26
|
Geoffrion D, Harissi-Dagher M. Improving glaucoma management for the Boston keratoprosthesis type 1: tubes versus lasers. EXPERT REVIEW OF OPHTHALMOLOGY 2020. [DOI: 10.1080/17469899.2020.1809378] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Dominique Geoffrion
- Department of Experimental Surgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
- Department of Ophthalmology, Université de Montreal, Centre Hospitalier de l’Université de Montréal (CHUM), Montreal, Quebec, Canada
| | - Mona Harissi-Dagher
- Department of Ophthalmology, Université de Montreal, Centre Hospitalier de l’Université de Montréal (CHUM), Montreal, Quebec, Canada
| |
Collapse
|
27
|
Chen X, Lei F, Zhou C, Chodosh J, Wang L, Huang Y, Dohlman CH, Paschalis EI. Glaucoma after Ocular Surgery or Trauma: The Role of Infiltrating Monocytes and Their Response to Cytokine Inhibitors. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:2056-2066. [PMID: 32693061 DOI: 10.1016/j.ajpath.2020.07.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 06/21/2020] [Accepted: 07/07/2020] [Indexed: 12/11/2022]
Abstract
Glaucoma is a frequent and devastating long-term complication following ocular trauma, including corneal surgery, open globe injury, chemical burn, and infection. Postevent inflammation and neuroglial remodeling play a key role in subsequent ganglion cell apoptosis and glaucoma. To this end, this study was designed to investigate the amplifying role of monocyte infiltration into the retina. By using three different ocular injury mouse models (corneal suture, penetrating keratoplasty, and globe injury) and monocyte fate mapping techniques, we show that ocular trauma or surgery can cause robust infiltration of bone marrow-derived monocytes into the retina and subsequent neuroinflammation by up-regulation of Tnf, Il1b, and Il6 mRNA within 24 hours. This is accompanied by ganglion cell apoptosis and neurodegeneration. Prompt inhibition of tumor necrosis factor-α or IL-1β markedly suppresses monocyte infiltration and ganglion cell loss. Thus, acute ocular injury (surgical or trauma) can lead to rapid neuroretinal inflammation and subsequent ganglion cell loss, the hallmark of glaucoma. Infiltrating monocytes play a central role in this process, likely amplifying the inflammatory cascade, aiding in the activation of retinal microglia. Prompt administration of cytokine inhibitors after ocular injury prevents this infiltration and ameliorates the damage to the retina-suggesting that it may be used prophylactically for neuroprotection against post-traumatic glaucoma.
Collapse
Affiliation(s)
- Xiaoniao Chen
- Boston Keratoprosthesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts; Department of Ophthalmology, Chinese PLA General Hospital, Beijing, China
| | - Fengyang Lei
- Boston Keratoprosthesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts
| | - Chengxin Zhou
- Boston Keratoprosthesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts
| | - James Chodosh
- Boston Keratoprosthesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts
| | - Liqiang Wang
- Department of Ophthalmology, Chinese PLA General Hospital, Beijing, China
| | - Yifei Huang
- Department of Ophthalmology, Chinese PLA General Hospital, Beijing, China
| | - Claes H Dohlman
- Boston Keratoprosthesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts.
| | - Eleftherios I Paschalis
- Boston Keratoprosthesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
28
|
Chen L, Liu Y, Zheng XS, Zheng H, Liu PP, Yang XX, Liu Y. Alarmins from conjunctival fibroblasts up-regulate matrix metalloproteinases in corneal fibroblasts. Int J Ophthalmol 2020; 13:1031-1038. [PMID: 32685388 DOI: 10.18240/ijo.2020.07.03] [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] [Received: 10/21/2019] [Accepted: 03/20/2020] [Indexed: 01/10/2023] Open
Abstract
AIM To explore the effects of alarmins produced by necrotic human conjunctival fibroblasts on the release of matrix metalloproteinases (MMPs) by human corneal fibroblasts (HCFs). METHODS A necrotic cell supernatant (NHCS) was prepared by subjecting human conjunctival fibroblasts to three cycles of freezing and thawing. The amounts of interleukin (IL)-1β and tumor necrosis factor (TNF)-α in NHCS were determined by enzyme-linked immunosorbent assays. HCFs exposed to NHCS or other agents in culture were assayed for the release of MMPs as well as for intracellular signaling by immunoblot analysis. The abundance of MMP mRNAs in HCFs was examined by reverse transcription and real-time polymerase chain reaction analysis. RESULTS NHCS increased the release of MMP-1 and MMP-3 by HCFs as well as the amounts of the corresponding mRNAs in the cells. NHCS also induced activation of mitogen-activated protein kinase (MAPK) signaling pathways mediated by extracellular signal-regulated kinase (ERK), p38, and c-Jun NH2-terminal kinase (JNK) as well as elicited that of the nuclear factor (NF)-κB signaling pathway by promoting phosphorylation of the endogenous NF-κB inhibitor IκB-α. Inhibitors of MAPK and NF-κB signaling as well as IL-1 and TNF-α receptor antagonists attenuated the NHCS-induced release of MMP-1 and MMP-3 by HCFs. Furthermore, IL-1β and TNF-α were both detected in NHCS, and treatment of HCFs with these cytokines induced the release of MMP-1 and MMP-3 in a concentration-dependent manner. CONCLUSION Alarmins, including IL-1β and TNF-α, produced by necrotic human conjunctival fibroblasts triggered MMP release in HCFs through activation of MAPK and NF-κB signaling. IL-1β and TNF-α are therefore potential therapeutic targets for the amelioration of corneal stromal degradation in severe ocular burns.
Collapse
Affiliation(s)
- Lin Chen
- Department of Ophthalmology, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, Guangdong Province, China
| | - Ye Liu
- Department of Pathology, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, Guangdong Province, China
| | - Xiao-Shuo Zheng
- Department of Ophthalmology, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, Guangdong Province, China
| | - Hui Zheng
- Department of Ophthalmology, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, Guangdong Province, China
| | - Ping-Ping Liu
- Department of Ophthalmology, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, Guangdong Province, China
| | - Xiu-Xia Yang
- Department of Ophthalmology, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, Guangdong Province, China
| | - Yang Liu
- Department of Ophthalmology, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, Guangdong Province, China
| |
Collapse
|
29
|
Zhou C, Singh A, Qian G, Wolkow N, Dohlman CH, Vavvas DG, Chodosh J, Paschalis EI. Microporous Drug Delivery System for Sustained Anti-VEGF Delivery to the Eye. Transl Vis Sci Technol 2020; 9:5. [PMID: 32855852 PMCID: PMC7422759 DOI: 10.1167/tvst.9.8.5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 05/04/2020] [Indexed: 11/24/2022] Open
Abstract
Purpose To describe a novel microporous drug delivery system (DDS) for sustained anti- vascular endothelial growth factor (VEGF) delivery to the eye and to evaluate its efficacy in a corneal injury model. Methods A macro-porous DDS (1.5 × 1.5 × 4 mm) loaded with 2 mg of bevacizumab was implanted subconjunctivally in three Dutch-belted pigmented rabbits after corneal alkali injury (2N NaOH). Three rabbits received sham DDS. Animals were followed for three months and assessed in vivo and ex vivo for corneal neovascularization (NV), epithelial defect, stromal scarring, endothelial cell loss, and expression of angiogenic and inflammatory markers in the cornea and retina. Results Anti-VEGF DDS treatment led to complete inhibition of superior cornea NV and complete corneal re-epithelialization by day 58 whereas sham DDS resulted in severe cornea NV and persistent epithelial defect (9%∼12% of total cornea area) through the end of the study. Histologically, anti-VEGF DDS significantly reduced CD45+ and F4/80 CD11b+ cell accumulation (79%, P < 0.05) in the cornea, ameliorated tumor necrosis factor–α expression (90%, P < 0.05), reduced corneal stromal scarring and prevented corneal endothelial cell loss, as compared to sham DDS. Moreover, anti-VEGF DDS achieved retinal penetration and reduction in retinal VEGF levels at 3 months. Conclusions Use of subconjunctival anti-VEGF DDS suppresses cornea NV, inflammation, stromal scarring, prevents endothelial cell loss, and abrogates retinal VEGF upregulation in a rabbit corneal alkali burn model. Moreover, it delivers anti-VEGF antibodies to the retina for three months. This delivery platform could enable antibody therapy of other corneal and retinal vascular pathologies. Translational Relevance We describe a method for sustained anti-VEGF delivery to the eye for the treatment of ocular injuries.
