1
|
Qi Q, Su D, Zhuang S, Yao S, Heindl LM, Fan X, Lin M, Li J, Pang Y. Progress in Nanotechnology for Treating Ocular Surface Chemical Injuries: Reflecting on Advances in Ophthalmology. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2407340. [PMID: 39755928 PMCID: PMC11809354 DOI: 10.1002/advs.202407340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 11/26/2024] [Indexed: 01/06/2025]
Abstract
Ocular surface chemical injuries often result in permanent visual impairment and necessitate complex, long-term treatments. Immediate and extensive irrigation serves as the first-line intervention, followed by various therapeutic protocols applied throughout different stages of the condition. To optimize outcomes, conventional regimens increasingly incorporate biological agents and surgical techniques. In recent years, nanotechnology has made significant strides, revolutionizing the management of ocular surface chemical injuries by enabling sustained drug release, enhancing treatment efficacy, and minimizing side effects. This review provides a comprehensive analysis of the etiology, epidemiology, classification, and conventional therapies for ocular chemical burns, with a special focus on nanotechnology-based drug delivery systems in managing ocular surface chemical injuries. Twelve categories of nanocarrier platforms are examined, including liposomes, nanoemulsions, nanomicelles, nanowafers, nanostructured lipid carriers, nanoparticles, hydrogels, dendrimers, nanocomplexes, nanofibers, nanozymes, and nanocomposite materials, highlighting their advantages in targeted delivery, biocompatibility, and improved healing efficacy. Additionally, current challenges and limitations in the field are discussed and the future potential of nanotechnology in treating ocular diseases is explored. This review presents the most extensive examination of this topic to date, aiming to link recent advancements with broader therapeutic strategies.
Collapse
Affiliation(s)
- Qiaoran Qi
- Department of OphthalmologyNinth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011China
- Shanghai Key Laboratory of Orbital Diseases and Ocular OncologyCenter for Basic Medical Research and Innovation in Visual System DiseasesMinistry of EducationShanghai200011China
| | - Dai Su
- Department of OphthalmologyNinth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011China
- Shanghai Key Laboratory of Orbital Diseases and Ocular OncologyCenter for Basic Medical Research and Innovation in Visual System DiseasesMinistry of EducationShanghai200011China
| | - Shuqin Zhuang
- Department of OphthalmologyNinth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011China
- Shanghai Key Laboratory of Orbital Diseases and Ocular OncologyCenter for Basic Medical Research and Innovation in Visual System DiseasesMinistry of EducationShanghai200011China
| | - Sunyuan Yao
- Department of OphthalmologyNinth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011China
- Shanghai Key Laboratory of Orbital Diseases and Ocular OncologyCenter for Basic Medical Research and Innovation in Visual System DiseasesMinistry of EducationShanghai200011China
| | - Ludwig M. Heindl
- Department of OphthalmologyFaculty of Medicine and University Hospital CologneUniversity of Cologne50937CologneGermany
- Center for Integrated Oncology (CIO)Aachen‐Bonn‐Cologne‐DuesseldorfCologneGermany
| | - Xianqun Fan
- Department of OphthalmologyNinth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011China
- Shanghai Key Laboratory of Orbital Diseases and Ocular OncologyCenter for Basic Medical Research and Innovation in Visual System DiseasesMinistry of EducationShanghai200011China
| | - Ming Lin
- Department of OphthalmologyNinth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011China
- Shanghai Key Laboratory of Orbital Diseases and Ocular OncologyCenter for Basic Medical Research and Innovation in Visual System DiseasesMinistry of EducationShanghai200011China
| | - Jin Li
- Department of OphthalmologyNinth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011China
- Shanghai Key Laboratory of Orbital Diseases and Ocular OncologyCenter for Basic Medical Research and Innovation in Visual System DiseasesMinistry of EducationShanghai200011China
| | - Yan Pang
- Department of OphthalmologyNinth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011China
- Shanghai Key Laboratory of Orbital Diseases and Ocular OncologyCenter for Basic Medical Research and Innovation in Visual System DiseasesMinistry of EducationShanghai200011China
- Shanghai Frontiers Science Center of Drug Target Identification and DeliverySchool of Pharmaceutical SciencesShanghai Jiao Tong UniversityShanghai200240China
| |
Collapse
|
2
|
Shimazaki J, Tomida D, Yagi-Yaguchi Y, Satake Y, Yamaguchi T. Topical tacrolimus for high-risk corneal transplantation: a randomized, clinical trial. BMC Ophthalmol 2024; 24:251. [PMID: 38867175 PMCID: PMC11167901 DOI: 10.1186/s12886-024-03506-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 05/29/2024] [Indexed: 06/14/2024] Open
Abstract
BACKGROUND The prevalence of rejection is 10-30% in penetrating keratoplasty (PKP) case, and the rate is higher in cases of high-risk patients. Although using topical corticosteroids is a standard method for management the rejection of post-PKP patients, it may not be sufficiently potent in high-risk patients. Topical administration of tacrolimus (TAC) may be effective in suppression rejection after corneal transplantation. This study aimed to investigate the efficacy and safety of topical TAC in high-risk PKP patients in Japan. METHODS This study was a single centre, single-blinded, randomized controlled trial. Patients with a history of PKP, graft rejection, atopic dermatitis, or deep corneal neovascularisation who underwent PKP were enrolled. They were randomly assigned to receive 0.1% TAC ophthalmic suspension or artificial tear (AT) up to week 52 after surgery. All participants received 0.1% betamethasone up to week 13 after surgery then they received 0.1% fluorometholone up to week 52. The incidence of immunological rejection during the observation period was the main outcome measure in this study. RESULTS Thirty patients were enrolled in this study, and 12 eyes in the TAC group and 13 eyes in the AT group completed the study, respectively. Five out of 30 patients discontinued participation after providing informed consent. No serious adverse effects were developed in patients who received 0.1% TAC ophthalmic suspension. No rejection episodes occurred in the TAC group, while one eye in the AT group had rejection. Graft clarity, best spectacle-corrected visual acuity, intraocular pressure, and corneal endothelial cell density were not significantly different between the TAC and AT groups. CONCLUSION Our results demonstrated that good tolerability of 0.1% TAC ophthalmic suspension. However, we failed to demonstrate its efficacy in preventing immunological rejection in high-risk patients undergoing PKP. TRIAL REGISTRATION This study was first registered in the University Hospital Medical Information Network (UMIN000029669, Date of registration: November 1, 2017). With the enforcement of the Clinical Trial Act in Japan, the study re-registered in the Japan Registry of Clinical Trials (jRCTs031180342, Date of registration: March 18, 2019).