Collapse
Affiliation(s)
- Chengxin Zhou
- Boston Keratoprosthesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear and Schepens Eye Research Institute, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Arushi Singh
- Boston Keratoprosthesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear and Schepens Eye Research Institute, Boston, MA, USA
| | - Grace Qian
- Boston Keratoprosthesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear and Schepens Eye Research Institute, Boston, MA, USA
| | - Natalie Wolkow
- Harvard Medical School, Boston, MA, USA.,David G. Cogan Laboratory of Eye Pathology and Ophthalmic Plastic Surgery Service, Massachusetts Eye and Ear Infirmary, Boston, MA, USA
| | - Claes H Dohlman
- Boston Keratoprosthesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear and Schepens Eye Research Institute, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Demetrios G Vavvas
- Angiogenesis Laboratory, Retina Service, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, USA
| | - James Chodosh
- Boston Keratoprosthesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear and Schepens Eye Research Institute, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA.,Disruptive Technology Laboratory (D.T.L.), Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Eleftherios I Paschalis
- Boston Keratoprosthesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear and Schepens Eye Research Institute, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA.,Disruptive Technology Laboratory (D.T.L.), Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
30
|
Abstract
To review clinical aspects and cellular and molecular steps in the development of long-term glaucoma after corneal surgery or acute trauma—especially the pivotal role of tumor necrosis factor alpha (TNF-α), the rapidity of the secondary damage to the retinal ganglion cells, and the clinical promise of early antiinflammatory intervention.
Collapse
|
31
|
Choi SH, Kim MK, Oh JY. Glaucoma after ocular chemical burns: Incidence, risk factors, and outcome. Sci Rep 2020; 10:4763. [PMID: 32179804 PMCID: PMC7076008 DOI: 10.1038/s41598-020-61822-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 03/04/2020] [Indexed: 11/29/2022] Open
Abstract
Effects of chemical injuries on the cornea and limbus have been widely studied; however, little is known about glaucoma after ocular chemical injuries. We herein investigated the incidence, risk factors, and outcome of glaucoma in patients with ocular chemical burns. Medical records were reviewed of patients who visited our clinic for chemical injuries to the ocular surface. Patients were divided into glaucoma and non-glaucoma groups based on high intraocular pressure (IOP) readings. Clinical characteristics, treatment method, and therapeutic and visual outcomes were compared between the two groups. Of 29 patients (40 eyes), 9 patients (15 eyes, 37.5%) were diagnosed with glaucoma at 2.64 ± 2.92 months after injury. Factors associated with glaucoma included male gender (p = 0.0114), bilateral ocular involvement (p = 0.0478), severe ocular surface involvement (Dua grades IV-VI, p = 0.0180), poor initial visual acuity (p = 0.0136), high initial IOP (p < 0.0001), pupil involvement at initial examination (p = 0.0051), and the need for amniotic membrane transplantation in the acute stage (p = 0.0079). At final follow-up, IOP was uncontrolled in 3 eyes (20.0%), and visual acuity was worse in the glaucoma group than in the non-glaucoma group (logMAR 2.94 ± 1.86 vs 0.34 ± 0.69, p < 0.0001). These findings suggest that careful evaluation and intensive treatment for glaucoma are essential in patients with severe ocular burns.
Collapse
Affiliation(s)
- Se Hyun Choi
- Department of Ophthalmology, Hallym University Sacred Heart Hospital, Anyang-si, Gyeonggi-do, Republic of Korea.,Laboratory of Ocular Regenerative Medicine and Immunology, Seoul National University Hospital Biomedical Research Institute, Seoul, Korea
| | - Mee Kum Kim
- Laboratory of Ocular Regenerative Medicine and Immunology, Seoul National University Hospital Biomedical Research Institute, Seoul, Korea.,Department of Ophthalmology, Seoul National University College of Medicine, Seoul, Korea
| | - Joo Youn Oh
- Laboratory of Ocular Regenerative Medicine and Immunology, Seoul National University Hospital Biomedical Research Institute, Seoul, Korea. .,Department of Ophthalmology, Seoul National University College of Medicine, Seoul, Korea.
| |
Collapse
|
32
|
RIP1 kinase mediates angiogenesis by modulating macrophages in experimental neovascularization. Proc Natl Acad Sci U S A 2019; 116:23705-23713. [PMID: 31685620 PMCID: PMC6876205 DOI: 10.1073/pnas.1908355116] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Pathological angiogenesis has been implicated in diverse pathologies. Infiltrating macrophages, especially those activated to M2-like phenotype, are critically important for angiogenesis. Although the role of RIP1 kinase in the regulation of apoptosis, necroptosis, and inflammation has been well-established, its role in angiogenesis remains elusive, despite being abundantly expressed in angiogenesis-related infiltrating macrophages. This study demonstrates that RIP1 kinase inhibition attenuates angiogenesis in multiple mouse models of pathological angiogenesis in vivo and suggests a therapeutic role of RIP1 kinase inhibition in pathological angiogenesis. Mechanistically, the inhibitory effect on angiogenesis depends on RIP kinase inhibition-mediated caspase activation in infiltrating macrophages through suppression of M2-like polarization, and subsequent attenuation of pathological angiogenesis. Inflammation plays an important role in pathological angiogenesis. Receptor-interacting protein 1 (RIP1) is highly expressed in inflammatory cells and is known to play an important role in the regulation of apoptosis, necroptosis, and inflammation; however, a comprehensive description of its role in angiogenesis remains elusive. Here, we show that RIP1 is abundantly expressed in infiltrating macrophages during angiogenesis, and genetic or pharmacological inhibition of RIP1 kinase activity using kinase-inactive RIP1K45A/K45A mice or necrostatin-1 attenuates angiogenesis in laser-induced choroidal neovascularization, Matrigel plug angiogenesis, and alkali injury-induced corneal neovascularization in mice. The inhibitory effect on angiogenesis is mediated by caspase activation through a kinase-independent function of RIP1 and RIP3. Mechanistically, infiltrating macrophages are the key target of RIP1 kinase inhibition to attenuate pathological angiogenesis. Inhibition of RIP1 kinase activity is associated with caspase activation in infiltrating macrophages and decreased expression of proangiogenic M2-like markers but not M1-like markers. Similarly, in vitro, catalytic inhibition of RIP1 down-regulates the expression of M2-like markers in interleukin-4–activated bone marrow-derived macrophages, and this effect is blocked by simultaneous caspase inhibition. Collectively, these results demonstrate a nonnecrotic function of RIP1 kinase activity and suggest that RIP1-mediated modulation of macrophage activation may be a therapeutic target of pathological angiogenesis.