Collapse
Affiliation(s)
- Jun Shimazaki
- Department of Ophthalmology, Tokyo Dental College Ichikawa General Hospital, 5-11-13 Sugano, Ichikawa, 272-8513, Chiba, Japan.
| | - Daisuke Tomida
- Department of Ophthalmology, Tokyo Dental College Ichikawa General Hospital, 5-11-13 Sugano, Ichikawa, 272-8513, Chiba, Japan
| | - Yukari Yagi-Yaguchi
- Department of Ophthalmology, Tokyo Dental College Ichikawa General Hospital, 5-11-13 Sugano, Ichikawa, 272-8513, Chiba, Japan
| | - Yoshiyuki Satake
- Department of Ophthalmology, Tokyo Dental College Ichikawa General Hospital, 5-11-13 Sugano, Ichikawa, 272-8513, Chiba, Japan
| | - Takefumi Yamaguchi
- Department of Ophthalmology, Tokyo Dental College Ichikawa General Hospital, 5-11-13 Sugano, Ichikawa, 272-8513, Chiba, Japan
| |
Collapse
|
3
|
Cao Z, Ramadan A, Tai A, Zetterberg F, Panjwani N. Anti-Angiogenic and Anti-Scarring Dual Effect of Galectin-3 Inhibition in Mouse Models of Corneal Wound Healing. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:447-458. [PMID: 38159722 DOI: 10.1016/j.ajpath.2023.11.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 11/05/2023] [Accepted: 11/28/2023] [Indexed: 01/03/2024]
Abstract
Corneal scarring is the third leading cause of global blindness. Neovascularization of ocular tissues is a major predisposing factor in scar development. Although corneal transplantation is effective in restoring vision, some patients are at high risk for graft rejection due to the presence of blood vessels in the injured cornea. Current treatment options for controlling corneal scarring are limited, and outcomes are typically poor. In this study, topical application of a small-molecule inhibitor of galectin-3, GB1265, in mouse models of corneal wound healing, led to the reduction of the following in injured corneas: i) corneal angiogenesis; ii) corneal fibrosis; iii) infiltration of immune cells; and iv) expression of the proinflammatory cytokine IL-1β. Four independent techniques (RNA sequencing, NanoString, real-time quantitative RT-PCR, and Western blot analysis) determined that decreased corneal opacity in the galectin-3 inhibitor-treated corneas was associated with decreases in the numbers of genes and signaling pathways known to promote fibrosis. These findings allowed for a high level of confidence in the conclusion that galectin-3 inhibition by the small-molecule inhibitor GB1265 has dual anti-angiogenic and anti-scarring effects. Targeting galectin-3 by GB1265 is, thus, attractive for the development of innovative therapies for a myriad of ocular and nonocular diseases characterized by pathologic angiogenesis and fibrosis.
Collapse
Affiliation(s)
- Zhiyi Cao
- New England Eye Center/Department of Ophthalmology, Tufts University School of Medicine, Boston, Massachusetts
| | - Abdulraouf Ramadan
- New England Eye Center/Department of Ophthalmology, Tufts University School of Medicine, Boston, Massachusetts
| | - Albert Tai
- Department of Immunology, Tufts University School of Medicine, Boston, Massachusetts
| | | | - Noorjahan Panjwani
- New England Eye Center/Department of Ophthalmology, Tufts University School of Medicine, Boston, Massachusetts; Department of Developmental, Molecular, and Chemical Biology, Tufts University School of Medicine, Boston, Massachusetts.