Collapse
|
33
|
Chornenka NM, Raetska YB, Dranitsina AS, Kalmikova OO, Dzerginskiy NE, Savchuk OM, Ostapchenko LI. Expression Levels of Genes Ptgs2 and Tgfb1 in Esophageal Burns and When Introducing Melanin. CYTOL GENET+ 2019. [DOI: 10.3103/s0095452719050050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
34
|
Ibrahim Al-Mashahedah AM, Kanwar RK, Kanwar JR. Utility of nanomedicine targeting scar-forming myofibroblasts to attenuate corneal scarring and haze. Nanomedicine (Lond) 2019; 14:1049-1072. [DOI: 10.2217/nnm-2017-0305] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Corneal scarring refers to the loss of normal corneal tissue, replaced by fibrotic tissue (during wound repair) thereby affecting corneal transparency and vision quality. The corneal wound healing process involves a complex series of physiological events resulting in the transformation of transparent keratocytes into opaque myofibroblasts; the prominent cause of irregular extracellular matrix synthesis leading to the development of corneal opacity/hazy vision. Globally, corneal scarring/haze is one of the most prevalent causes of blindness. Ocular trauma (physical and chemical) and microbial infections induce corneal tissue damage. Although great progress has been made in the clinical management of ocular diseases, the global rates of corneal blindness remain high, nonetheless. The topical conventional modalities treating corneal wounds/injuries have inherent limitations/side effects such as low bioavailability of a therapeutic agent, upregulation of the intraocular pressure and the toxicity/allergy of the drug. These limitations/side effects rather than treating the wound, often negatively affect the healing process, especially, when applied frequently for longer periods. Recently, there has been an increasing evidence provided by the preclinical studies that nanotechnology-based drug-delivery systems can improve drug bioavailability, through controlled drug release and targeted delivery. After reviewing the epidemiology, risk factors of corneal scarring/haze and the conventional ocular medicines, we review here the different nanodrug-delivery systems and potential drug candidates including nanoherbal formulations investigated for their efficacy to heal the damaged cornea. Finally, we discuss the challenges of using these nanomedicinal platforms.
Collapse
Affiliation(s)
- Aseel Mahmood Ibrahim Al-Mashahedah
- Nanomedicine-Laboratory of Immunology & Molecular Biomedical Research (NLIMBR), School of Medicine (SOM), Faculty of Health, Deakin University, Waurn Ponds, Geelong 3216, Australia
| | - Rupinder Kaur Kanwar
- Nanomedicine-Laboratory of Immunology & Molecular Biomedical Research (NLIMBR), School of Medicine (SOM), Faculty of Health, Deakin University, Waurn Ponds, Geelong 3216, Australia
| | - Jagat Rakesh Kanwar
- Nanomedicine-Laboratory of Immunology & Molecular Biomedical Research (NLIMBR), School of Medicine (SOM), Faculty of Health, Deakin University, Waurn Ponds, Geelong 3216, Australia
| |
Collapse
|
35
|
Pietraszkiewicz A, Hampton C, Caplash S, Lei L, Capetanaki Y, Tadvalkar G, Pal-Ghosh S, Stepp MA, Bargagna-Mohan P, Mohan R. Desmin deficiency is not sufficient to prevent corneal fibrosis. Exp Eye Res 2019; 180:155-163. [PMID: 30590024 PMCID: PMC6389382 DOI: 10.1016/j.exer.2018.12.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 11/08/2018] [Accepted: 12/22/2018] [Indexed: 12/16/2022]
Abstract
The type III intermediate filament (IF) proteins vimentin and desmin are sequentially overexpressed in stromal myofibroblasts over the period when fibrosis sets in after corneal injury. Prior findings have revealed vimentin-deficient mice are significantly protected from corneal fibrosis after alkali injury, which has implicated this IF protein as an important regulator of corneal fibrosis. It has remained as yet unproven whether desmin contributes in any significant manner to corneal fibrosis. Here we have employed desmin-deficient (Des KO) mice in the corneal alkali injury model and show that injured Des KO mice develop fibrosis and show similar levels of corneal opacity at 14 days post-injury as wild type (WT) mice and retain this phenotype even at 30d post injury. Des KO corneas from injured mice show upregulation of vimentin and alpha-smooth muscle actin expression to equivalent levels as WT corneas, illuminating that desmin deficiency does not interfere with myofibrobast differentiation. Employing the small molecule withaferin A (WFA), an inhibitor of vimentin, we show that WFA treatment causes the decrease in steady state levels of vimentin and serine 38 phosphorylated vimentin, the latter a biomarker associated with corneal fibrosis, and improved corneal clarity through blockade of myofibroblast differentiation. To investigate further the mechanism of fibrosis in desmin deficiency, we examined keratin 8 expression in the epithelium, and found reduced levels of this cytokeratin in injured Des KO corneas compared to WT corneas. This finding also corroborates the decrease of cell proliferation in injured Des KO corneas compared to that in WT corneas. The fibrotic phenotype of Des KO corneas also features abundant vascularization, further exemplifying the magnitude of corneal pathology. Together, these findings illuminate that desmin does not contribute significantly to corneal fibrosis in this injury model.
Collapse
Affiliation(s)
| | - Christopher Hampton
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT, USA
| | - Sonny Caplash
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT, USA
| | - Ling Lei
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT, USA
| | - Yassemi Capetanaki
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Gauri Tadvalkar
- Department of Ophthalmology, George Washington University, Washington, DC, USA
| | - Sonali Pal-Ghosh
- Department of Ophthalmology, George Washington University, Washington, DC, USA
| | - Mary Ann Stepp
- Department of Ophthalmology, George Washington University, Washington, DC, USA
| | - Paola Bargagna-Mohan
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT, USA
| | - Royce Mohan
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT, USA.