| |
Collapse
|
4
|
Zhu J, Inomata T, Nakamura M, Fujimoto K, Akasaki Y, Fujio K, Yanagawa A, Uchida K, Sung J, Negishi N, Nagino K, Okumura Y, Miura M, Shokirova H, Kuwahara M, Hirosawa K, Midorikawa-Inomata A, Eguchi A, Huang T, Yagita H, Habu S, Okumura K, Murakami A. Anti-CD80/86 antibodies inhibit inflammatory reaction and improve graft survival in a high-risk murine corneal transplantation rejection model. Sci Rep 2022; 12:4853. [PMID: 35318419 PMCID: PMC8941080 DOI: 10.1038/s41598-022-08949-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 03/15/2022] [Indexed: 12/27/2022] Open
Abstract
We investigated the effects of anti-CD80/86 antibodies in a murine high-risk corneal transplantation rejection model. A mixed lymphocyte reaction (MLR) assay was conducted with anti-CD80/86 antibodies. Inflammatory cytokine levels in the culture supernatant were measured using an enzyme-linked immunosorbent assay. Interferon (IFN)-γ-producing CD4+ T cell frequencies in the MLR were assessed using flow cytometry. In vivo, high-risk corneal allograft survival and IFN-γ-producing CD4+ T cell frequencies in corneal grafts were assessed with intraperitoneal injection of anti-CD80/86 antibodies compared to phosphate-buffered saline (PBS). RNA-sequencing was performed on corneal grafts 2 weeks post-transplantation. Anti-CD80/86 antibodies significantly decreased T-cell proliferation, IFN-γ+-producing CD4+ T cell frequencies, and IFN-γ, interleukin (IL)-1β, IL-2, IL-10, and tumor necrosis factor-α production in the MLR compared to PBS injection. Intraperitoneal injection of anti-CD80/86 antibodies significantly prolonged corneal graft survival and decreased IFN-γ+-producing CD4+ T cell frequencies compared to PBS injection. Gene set enrichment analysis showed that the gene sets mainly enriched in the control group were related to allograft rejection and inflammatory response compared to PBS injection. Anti-CD80/86 antibodies significantly prolonged corneal graft survival by inhibiting T-cell proliferation and inflammatory response.
Collapse
Affiliation(s)
- Jun Zhu
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.,Department of Ophthalmology, Subei People's Hospital Affiliated to Yangzhou University, Yangzhou, Jiangsu, China
| | - Takenori Inomata
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan. .,Department of Strategic Operating Room Management and Improvement, Juntendo University Graduate School of Medicine, Tokyo, Japan. .,Department of Hospital Administration, Juntendo University Graduate School of Medicine, Tokyo, Japan. .,Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan.
| | - Masahiro Nakamura
- Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Precision Health, Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Keiichi Fujimoto
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Yasutsugu Akasaki
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.,Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Kenta Fujio
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.,Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Ai Yanagawa
- Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Koichiro Uchida
- Center for Immune Therapeutics and Diagnosis, Juntendo University, Tokyo, Japan
| | - Jaemyoung Sung
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.,Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Naoko Negishi
- Atopy Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Department of Indoor Environment Neurophysiological Research, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Ken Nagino
- Department of Hospital Administration, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yuichi Okumura
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.,Department of Strategic Operating Room Management and Improvement, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Maria Miura
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.,Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hurramhon Shokirova
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Mizu Kuwahara
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.,Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Kunihiko Hirosawa
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.,Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Akie Midorikawa-Inomata
- Department of Hospital Administration, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Atsuko Eguchi
- Department of Hospital Administration, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Tianxiang Huang
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.,Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hideo Yagita
- Department of Immunology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Sonoko Habu
- Atopy Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Ko Okumura
- Center for Immune Therapeutics and Diagnosis, Juntendo University, Tokyo, Japan
| | - Akira Murakami
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.,Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
5
|
Liu S, Wong YL, Walkden A. Current Perspectives on Corneal Transplantation. Clin Ophthalmol 2022; 16:631-646. [PMID: 35282172 PMCID: PMC8904759 DOI: 10.2147/opth.s289359] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Accepted: 01/20/2022] [Indexed: 12/18/2022] Open
Abstract
Disease of the cornea is the third leading cause of blindness worldwide. Corneal graft surgery is one of the most successful forms of solid organ transplantations in humans, with ever-increasing developments in surgical technique. To date, approximately 4504 corneal transplants are performed in the United Kingdom each year. While full thickness transplantation was the most commonly performed keratoplasty over the last few decades, selective lamellar transplantation of the diseased layers of the cornea has been universally adopted. This comprehensive review aims to provide an updated synthesis on different types of corneal transplantations, their treatment outcomes, and the associated complications of each procedure in both adult and paediatric population. In addition, we also present an up-to-date summary of the emerging therapeutic approaches that have the potential to reduce the demand for donor-dependent keratoplasty.
Collapse
Affiliation(s)
- Siyin Liu
- Manchester Royal Eye Hospital, Manchester University NHS Foundation Trust, Manchester, UK
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Yee Ling Wong
- Manchester Royal Eye Hospital, Manchester University NHS Foundation Trust, Manchester, UK
| | - Andrew Walkden
- Manchester Royal Eye Hospital, Manchester University NHS Foundation Trust, Manchester, UK
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Correspondence: Andrew Walkden, Email
| |
Collapse
|
6
|
Mousa HM, Saban DR, Perez VL. The cornea IV immunology, infection, neovascularization, and surgery chapter 1: Corneal immunology. Exp Eye Res 2021; 205:108502. [PMID: 33607075 PMCID: PMC8462940 DOI: 10.1016/j.exer.2021.108502] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 02/07/2021] [Accepted: 02/10/2021] [Indexed: 12/27/2022]
Abstract
PURPOSE of Review: This review offers an informed and up-to-date insight on the immune profile of the cornea and the factors that govern the regulation of such a unique immune environment. SUMMARY The cornea is a unique tissue that performs the specialized task of allowing light to penetrate for visual interpretation. To accomplish this, the ocular surface requires a distinct immune environment that is achieved through unique structural, cellular and molecular factors. Not only must the cornea be able to fend off invasive infectious agents but also control the inflammatory response as to avoid collateral, and potentially blinding damage; particularly of post-mitotic cells such as the corneal endothelium. To combat infections, both innate and adaptive arms of the inflammatory immune response are at play in the cornea. Dendritic cells play a critical role in coordinating both these responses in order to fend off infections. On the other side of the spectrum, the ocular surface is also endowed with a variety of anatomic and physiologic components that aid in regulating the immune response to prevent excessive, potentially damaging, inflammation. This attenuation of the immune response is termed immune privilege. The balance between pro and anti-inflammatory reactions is key for preservation of the functional integrity of the cornea. RECENT FINDINGS The understanding of the molecular and cellular factors governing corneal immunology and its response to antigens is a growing field. Dendritic cells in the normal cornea play a crucial role in combating infections and coordinating the inflammatory arms of the immune response, particularly through coordination with T-helper cells. The role of neuropeptides is recently becoming more highlighted with different factors working on both sides of the inflammatory balance.
Collapse
Affiliation(s)
- Hazem M Mousa
- Foster Center for Ocular Immunology at Duke Eye Center, Duke University Medical Center, Durham, NC, USA
| | - Daniel R Saban
- Foster Center for Ocular Immunology at Duke Eye Center, Duke University Medical Center, Durham, NC, USA; Department of Immunology, Duke University School of Medicine, Durham, NC, USA
| | - Victor L Perez
- Foster Center for Ocular Immunology at Duke Eye Center, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
7
|
Inomata T, Fujimoto K, Okumura Y, Zhu J, Fujio K, Shokirova H, Miura M, Okano M, Funaki T, Sung J, Negishi N, Murakami A. Novel immunotherapeutic effects of topically administered ripasudil (K-115) on corneal allograft survival. Sci Rep 2020; 10:19817. [PMID: 33188243 PMCID: PMC7666179 DOI: 10.1038/s41598-020-76882-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 10/22/2020] [Indexed: 02/07/2023] Open
Abstract
Corneal allograft survival is mediated by the variety of immunological reactions and wound healing process. Our aim was to explore the effects of topical administration of ripasudil, a selective Rho-associated coiled-coil protein kinase inhibitor, on corneal allograft survival. Ripasudil was administered to mice thrice a day after allogeneic corneal transplantation. Corneal graft survival, opacity, neovascularization, re-epithelization, immune cell infiltration, and mRNA levels of angiogenic and pro-inflammatory factors in the grafted cornea and draining lymph nodes (dLNs) were evaluated with slit-lamp microscopy, immunohistochemistry, flow cytometry, and polymerase chain reaction. Graft survival was significantly prolonged with lower graft opacity and neovascularization scores in 0.4% and 2.0% ripasudil-treated groups, and mRNA levels of angiogenic and pro-inflammatory factors in ripasudil-treated grafted corneas were reduced. Moreover, 0.4% and 2.0% ripasudil reduced CD45+-infiltrated leukocyte frequency, Cd11b and Cd11c mRNA levels, and the frequencies of mature dendritic cells, IFNγ-, and IL-17- producing CD4+T cells in the dLNs of recipients. Re-epithelization rate of the grafted cornea was significantly higher in the 0.4% and 2.0% ripasudil groups than in the control. Topically applied ripasudil prolonged graft survival by downregulating neovascularization and inflammation factors, while promoting corneal re-epithelization, suggesting that ripasudil may be useful for suppressing immunological rejection in corneal transplantation.
Collapse
Affiliation(s)
- Takenori Inomata
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan. .,Department of Ophthalmology, Juntendo University Faculty of Medicine, Tokyo, Japan. .,Department of Strategic Operating Room Management and Improvement, Juntendo University Graduate School of Medicine, Tokyo, Japan. .,Department of Hospital Administration, Juntendo University Graduate School of Medicine, Tokyo, Japan. .,Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan.
| | - Keiichi Fujimoto
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.,Department of Ophthalmology, Juntendo University Faculty of Medicine, Tokyo, Japan.,Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yuichi Okumura
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.,Department of Ophthalmology, Juntendo University Faculty of Medicine, Tokyo, Japan.,Department of Strategic Operating Room Management and Improvement, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Jun Zhu
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Kenta Fujio
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.,Department of Ophthalmology, Juntendo University Faculty of Medicine, Tokyo, Japan.,Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hurramhon Shokirova
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Maria Miura
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.,Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Mikiko Okano
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Toshinari Funaki
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.,Department of Ophthalmology, Japanese Red Cross Medical Center, Tokyo, Japan
| | - Jaemyoung Sung
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.,Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Naoko Negishi
- Atopy (Allergic) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Department of Indoor Environment Neurophysiology Research, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Akira Murakami
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.,Department of Ophthalmology, Juntendo University Faculty of Medicine, Tokyo, Japan.,Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
8
|
Schultheiss M, Voykov B, Klemm M, Gross U, Schultheiss HP, Spitzer MS, Casagrande M. Scleral Inflammation around Collector Channels in Eyes with Primary Open-Angle Glaucoma. Ocul Immunol Inflamm 2020; 29:1338-1344. [DOI: 10.1080/09273948.2020.1746355] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Affiliation(s)
- Maximilian Schultheiss
- Department of Ophthalmology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Bogomil Voykov
- Centre of Ophthalmology, University Eye Hospital Tübingen, Tübingen, Germany
| | - Maren Klemm
- Department of Ophthalmology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Ulrich Gross