| |
Collapse
|
36
|
Paschalis EI, Taniguchi EV, Chodosh J, Pasquale LR, Colby K, Dohlman CH, Shen LQ. Blood Levels of Tumor Necrosis Factor Alpha and Its Type 2 Receptor Are Elevated in Patients with Boston Type I Keratoprosthesis. Curr Eye Res 2019; 44:599-606. [PMID: 30632412 DOI: 10.1080/02713683.2019.1568500] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Purpose: Boston keratoprosthesis (KPro) patients are prone to glaucoma even with well-controlled intraocular pressure (IOP). Recent experimental data have shown that soluble tumor necrosis factor alpha (TNF-α) after ocular injury may contribute to progressive retinal damage and subsequent glaucoma. This study evaluates the blood plasma levels of soluble TNF-α, TNF receptors 1 (TNFR1) and 2 (TNFR2), and leptin in patients with Boston type I KPro. Methods: Venous blood samples were collected from KPro patients with glaucoma (KPro G, n = 19), KPro patients without glaucoma (KPro NoG, n = 12), primary angle closure glaucoma without KPro (PACG, n = 13), and narrow angles without glaucoma or KPro (NA, n = 21). TNF-α, TNFR1, TNFR2, and leptin levels were quantified using the enzyme-linked immunosorbent assay. Erythrocyte sedimentation rate (ESR) was assessed using the Westergren test. Patients with underlying autoimmune conditions or diabetes were excluded from the study. Results: All groups had similar age, body mass index (BMI), IOP, and ESR (p ≥ 0.11). The mean time from KPro surgery to blood draw was 5.3 ± 3.7 years. Compared to NA patients (0.72 ± 0.3 pg/ml), KPro G and KPro NoG patients had higher blood plasma levels of TNF-α (1.18 ± 0.58 pg/ml, p = 0.006; 1.16 ± 0.50 pg/ml, p = 0.04, respectively). Similarly, KPro G patients had higher blood plasma levels of TNFR2 (2768 ± 1368 pg/ml) than NA patients (2020 ± 435 pg/ml, p = 0.048). In multivariate analysis, KPro status remained positively associated with TNF-α levels (β = 0.36; 95% confidence intervals [CI]: 0.14-0.58; p = 0.002) and TNFR2 levels (β = 458.3; 95% CI: 32.8-883.7; p = 0.035) after adjusting for age, gender, BMI, glaucoma status, and ESR. TNFR1 and leptin levels were not significantly different in the study groups. Conclusions: We detected elevated serum levels of TNF-α and TNFR2 in KPro patients. Longitudinal studies are needed to establish TNF-α and TNFR2 as serum biomarkers related to KPro surgery. Abbreviations: BCVA: best corrected visual acuity; BMI: body mass index; CDR: cup-to-disc ratio; EDTA: ethylenediaminetetraacetic acid; ELISA: enzyme-linked immunosorbent assay; ESR: erythrocyte sedimentation rate; HVF: Humphrey visual field; IOP: intraocular pressure; KPro G: keratoprosthesis with glaucoma; KPro NoG: keratoprosthesis without glaucoma; KPro: keratoprosthesis; MD: mean deviation; NA: narrow angle; non-KPro: without keratoprosthesis; PACG: primary angle closure glaucoma; RNFL: retinal nerve fiber layer; TNF-α: tumor necrosis factor alpha; TNFR1: tumor necrosis factor receptor 1; TNFR2: tumor necrosis factor receptor 2.
Collapse
Affiliation(s)
- Eleftherios I Paschalis
- a Department of Ophthalmology , Harvard Medical School, Massachusetts Eye and Ear , Boston , MA , USA.,b Boston Keratoprosthesis Laboratory , Massachusetts Eye and Ear - Schepens Eye Research Institute, Harvard Medical School , Boston , MA , USA.,c Disruptive Technology Laboratory, Department of Ophthalmology , Massachusetts Eye and Ear, Harvard Medical School , Boston , MA , USA
| | - Elise V Taniguchi
- a Department of Ophthalmology , Harvard Medical School, Massachusetts Eye and Ear , Boston , MA , USA.,b Boston Keratoprosthesis Laboratory , Massachusetts Eye and Ear - Schepens Eye Research Institute, Harvard Medical School , Boston , MA , USA.,d Department of Ophthalmology , Universidade Federal de Sao Paulo , Sao Paulo , Brazil
| | - James Chodosh
- a Department of Ophthalmology , Harvard Medical School, Massachusetts Eye and Ear , Boston , MA , USA.,b Boston Keratoprosthesis Laboratory , Massachusetts Eye and Ear - Schepens Eye Research Institute, Harvard Medical School , Boston , MA , USA.,c Disruptive Technology Laboratory, Department of Ophthalmology , Massachusetts Eye and Ear, Harvard Medical School , Boston , MA , USA
| | - Louis R Pasquale
- e Department of Ophthalmology , Icahn School of Medicine, Mount Sinai Hospital , New York , NY , USA
| | - Kathryn Colby
- f Department of Ophthalmology , University of Chicago , Chicago , IL , USA
| | - Claes H Dohlman
- a Department of Ophthalmology , Harvard Medical School, Massachusetts Eye and Ear , Boston , MA , USA.,b Boston Keratoprosthesis Laboratory , Massachusetts Eye and Ear - Schepens Eye Research Institute, Harvard Medical School , Boston , MA , USA
| | - Lucy Q Shen
- a Department of Ophthalmology , Harvard Medical School, Massachusetts Eye and Ear , Boston , MA , USA
| |
Collapse
|
37
|
Correlation of the expression of inflammatory factors with expression of apoptosis-related genes Bax and Bcl-2, in burned rats. Exp Ther Med 2018; 17:1790-1796. [PMID: 30783451 PMCID: PMC6364214 DOI: 10.3892/etm.2018.7118] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 12/03/2018] [Indexed: 12/25/2022] Open
Abstract
Correlation of the expression of inflammatory factors with expression of apoptosis-related genes, B-cell lymphoma 2 (Bcl-2) and Bcl-2 associated X protein (Bax), in burned rats was investigated. Forty healthy Sprague-Dawley rats were selected and randomly divided into SHAM group (n=10), I° burn group (n=10), II° burn group (n=10) and III° burn group (n=10). Changes in tumor necrosis factor-α (TNF-α), Bax messenger ribonucleic acid (mRNA), Bcl-2 mRNA, Bax protein and Bcl-2 protein expression levels were detected. The correlation of TNF-α, Bax and Bcl-2 with the degree of burn in rats was observed, and the correlation of TNF-α with Bax and Bcl-2 was also analyzed. Moreover, Bax mRNA and Bcl-2 mRNA were detected via reverse transcription-quantitative polymerase chain reaction, and TNF-α, Bax protein and Bcl-2 protein were detected via enzyme-linked immunosorbent assay. In burn groups, TNF-α, Bax mRNA and Bax protein levels were significantly increased at each time point compared with those at the previous time point (P<0.05), but Bcl-2 mRNA and protein levels were significantly decreased compared with those at the previous time point (P<0.05). At the same time point, TNF-α, Bax mRNA, Bcl-2 mRNA, Bax protein and Bcl-2 protein expression levels had statistically significant differences between any given two groups (P<0.05). The TNF-α expression level was positively correlated with Bax expression levels and negatively correlated with Bcl-2 expression levels. Additionally, TNF-α, Bax mRNA and Bax protein had positive correlations with the degree of burn and time after burn, while Bcl-2 mRNA and Bcl-2 protein had negative correlations with the degree of burn and time after burn. Continuous monitoring of changes in the TNF-α level can be used as a means to evaluate the degree of burn and apoptosis, and to prevent the deepening of burn wounds, thus facilitating the early clinical evaluation of prognosis.