- Institute for Cardiac Diagnostics and Therapy (IKDT), Berlin, Germany
| | | | - Martin S. Spitzer
- Department of Ophthalmology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Maria Casagrande
- Department of Ophthalmology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| |
Collapse
|
9
|
Vanathi M, Shukla R, Balakrishnan P, Dwivedi R, Gupta N, Tandon R. Evaluation of thrombospondin-1 gene polymorphisms in corneal allograft rejection in Asian Indian patients. Indian J Ophthalmol 2020; 68:565-572. [PMID: 32174570 PMCID: PMC7210848 DOI: 10.4103/ijo.ijo_552_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Purpose: To evaluate the frequency and the association of Thrombospondin 1 (THBS1) gene single nucleotide polymorphisms (SNPs) in Asian Indian patients with optical full thickness corneal grafting surgery. Methods: Prospective case–control analysis of optical penetrating keratoplasty patients with and without immune rejection and controls for genotyping of 3 THBS1 gene SNPs (rs1478604 A>G; rs2228261 C>T; rs2228262 A>G) by Amplification Refractory Mutation System-Polymerase Chain Reaction (ARMS PCR). Results: Among 58 patients [45 with immune allograft rejection (DNA isolation was possible in 38 samples) and 13 without immune corneal allograft rejection] and 65 controls, allele frequencies observed for rs1478604 (A>G) are A: 69.7% and 72.6%, G: 30.2% and 27.3%; for rs2228261 (C>T) are T: 70.2% and 62.3%, C: 29.7% and 37.6%; and for rs2228262 (A>G) A: 97.4% and 98.4%; G 2.5% and 1.5% respectively. Genotype frequencies were rs1478604 (A>G) AA: 57.8% and 59.3%, AG 23.6% and 26.5%; GG 18.4% and 14%; for rs2228261 (C>T) TT: 40.5% and 33.8%, TC: 59% and 56.9%, CC: 0% and 9.2%; for rs2228262 (A>G) AA: 94.8% and 96.8%, AG: 5.1% and 3.1% in rejection and controls respectively. The allele and genotype frequency for the 3 described THSB1 SNPs did not show any difference between the corneal graft immune rejection patients and controls. Conclusion: Asian Indian population evaluated for THBS1 gene SNPs by ARMS PCR genotyping in Asian Indian population did not show any genetic association to immune rejection occurrence in our study.
Collapse
Affiliation(s)
- Murugesan Vanathi
- Cornea, Ocular Surface, Cataract and Refractive Services, Dr R P Centre for Ophthalmic Sciences, New Delhi, India
| | - Rashmi Shukla
- Department of Pediatric Genetics, All India Institute of Medical Sciences, New Delhi, India
| | - Prahlad Balakrishnan
- Department of Pediatric Genetics, All India Institute of Medical Sciences, New Delhi, India
| | - Roopa Dwivedi
- Cornea, Ocular Surface, Cataract and Refractive Services, Dr R P Centre for Ophthalmic Sciences, New Delhi, India
| | - Noopur Gupta
- Cornea, Ocular Surface, Cataract and Refractive Services, Dr R P Centre for Ophthalmic Sciences, New Delhi, India
| | - Radhika Tandon
- Cornea, Ocular Surface, Cataract and Refractive Services, Dr R P Centre for Ophthalmic Sciences, New Delhi, India
| |
Collapse
|
10
|
Molter Y, Milioti G, Langenbucher A, Seitz B. Zeitpunkt, Rezidive und Prognose der immunologischen Abstoßungsreaktion nach Keratoplastik. Ophthalmologe 2019; 117:548-556. [DOI: 10.1007/s00347-019-00975-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
11
|
Schlereth SL, Karlstetter M, Hos D, Matthaei M, Cursiefen C, Heindl LM. Detection of Pro- and Antiangiogenic Factors in the Human Sclera. Curr Eye Res 2018; 44:172-184. [PMID: 30358460 DOI: 10.1080/02713683.2018.1540704] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE Avascular tissues can be used to identify antilymph- or antihemangiogenic factors. The human sclera-the outer covering layer of the eye, lacks lymphatic vessels and contains only a superficial network of blood vessels and was used here to identify endogenous antiangiogenic factors. METHODS Expression levels of a panel of 96 known pro- and antiangiogenic factors were analyzed in 12 scleral or conjunctival control samples from normal human donors using real-time PCR. In vitro, scleral homogenate was cocultured with blood- and lymphatic endothelial cells (BECs and LECs) and immunohistochemistry was performed of scleral fibroblasts and BECs. RESULTS Three antiangiogenic factors were significantly upregulated in the human sclera compared to the conjunctiva, including FBLN5 (fibulin 5), SERPINF1 (serpin peptidase inhibitor, clade F, member 1 = pigment epithelium derived factor) and TIMP2 (Tissue inhibitor of metalloproteinases 2). Six proangiogenic factors were significantly downregulated in the sclera, including FLT4 (Fms-related tyrosine kinase 4=VEGF-R3), HGF (hepatocyte growth factor), KIT (CD117 / c-kit), PROX1 (prospero homeobox 1), SEMA3F (semaphorin-3F) and TGFA (transforming growth factor alpha). In vitro, scleral homogenate inhibited the growth of both BECs and LECs. Immunohistochemistry labeling of three major antiangiogenic factors from scleral tissue confirmed TIMP3 and PEDF expression both in scleral fibroblasts and in blood endothelial cells, whereas TIMP2 was not detectable. CONCLUSION Balancing anti- and proangiogenic factors actively regulates human scleral avascularity, inhibits endothelial cell growth in vitro, and thus may help maintaining the vascular privilege of the inner eye.