Collapse
|
38
|
Paschalis EI, Lei F, Zhou C, Chen XN, Kapoulea V, Hui PC, Dana R, Chodosh J, Vavvas DG, Dohlman CH. Microglia Regulate Neuroglia Remodeling in Various Ocular and Retinal Injuries. THE JOURNAL OF IMMUNOLOGY 2018; 202:539-549. [PMID: 30541880 DOI: 10.4049/jimmunol.1800982] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 11/04/2018] [Indexed: 11/19/2022]
Abstract
Reactive microglia and infiltrating peripheral monocytes have been implicated in many neurodegenerative diseases of the retina and CNS. However, their specific contribution in retinal degeneration remains unclear. We recently showed that peripheral monocytes that infiltrate the retina after ocular injury in mice become permanently engrafted into the tissue, establishing a proinflammatory phenotype that promotes neurodegeneration. In this study, we show that microglia regulate the process of neuroglia remodeling during ocular injury, and their depletion results in marked upregulation of inflammatory markers, such as Il17f, Tnfsf11, Ccl4, Il1a, Ccr2, Il4, Il5, and Csf2 in the retina, and abnormal engraftment of peripheral CCR2+ CX3CR1+ monocytes into the retina, which is associated with increased retinal ganglion cell loss, retinal nerve fiber layer thinning, and pigmentation onto the retinal surface. Furthermore, we show that other types of ocular injuries, such as penetrating corneal trauma and ocular hypertension also cause similar changes. However, optic nerve crush injury-mediated retinal ganglion cell loss evokes neither peripheral monocyte response in the retina nor pigmentation, although peripheral CX3CR1+ and CCR2+ monocytes infiltrate the optic nerve injury site and remain present for months. Our study suggests that microglia are key regulators of peripheral monocyte infiltration and retinal pigment epithelium migration, and their depletion results in abnormal neuroglia remodeling that exacerbates neuroretinal tissue damage. This mechanism of retinal damage through neuroglia remodeling may be clinically important for the treatment of patients with ocular injuries, including surgical traumas.
Collapse
Affiliation(s)
- Eleftherios I Paschalis
- Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114; .,Massachusetts Eye and Ear/Schepens Eye Research Institute, Boston Keratoprosthesis Laboratory, Harvard Medical School, Boston, MA 02114.,Disruptive Technology Laboratory, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114; and
| | - Fengyang Lei
- Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114.,Massachusetts Eye and Ear/Schepens Eye Research Institute, Boston Keratoprosthesis Laboratory, Harvard Medical School, Boston, MA 02114.,Disruptive Technology Laboratory, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114; and
| | - Chengxin Zhou
- Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114.,Massachusetts Eye and Ear/Schepens Eye Research Institute, Boston Keratoprosthesis Laboratory, Harvard Medical School, Boston, MA 02114.,Disruptive Technology Laboratory, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114; and
| | - Xiaohong Nancy Chen
- Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114.,Angiogenesis Laboratory, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114
| | - Vassiliki Kapoulea
- Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114.,Massachusetts Eye and Ear/Schepens Eye Research Institute, Boston Keratoprosthesis Laboratory, Harvard Medical School, Boston, MA 02114
| | - Pui-Chuen Hui
- Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114.,Massachusetts Eye and Ear/Schepens Eye Research Institute, Boston Keratoprosthesis Laboratory, Harvard Medical School, Boston, MA 02114.,Disruptive Technology Laboratory, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114; and
| | - Reza Dana
- Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114.,Massachusetts Eye and Ear/Schepens Eye Research Institute, Boston Keratoprosthesis Laboratory, Harvard Medical School, Boston, MA 02114
| | - James Chodosh
- Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114.,Massachusetts Eye and Ear/Schepens Eye Research Institute, Boston Keratoprosthesis Laboratory, Harvard Medical School, Boston, MA 02114.,Disruptive Technology Laboratory, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114; and
| | - Demetrios G Vavvas
- Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114.,Angiogenesis Laboratory, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114
| | - Claes H Dohlman
- Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114.,Massachusetts Eye and Ear/Schepens Eye Research Institute, Boston Keratoprosthesis Laboratory, Harvard Medical School, Boston, MA 02114
| |
Collapse
|
39
|
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
|
40
|
Controlled delivery of pirfenidone through vitamin E-loaded contact lens ameliorates corneal inflammation. Drug Deliv Transl Res 2018; 8:1114-1126. [DOI: 10.1007/s13346-018-0541-5] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
41
|
Paschalis EI, Lei F, Zhou C, Kapoulea V, Thanos A, Dana R, Vavvas DG, Chodosh J, Dohlman CH. The Role of Microglia and Peripheral Monocytes in Retinal Damage after Corneal Chemical Injury. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:1580-1596. [PMID: 29630857 DOI: 10.1016/j.ajpath.2018.03.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Revised: 03/04/2018] [Accepted: 03/26/2018] [Indexed: 12/17/2022]
Abstract
Eyes that have experienced alkali burn to the surface are excessively susceptible to subsequent severe glaucoma and retinal ganglion cell loss, despite maximal efforts to prevent or slow down the disease. Recently, we have shown, in mice and rabbits, that such retinal damage is neither mediated by the alkali itself reaching the retina nor by intraocular pressure elevation. Rather, it is caused by the up-regulation of tumor necrosis factor-α (TNF-α), which rapidly diffuses posteriorly, causing retinal ganglion cell apoptosis and CD45+ cell activation. Herein, we investigated the involvement of peripheral blood monocytes and microglia in retinal damage. Using CX3CR1+/EGFP::CCR2+/RFP reporter mice and bone marrow chimeras, we show that peripheral CX3CR1+CD45hiCD11b+MHC-II+ monocytes infiltrate into the retina from the optic nerve at 24 hours after the burn and release further TNF-α. A secondary source of peripheral monocyte response originates from a rare population of patrolling myeloid CCR2+ cells of the retina that differentiate into CX3CR1+ macrophages within hours after the injury. As a result, CX3CR1+CD45loCD11b+ microglia become reactive at 7 days, causing further TNF-α release. Prompt TNF-α inhibition after corneal burn suppresses monocyte infiltration and microglia activation, and protects the retina. This study may prove relevant to other injuries of the central nervous system.
Collapse
Affiliation(s)
- Eleftherios I Paschalis
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts; Massachusetts Eye and Ear/Schepens Eye Research Institute, Boston Keratoprosthesis Laboratory, Harvard Medical School, Boston, Massachusetts; Disruptive Technology Laboratory, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts.