Collapse
Affiliation(s)
- Simona L Schlereth
- a Department of Ophthalmology , University of Cologne , Cologne , Germany
| | - Marcus Karlstetter
- a Department of Ophthalmology , University of Cologne , Cologne , Germany
| | - Deniz Hos
- a Department of Ophthalmology , University of Cologne , Cologne , Germany
| | - Mario Matthaei
- a Department of Ophthalmology , University of Cologne , Cologne , Germany
| | - Claus Cursiefen
- a Department of Ophthalmology , University of Cologne , Cologne , Germany
| | - Ludwig M Heindl
- a Department of Ophthalmology , University of Cologne , Cologne , Germany
| |
Collapse
|
12
|
Fine Needle-Diathermy Regresses Pathological Corneal (Lymph)Angiogenesis and Promotes High-Risk Corneal Transplant Survival. Sci Rep 2018; 8:5707. [PMID: 29632336 PMCID: PMC5890271 DOI: 10.1038/s41598-018-24037-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 03/19/2018] [Indexed: 01/23/2023] Open
Abstract
Pathological corneal hem- and lymphangiogenesis are prime risk factors for corneal graft rejection. Fine needle-diathermy (FND) is an option to regress corneal blood vessels; however, whether this treatment besides clinically visible blood vessels also affects invisible lymphatic vessels is so far unknown. Here we test the hypothesis that FND destroys not only blood but also lymphatic vessels, thereby promotes corneal high-risk graft survival. The effect of FND was studied in vivo using BALB/c mice and the model of suture-induced corneal neovascularization. Mice were divided into three groups: FND, ANTI (anti-inflammatory therapy) and NON (control). Five, 7, 10 and 20 days after cauterization, corneas were harvested and stained with LYVE-1, CD31 to quantify (lymph)angiogenesis. The long-term survival of allografts was compared between the three groups. FND caused significant regression of both blood and lymphatic vessels compared to the control group at all time points (p < 0.05) with the most obvious effect at day 7 (p < 0.01). Graft survival was significantly prolonged when transplants were placed into the FND pretreated group (p < 0.0001). The effect of the anti-inflammatory therapy alone was less effective compared to FND (p < 0.05). This novel lymphangioregressive effect of FND can be used clinically to precondition high-risk recipients to promote graft survival.
Collapse
|
13
|
Liu S, Romano V, Steger B, Kaye SB, Hamill KJ, Willoughby CE. Gene-based antiangiogenic applications for corneal neovascularization. Surv Ophthalmol 2018; 63:193-213. [DOI: 10.1016/j.survophthal.2017.10.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 10/09/2017] [Accepted: 10/12/2017] [Indexed: 12/22/2022]
|
14
|
Eslani M, Putra I, Shen X, Hamouie J, Tadepalli A, Anwar KN, Kink JA, Ghassemi S, Agnihotri G, Reshetylo S, Mashaghi A, Dana R, Hematti P, Djalilian AR. Cornea-Derived Mesenchymal Stromal Cells Therapeutically Modulate Macrophage Immunophenotype and Angiogenic Function. Stem Cells 2018; 36:775-784. [PMID: 29341332 DOI: 10.1002/stem.2781] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2017] [Revised: 01/02/2018] [Accepted: 01/08/2018] [Indexed: 12/12/2022]
Abstract
Macrophages are crucial drivers of inflammatory corneal neovascularization and thus are potential targets for immunomodulatory therapies. We hypothesized that therapeutic use of cornea-derived mesenchymal stromal cells (cMSCs) may alter the function of macrophages. We found that cMSCs can modulate the phenotype and angiogenic function of macrophages. In vitro, cMSCs induce apoptosis of macrophages while preferentially promoting a distinct CD14hi CD16hi CD163hi CD206hi immunophenotype that has significantly reduced angiogenic effects based on in vitro angiogenesis assays. In vivo, application of cMSCs to murine corneas after injury leads to reduced macrophage infiltration and higher expression of CD206 in macrophages. Macrophages cocultured ("educated") by cMSCs express significantly higher levels of anti-angiogenic and anti-inflammatory factors compared with control macrophages. In vivo, injured corneas treated with cMSC-educated macrophages demonstrate significantly less neovascularization compared with corneas treated with control macrophages. Knocking down the expression of pigment epithelial derived factor (PEDF) in cMSCs significantly abrogates its modulating effects on macrophages, as shown by the reduced rate of apoptosis, decreased expression of sFLT-1/PEDF, and increased expression of vascular endothelial growth factor-A in the cocultured macrophages. Similarly, cMSCs isolated from PEDF knockout mice are less effective compared with wild-type cMSCs at inhibiting macrophage infiltration when applied to wild-type corneas after injury. Overall, these results demonstrate that cMSCs therapeutically suppress the angiogenic capacity of macrophages and highlight the role of cMSC secreted PEDF in the modulation of macrophage phenotype and function. Stem Cells 2018;36:775-784.
Collapse
Affiliation(s)
- Medi Eslani
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Ilham Putra
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Xiang Shen
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Judy Hamouie
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Asha Tadepalli
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Khandaker N Anwar
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, Illinois, USA
| | - John A Kink
- Department of Medicine and University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Samaneh Ghassemi
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Gaurav Agnihotri
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Sofiya Reshetylo
- Department of Medicine and University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Alireza Mashaghi
- Faculty of Mathematics and Natural Sciences, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Reza Dana
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, USA
| | - Peiman Hematti
- Department of Medicine and University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Ali R Djalilian
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, Illinois, USA
| |
Collapse
|
15
|
Abstract
The eye has become a useful site for the investigation and understanding of local and systemic immune responses. The ease of access and transparency of the cornea permits direct visualization of ocular structures, blood vessels, and lymphatic vessels, allowing for the tracking of normal and pathological biological processes in real time. As a window to the immune system, we have used the eye to dissect the mechanisms of corneal inflammatory reactions that include innate and adaptive immune responses. We have identified that the ocular microenvironment regulates these immune responses by recruiting different populations of inflammatory cells to the cornea through local production of selected chemokines. Moreover, crosstalk between T cells and macrophages is a common and crucial step in the development of ocular immune responses to corneal alloantigens. This review summarizes the data generated by our group using intravital fluorescent confocal microscopy to capture the tempo, magnitude, and function of innate and adaptive corneal immune responses.
Collapse
Affiliation(s)
- Victor L Perez
- *the Department of Ophthalmology, Bascom Palmer Eye Institute; and †the Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL
| |
Collapse
|
16
|
INOMATA TAKENORI. A New Immunotherapy Using Regulatory T-Cells for High-Risk Corneal Transplantation. JUNTENDO MEDICAL JOURNAL 2017. [DOI: 10.14789/jmj.63.2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- TAKENORI INOMATA
- Department of Ophthalmology, Juntendo University Faculty of Medicine
| |
Collapse
|
17
|
Inomata T, Hua J, Di Zazzo A, Dana R. Impaired Function of Peripherally Induced Regulatory T Cells in Hosts at High Risk of Graft Rejection. Sci Rep 2016; 6:39924. [PMID: 28008995 PMCID: PMC5180229 DOI: 10.1038/srep39924] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 11/29/2016] [Indexed: 12/27/2022] Open
Abstract
Regulatory T cells (Tregs) are crucial for allograft survival. Tregs can be divided into thymus-derived natural Tregs (tTregs) and peripherally-derived induced Tregs (pTregs). Here, we determine whether the suppressive function of Treg subsets is hampered in hosts who are at high risk for rejecting their graft. To induce graft beds that promote high risk of transplant rejection, intrastromal corneal sutures were placed two weeks prior to the transplant procedure in mice. We demonstrate that in high-risk recipients the frequencies and function of pTregs (but not tTregs) are suppressed. Reduced function of pTregs correlated with decreased expression of CTLA-4, interleukin-10, and transforming growth factor-β. Adoptive transfer of pTregs from mice at low risk of subsequent graft rejection is able to rescue graft survival in recipients that are at high risk of rejecting their grafts. Our data suggest that impaired function of pTregs, but not tTregs, mediates the loss of immune tolerance and promotes allograft rejection.
Collapse
Affiliation(s)
- Takenori Inomata
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Jing Hua
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Antonio Di Zazzo
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Reza Dana
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
18
|
In Vivo Expansion of Regulatory T Cells by Low-Dose Interleukin-2 Treatment Increases Allograft Survival in Corneal Transplantation. Transplantation 2016; 100:525-32. [PMID: 26881788 DOI: 10.1097/tp.0000000000001044] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
BACKGROUND Corneal allograft survival dramatically decreases in hosts with inflamed or vascularized recipient beds. We have previously shown that in rejected corneal allografts regulatory T cells (Treg) demonstrate diminished Foxp3 expression and immunoregulatory function. Treatment with low doses of IL-2 selectively expands Treg and has been proposed for the treatment of autoimmune diseases. In this study, we investigated the effect of low-dose IL-2 administration on Treg function and corneal allograft survival. METHODS Allogeneic corneal transplantation was performed on inflamed host beds. Low-dose systemic IL-2 was administered starting 3 days before grafting until 6 weeks after transplantation. Frequencies of Treg and their immunosuppressive function and antigen specificity were assessed using flow cytometry, in vitro proliferation assays, and adoptive transfer experiments. Frequencies of effector T cells (Teff) and graft infiltrating immune cells were measured at 2 weeks posttransplantation. Long-term allograft survival was evaluated for up to 9 weeks using Kaplan-Meier survival analysis. RESULTS Treatment with low-dose IL-2 significantly increased frequencies of CD4CD25Foxp3 Treg and their immunosuppressive function. It also suppressed alloimmune response as shown by the decreased CD4 IFNγ T cell frequencies and graft infiltration of CD45 and CD4 cells. Clinical evaluation of the grafts showed significant improvement in long-term corneal allograft survival in the IL-2 treated group compared with controls. CONCLUSIONS Our study is the first to report that treatment with low-dose IL-2 increases survival of corneal allografts. We propose that IL-2-mediated Treg expansion can be an effective tool to prevent alloimmunity and to improve long-term allograft survival in transplantation.