| | - Fengyang Lei
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts; Massachusetts Eye and Ear/Schepens Eye Research Institute, Boston Keratoprosthesis Laboratory, Harvard Medical School, Boston, Massachusetts; Disruptive Technology Laboratory, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts
| | - Chengxin Zhou
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts; Massachusetts Eye and Ear/Schepens Eye Research Institute, Boston Keratoprosthesis Laboratory, Harvard Medical School, Boston, Massachusetts; Disruptive Technology Laboratory, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts
| | - Vassiliki Kapoulea
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts; Massachusetts Eye and Ear/Schepens Eye Research Institute, Boston Keratoprosthesis Laboratory, Harvard Medical School, Boston, Massachusetts; Disruptive Technology Laboratory, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts
| | - Aristomenis Thanos
- Angiogenesis Laboratory, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts
| | - Reza Dana
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts
| | - Demetrios G Vavvas
- Angiogenesis Laboratory, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts
| | - James Chodosh
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts; Disruptive Technology Laboratory, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts
| | - Claes H Dohlman
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts; Massachusetts Eye and Ear/Schepens Eye Research Institute, Boston Keratoprosthesis Laboratory, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
42
|
Dohlman CH, Cade F, Regatieri CV, Zhou C, Lei F, Crnej A, Harissi-Dagher M, Robert MC, Papaliodis GN, Chen D, Aquavella JV, Akpek EK, Aldave AJ, Sippel KC, DʼAmico DJ, Dohlman JG, Fagerholm P, Wang L, Shen LQ, González-Andrades M, Chodosh J, Kenyon KR, Foster CS, Pineda R, Melki S, Colby KA, Ciolino JB, Vavvas DG, Kinoshita S, Dana R, Paschalis EI. Chemical Burns of the Eye: The Role of Retinal Injury and New Therapeutic Possibilities. Cornea 2018; 37:248-251. [PMID: 29135604 PMCID: PMC8728745 DOI: 10.1097/ico.0000000000001438] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE To propose a new treatment paradigm for chemical burns to the eye - in the acute and chronic phases. METHODS Recent laboratory and clinical data on the biology and treatment of chemical burns are analyzed. RESULTS Corneal blindness from chemical burns can now be successfully treated with a keratoprosthesis, on immediate and intermediate bases. Long term outcomes, however, are hampered by early retinal damage causing glaucoma. New data suggest that rapid diffusion of inflammatory cytokines posteriorly (TNF-α, etc) can severely damage the ganglion cells. Prompt anti-TNF-α treatment is markedly neuroprotective. Long term profound reduction of the intraocular pressure is also vital. CONCLUSION A new regimen, in addition to standard treatment, for severe chemical burns is proposed. This involves tumor necrosis factor alpha (TNF-α) inhibition promptly after the accident (primarily for retinal neuroprotection), prophylactic maximal lowering of the intraocular pressure (starting immediately), and keratoprosthesis implantation in a later quiet state.
Collapse
Affiliation(s)
- Claes H Dohlman
- Cornea Service and Boston Keratoprosthesis Laboratory, Massachusetts Eye and Ear Infirmary and Schepens Eye Research Institute, Harvard Medical School, Boston, MA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Zhou C, Robert MC, Kapoulea V, Lei F, Stagner AM, Jakobiec FA, Dohlman CH, Paschalis EI. Sustained Subconjunctival Delivery of Infliximab Protects the Cornea and Retina Following Alkali Burn to the Eye. Invest Ophthalmol Vis Sci 2017; 58:96-105. [PMID: 28114570 PMCID: PMC5231904 DOI: 10.1167/iovs.16-20339] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose Tumor necrosis factor (TNF)-α is upregulated in eyes following corneal alkali injury and contributes to corneal and also retinal damage. Prompt TNF-α inhibition by systemic infliximab ameliorates retinal damage and improves corneal wound healing. However, systemic administration of TNF-α inhibitors carries risk of significant complications, whereas topical eye-drop delivery is hindered by poor ocular bioavailability and the need for patient adherence. This study investigates the efficacy of subconjunctival delivery of TNF-α antibodies using a polymer-based drug delivery system (DDS). Methods The drug delivery system was prepared using porous polydimethylsiloxane/polyvinyl alcohol composite fabrication and loaded with 85 μg of infliximab. Six Dutch-belted pigmented rabbits received ocular alkali burn with NaOH. Immediately after the burn, subconjunctival implantation of anti-TNF-α DDS was performed in three rabbits while another three received sham DDS (without antibody). Rabbits were followed with photography for 3 months. Results After 3 months, the device was found to be well tolerated by the host and the eyes exhibited less corneal damage as compared to eyes implanted with a sham DDS without drug. The low dose treatment suppressed CD45 and TNF-α expression in the burned cornea and inhibited retinal ganglion cell apoptosis and optic nerve degeneration, as compared to the sham DDS treated eyes. Immunolocalization revealed drug penetration in the conjunctiva, cornea, iris, and choroid, with residual infliximab in the DDS 3 months after implantation. Conclusions This reduced-risk biologic DDS improves corneal wound healing and provides retinal neuroprotection, and may be applicable not only to alkali burns but also to other inflammatory surgical procedures such as penetrating keratoplasty and keratoprosthesis implantation.
Collapse
Affiliation(s)
- Chengxin Zhou
- Boston Keratoprosthesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear and Schepens Eye Research Institute, Boston, Massachusetts, United States 2Harvard Medical School, Boston, Massachusetts, United States
| | - Marie-Claude Robert
- Department of Ophthalmology, Université de Montreal, Montreal, Quebec, Canada 4Centre Hospitalier de l'Université de Montreal, Hospital Notre-Dame, Montreal, Quebec, Canada
| | - Vassiliki Kapoulea
- Boston Keratoprosthesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear and Schepens Eye Research Institute, Boston, Massachusetts, United States 2Harvard Medical School, Boston, Massachusetts, United States
| | - Fengyang Lei
- Boston Keratoprosthesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear and Schepens Eye Research Institute, Boston, Massachusetts, United States 2Harvard Medical School, Boston, Massachusetts, United States
| | - Anna M Stagner
- Harvard Medical School, Boston, Massachusetts, United States 5David G. Cogan Ophthalmic Pathology Laboratory, Massachusetts Eye and Ear, Boston, Massachusetts, United States
| | - Frederick A Jakobiec
- Harvard Medical School, Boston, Massachusetts, United States 5David G. Cogan Ophthalmic Pathology Laboratory, Massachusetts Eye and Ear, Boston, Massachusetts, United States
| | - Claes H Dohlman
- Boston Keratoprosthesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear and Schepens Eye Research Institute, Boston, Massachusetts, United States 2Harvard Medical School, Boston, Massachusetts, United States
| | - Eleftherios I Paschalis
- Boston Keratoprosthesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear and Schepens Eye Research Institute, Boston, Massachusetts, United States 2Harvard Medical School, Boston, Massachusetts, United States 6Disruptive Technology Laboratory (D.T.L.), Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States
| |
Collapse
|
44
|
Choi H, Phillips C, Oh JY, Stock EM, Kim DK, Won JK, Fulcher S. Comprehensive Modeling of Corneal Alkali Injury in the Rat Eye. Curr Eye Res 2017. [DOI: 10.1080/02713683.2017.1317817] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Hosoon Choi
- Department of Basic Research, Central Texas Veterans Research Foundation, Temple, TX, USA
| | - Casie Phillips
- Department of Basic Research, Central Texas Veterans Research Foundation, Temple, TX, USA
| | - Joo Youn Oh
- Department of Ophthalmology, Seoul National University Hospital, Daehak-ro, Jongno-gu, Seoul, Republic of Korea
| | - Eileen M. Stock
- Cooperative Studies Program Coordinating Center, VA Maryland Health Care System, Perry Point, MD, USA
| | - Dong-Ki Kim
- Institute for Regenerative Medicine, Texas A&M Health Science Center, College Station, TX, USA
| | - Jae-Kyung Won
- Department of Pathology, Seoul National University Hospital, Daehak-ro, Jongno-gu, Seoul, Republic of Korea
| | - Samuel Fulcher
- Department of Surgery, Ophthalmology Section, Central Texas Veterans Health Care System, Temple, TX, USA
| |
Collapse
|
45
|
Paschalis EI, Zhou C, Lei F, Scott N, Kapoulea V, Robert MC, Vavvas D, Dana R, Chodosh J, Dohlman CH. Mechanisms of Retinal Damage after Ocular Alkali Burns. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:1327-1342. [PMID: 28412300 PMCID: PMC5455067 DOI: 10.1016/j.ajpath.2017.02.005] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2017] [Revised: 02/06/2017] [Accepted: 02/14/2017] [Indexed: 01/01/2023]
Abstract
Alkali burns to the eye constitute a leading cause of worldwide blindness. In recent case series, corneal transplantation revealed unexpected damage to the retina and optic nerve in chemically burned eyes. We investigated the physical, biochemical, and immunological components of retinal injury after alkali burn and explored a novel neuroprotective regimen suitable for prompt administration in emergency departments. Thus, in vivo pH, oxygen, and oxidation reduction measurements were performed in the anterior and posterior segment of mouse and rabbit eyes using implantable microsensors. Tissue inflammation was assessed by immunohistochemistry and flow cytometry. The experiments confirmed that the retinal damage is not mediated by direct effect of the alkali, which is effectively buffered by the anterior segment. Rather, pH, oxygen, and oxidation reduction changes were restricted to the cornea and the anterior chamber, where they caused profound uveal inflammation and release of proinflammatory cytokines. The latter rapidly diffuse to the posterior segment, triggering retinal damage. Tumor necrosis factor-α was identified as a key proinflammatory mediator of retinal ganglion cell death. Blockade, by either monoclonal antibody or tumor necrosis factor receptor gene knockout, reduced inflammation and retinal ganglion cell loss. Intraocular pressure elevation was not observed in experimental alkali burns. These findings illuminate the mechanism by which alkali burns cause retinal damage and may have importance in designing therapies for retinal protection.
Collapse
MESH Headings
- Alkalies
- Animals
- Apoptosis/drug effects
- Apoptosis/physiology
- Burns, Chemical/drug therapy
- Burns, Chemical/etiology
- Burns, Chemical/metabolism
- Burns, Chemical/pathology
- Cornea/immunology
- Corneal Injuries/drug therapy
- Corneal Injuries/etiology
- Corneal Injuries/metabolism
- Corneal Injuries/pathology
- Disease Models, Animal
- Drug Evaluation, Preclinical/methods
- Eye Burns/drug therapy
- Eye Burns/etiology
- Eye Burns/metabolism
- Eye Burns/pathology
- Hydrogen-Ion Concentration
- Infliximab/pharmacology
- Infliximab/therapeutic use
- Mice, Inbred C57BL
- Mice, Knockout
- Neuroprotective Agents/pharmacology
- Neuroprotective Agents/therapeutic use
- Oxidation-Reduction
- Rabbits
- Receptors, Tumor Necrosis Factor, Type I/deficiency
- Receptors, Tumor Necrosis Factor, Type I/genetics
- Receptors, Tumor Necrosis Factor, Type II/deficiency
- Receptors, Tumor Necrosis Factor, Type II/genetics
- Retina/immunology
- Retina/injuries
- Retina/metabolism
- Retina/pathology
- Retinal Ganglion Cells/drug effects
- Retinal Ganglion Cells/pathology
- Sodium Hydroxide
- Tumor Necrosis Factor-alpha/antagonists & inhibitors
- Tumor Necrosis Factor-alpha/metabolism
- Uvea/metabolism
- Uveitis, Anterior/chemically induced
- Uveitis, Anterior/metabolism
- Uveitis, Anterior/pathology
- Uveitis, Anterior/prevention & control
Collapse
Affiliation(s)
- Eleftherios I Paschalis
- Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts; Boston Keratoprosthesis Laboratory, Schepens Eye Research Institute, Harvard Medical School, Boston, Massachusetts; Disruptive Technology Laboratory, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts.
| | - Chengxin Zhou
- Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts; Boston Keratoprosthesis Laboratory, Schepens Eye Research Institute, Harvard Medical School, Boston, Massachusetts; Disruptive Technology Laboratory, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Fengyang Lei
- Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts; Boston Keratoprosthesis Laboratory, Schepens Eye Research Institute, Harvard Medical School, Boston, Massachusetts; Disruptive Technology Laboratory, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Nathan Scott
- Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts; Boston Keratoprosthesis Laboratory, Schepens Eye Research Institute, Harvard Medical School, Boston, Massachusetts; Disruptive Technology Laboratory, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Vassiliki Kapoulea
- Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts; Boston Keratoprosthesis Laboratory, Schepens Eye Research Institute, Harvard Medical School, Boston, Massachusetts; Disruptive Technology Laboratory, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Marie-Claude Robert
- Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts; Boston Keratoprosthesis Laboratory, Schepens Eye Research Institute, Harvard Medical School, Boston, Massachusetts; Centre Hospitalier de l'Universite de Montreal, Hospital Notre-Dame, Montreal, Quebec, Canada
| | - Demetrios Vavvas
- Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts; Angiogenesis Laboratory, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Reza Dana
- Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - James Chodosh
- Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts; Disruptive Technology Laboratory, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Claes H Dohlman
- Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts; Boston Keratoprosthesis Laboratory, Schepens Eye Research Institute, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
46
|
Expression of peptidylarginine deiminase 4 in an alkali injury model of retinal gliosis. Biochem Biophys Res Commun 2017; 487:134-139. [PMID: 28400047 DOI: 10.1016/j.bbrc.2017.04.031] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 04/07/2017] [Indexed: 11/20/2022]
Abstract
Citrullination is an important posttranslational modification that occurs during retinal gliosis. We examined the expression of peptidyl arginine deiminases (PADs) to identify the PADs that mediate citrullination in a model of alkali-induced retinal gliosis. Mouse corneas were exposed to 1.0 N NaOH and posterior eye tissue from injured and control uninjured eyes was evaluated for transcript levels of various PADs by reverse-transcription polymerase chain reaction (RT-PCR), and quantitative RT-PCR (qPCR). Retinas were also subjected to immunohistochemistry (IHC) for glial fibrillary acidic protein (GFAP), citrullinated species, PAD2, and PAD4 and tissue levels of GFAP, citrullinated species, and PAD4 were measured by western blots. In other experiments, the PAD4 inhibitor streptonigrin was injected intravitreally into injured eyes ex vivo to test inhibitory activity in an organ culture system. We found that uninjured retina and choroid expressed Pad2 and Pad4 transcripts. Pad4 transcript levels increased by day 7 post-injury (p < 0.05), whereas Pad2 levels did not change significantly (p > 0.05) by qPCR. By IHC, PAD2 was expressed in uninjured eyes along ganglion cell astrocytes, but in injured retina PAD2 was downregulated at 7 days. On the other hand, PAD4 showed increased staining in the retina upon injury revealing a pattern that overlapped with filamentous GFAP staining in Müller glial processes by 7 days. Injury-induced citrullination and soluble GFAP protein levels were reduced by PAD4 inhibition in western blot experiments of organ cultures. Together, our findings for the first time identify PAD4 as a novel injury-inducible druggable target for retinal gliosis.
Collapse
|
47
|
Khalili H, Lee RW, Khaw PT, Brocchini S, Dick AD, Copland DA. An anti-TNF-α antibody mimetic to treat ocular inflammation. Sci Rep 2016; 6:36905. [PMID: 27874029 PMCID: PMC5118814 DOI: 10.1038/srep36905] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 10/20/2016] [Indexed: 12/14/2022] Open
Abstract
Infliximab is an antibody that neutralizes TNF-α and is used principally by systemic administration to treat many inflammatory disorders. We prepared the antibody mimetic Fab-PEG-Fab (FpFinfliximab) for direct intravitreal injection to assess whether such formulations have biological activity and potential utility for ocular use. FpFinfliximab was designed to address side effects caused by antibody degradation and the presence of the Fc region. Surface plasmon resonance analysis indicated that infliximab and FpFinfliximab maintained binding affinity for both human and murine recombinant TNF-α. No Fc mediated RPE cellular uptake was observed for FpFinfliximab. Both Infliximab and FpFinfliximab suppressed ocular inflammation by reducing the number of CD45+ infiltrate cells in the EAU mice after a single intravitreal injection at the onset of peak disease. These results offer an opportunity to develop and formulate for ocular use, FpF molecules designed for single and potentially multiple targets using bi-specific FpFs.
Collapse
Affiliation(s)
- Hanieh Khalili
- UCL School of Pharmacy, London, UK.,National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, UK.,University of East London, School of Health, Sport and Bioscience, Water lane, Stratford campus, London, E15 4LZ, UK
| | - Richard W Lee
- National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, UK.,School of Clinical Sciences, University of Bristol, Bristol, UK
| | - Peng T Khaw
- National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, UK
| | - Steve Brocchini
- UCL School of Pharmacy, London, UK.,National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, UK
| | - Andrew D Dick
- National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, UK.,School of Clinical Sciences, University of Bristol, Bristol, UK
| | - David A Copland
- National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, UK.,School of Clinical Sciences, University of Bristol, Bristol, UK
| |
Collapse
|
48
|
Črnej A, Omoto M, Dohlman TH, Gonzalez-Andrades M, Paschalis EI, Cruzat A, Vu THK, Doorenbos M, Chen DF, Dohlman CH, Dana R. Effect of Penetrating Keratoplasty and Keratoprosthesis Implantation on the Posterior Segment of the Eye. Invest Ophthalmol Vis Sci 2016; 57:1643-8. [PMID: 27054516 PMCID: PMC4829084 DOI: 10.1167/iovs.15-17557] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE To compare the effects of post-penetrating keratoplasty (PK) and post-keratoprosthesis (KPro) surgery-related inflammation on the posterior segment of the eye and to assess inhibition of tumor necrosis factor alpha (TNFα) and interleukin-1 beta (IL-1β) on these effects. METHODS BALB/C (syngeneic) or C57BL/6 (allogeneic) corneas were transplanted onto BALB/C host beds as part of PK or miniature KPro (m-KPro) implantation. Intraocular pressure (IOP) was measured via an intracameral pressure sensor; tissues were harvested and analyzed 8 weeks after surgery. Expression of TNFα and IL-1β in the retina was analyzed using real-time quantitative (q)PCR. Optic nerve degeneration (axon count, circularity, and area) was assessed quantitatively using ImageJ software. After m-KPro implantation, mice were treated with saline, anti-TNFα, or anti-IL-1β antibody, and axonal loss was assessed after 10 weeks. RESULTS Mean IOP was within normal limits in the operated and fellow eyes in all groups. The mRNA expression of TNFα and IL-1β was highest in m-KPro groups with either syngeneic or an allogeneic carrier. We observed optic nerve degeneration in both allogeneic PK and m-KPro implanted eyes with an allogeneic carrier. However, TNFα blockade significantly reduced axonal loss by 35%. CONCLUSIONS Allogeneic PK and m-KPro implants with an allogeneic carrier lead to chronic inflammation in the posterior segment of the eye, resulting in optic nerve degeneration. In addition, blockade of TNFα prevents axonal degeneration in this preclinical model of allogeneic m-KPro (alloKPro) implantation.
Collapse
|
49
|
Keratoprosthesis: A Review of Recent Advances in the Field. J Funct Biomater 2016; 7:jfb7020013. [PMID: 27213461 PMCID: PMC4932470 DOI: 10.3390/jfb7020013] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2016] [Revised: 05/04/2016] [Accepted: 05/13/2016] [Indexed: 12/24/2022] Open
Abstract
Since its discovery in the years of the French Revolution, the field of keratoprostheses has evolved significantly. However, the path towards its present state has not always been an easy one. Initially discarded for its devastating complications, the introduction of new materials and the discovery of antibiotics in the last century gave new life to the field. Since then, the use of keratoprostheses for severe ocular surface disorders and corneal opacities has increased significantly, to the point that it has become a standard procedure for corneal specialists worldwide. Although the rate of complications has significantly been reduced, these can impede the long-term success, since some of them can be visually devastating. In an attempt to overcome these complications, researchers in the field have been recently working on improving the design of the currently available devices, by introducing the use of new materials that are more biocompatible with the eye. Here we present an update on the most recent research in the field.
Collapse
|
50
|
Bai JQ, Qin HF, Zhao SH. Research on mouse model of grade II corneal alkali burn. Int J Ophthalmol 2016; 9:487-90. [PMID: 27162717 DOI: 10.18240/ijo.2016.04.02] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Accepted: 06/16/2015] [Indexed: 11/23/2022] Open
Abstract
AIM To choose appropriate concentration of sodium hydroxide (NaOH) solution to establish a stable and consistent corneal alkali burn mouse model in grade II. METHODS The mice (n=60) were randomly divided into four groups and 15 mice each group. Corneal alkali burns were induced by placing circle filter paper soaked with NaOH solutions on the right central cornea for 30s. The concentrations of NaOH solutions of groups A, B, C, and D were 0.1 mol/L, 0.15 mol/L, 0.2 mol/L, and 1.0 mol/L respectively. Then these corneas were irrigated with 20 mL physiological saline (0.9% NaCl). On day 7 postburn, slit lamp microscope was used to observe corneal opacity, corneal epithelial sodium fluorescein staining positive rate, incidence of corneal ulcer and corneal neovascularization, meanwhile pictures of the anterior eyes were taken. Cirrus spectral domain optical coherence tomography was used to scan cornea to observe corneal epithelial defect and corneal ulcer. RESULTS Corneal opacity scores (x±s) were not significantly different between the group A and group B (P=0.097). Incidence of corneal ulcer in group B was significantly higher than that in group A (P=0.035). Incidence of corneal ulcer and perforation rate in group B was lower than that in group C. Group C and D had corneal neovascularization, and incidence of corneal neovascularization in group D was significantly higher than that in group C (P=0.000). CONCLUSION Using 0.15 mol/L NaOH can establish grade II mouse model of corneal alkali burns.
Collapse
Affiliation(s)
- Jun-Qiang Bai
- Department of Ophthalmology, Changhai Hospital, the Second Military Medical University, Shanghai 200433, China
| | - Hai-Feng Qin
- Department of Ophthalmology, Changhai Hospital, the Second Military Medical University, Shanghai 200433, China
| | - Shi-Hong Zhao
- Department of Ophthalmology, Changhai Hospital, the Second Military Medical University, Shanghai 200433, China
| |
Collapse
|