Collapse
|
19
|
Schlereth SL, Kremers S, Cursiefen C, Heindl LM. Using a Laminating Technique to Perform Confocal Microscopy of the Human Sclera. J Vis Exp 2016. [PMID: 27213987 DOI: 10.3791/53920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
The sclera is a dense connective tissue that covers and protects the eye. It mainly consists of dense collagen bundles (types I, III, IV, V, VI, and VII). Due to its autofluorescence, opaqueness, and thickness, it has not been found suitable for confocal microscopy. An alternative approach to the one presented here, which uses formalin-fixed sclera embedded in paraffin for immunohistochemistry, has technical challenges, especially when preheating the tissue for antigen retrieval. Since the sclera is relatively poor in both cells and vessels, the use of larger tissue samples was explored to help prevent overlooking cells and to understand their localization in relation to vessels and other anatomical sites. To allow for the analysis of larger tissue samples under the confocal microscope, a laminating technique was performed to create thin layers from the sclera. Following the analysis of results of CD31 blood vessels and lymphatic vessel endothelial hyaluronan receptor 1 (LYVE1) positive cells, for which approval for scientific examination was obtained, the advantages and limitations of this method are discussed.
Collapse
|
20
|
Yu T, Rajendran V, Griffith M, Forrester JV, Kuffová L. High-risk corneal allografts: A therapeutic challenge. World J Transplant 2016; 6:10-27. [PMID: 27011902 PMCID: PMC4801785 DOI: 10.5500/wjt.v6.i1.10] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 10/03/2015] [Accepted: 12/04/2015] [Indexed: 02/05/2023] Open
Abstract
Corneal transplantation is the most common surgical procedure amongst solid organ transplants with a high survival rate of 86% at 1-year post-grafting. This high success rate has been attributed to the immune privilege of the eye. However, mechanisms originally thought to promote immune privilege, such as the lack of antigen presenting cells and vessels in the cornea, are challenged by recent studies. Nevertheless, the immunological and physiological features of the cornea promoting a relatively weak alloimmune response is likely responsible for the high survival rate in “low-risk” settings. Furthermore, although corneal graft survival in “low-risk” recipients is favourable, the prognosis in “high-risk” recipients for corneal graft is poor. In “high-risk” grafts, the process of indirect allorecognition is accelerated by the enhanced innate and adaptive immune responses due to pre-existing inflammation and neovascularization of the host bed. This leads to the irreversible rejection of the allograft and ultimately graft failure. Many therapeutic measures are being tested in pre-clinical and clinical studies to counter the immunological challenge of “high-risk” recipients. Despite the prevailing dogma, recent data suggest that tissue matching together with use of systemic immunosuppression may increase the likelihood of graft acceptance in “high-risk” recipients. However, immunosuppressive drugs are accompanied with intolerance/side effects and toxicity, and therefore, novel cell-based therapies are in development which target host immune cells and restore immune homeostasis without significant side effect of treatment. In addition, developments in regenerative medicine may be able to solve both important short comings of allotransplantation: (1) graft rejection and ultimate graft failure; and (2) the lack of suitable donor corneas. The advances in technology and research indicate that wider therapeutic choices for patients may be available to address the worldwide problem of corneal blindness in both “low-risk” and “high-risk” hosts.
Collapse
|
21
|
Schöllhorn L, Bock F, Cursiefen C. Thrombospondin-1 as a Regulator of Corneal Inflammation and Lymphangiogenesis: Effects on Dry Eye Disease and Corneal Graft Immunology. J Ocul Pharmacol Ther 2015; 31:376-85. [PMID: 26154823 DOI: 10.1089/jop.2015.0020] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Thrombospondin-1 (TSP-1) is a matricellular glycoprotein that belongs to a family of evolutionary highly conserved calcium-binding proteins consisting of 5 members (TSP-1-TSP-5). In the eye, TSP-1 is expressed by several ocular cell types and is also detectable in the aqueous humor and the vitreous body. So far, TSP-1 is one of the major activators of TGFβ, suggesting a strong influence on various important cellular functions and interactions such as differentiation, migration, and wound healing. TSP-1 is also a key endogenous inhibitor of hem- and lymphangiogenesis. Several lines of evidence indicate a crucial role of TSP-1 in maintaining the ocular immune and angiogenic privilege, for example, by regulating T lymphocytes and the tolerance-promoting properties of ocular antigen-presenting cells. This review discusses the role of TSP-1 in dry eye disease and corneal graft rejection through its effects on hem- and lymphangiogenesis, as well as on the underlying immune responses. Recent work will be reviewed showing by which molecular mechanism TSP-1 modulates inflammatory processes during ocular diseases. This opens potential new treatment avenues in inflammatory and (lymph)angiogenic ocular diseases.
Collapse
Affiliation(s)
- Laura Schöllhorn
- Department of Ophthalmology, University of Cologne , Cologne, Germany
| | - Felix Bock
- Department of Ophthalmology, University of Cologne , Cologne, Germany
| | - Claus Cursiefen
- Department of Ophthalmology, University of Cologne , Cologne, Germany
| |
Collapse
|
22
|
Antilymphangiogenic therapy to promote transplant survival and to reduce cancer metastasis: what can we learn from the eye? Semin Cell Dev Biol 2014; 38:117-30. [PMID: 25460541 DOI: 10.1016/j.semcdb.2014.11.003] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 10/01/2014] [Accepted: 11/12/2014] [Indexed: 12/22/2022]
Abstract
The lymphatic vasculature is - amongst other tasks - essentially involved in inflammation, (auto)immunity, graft rejection and cancer metastasis. The eye is mainly devoid of lymphatic vessels except for its adnexa, the conjunctiva and the limbus. However, several pathologic conditions can result in the secondary ingrowth of lymphatic vessels into physiologically alymphatic parts of the eye such as the cornea or the inner eye. Therefore, the cornea has served as an excellent in vivo model system to study lymphangiogenesis, and findings from such studies have substantially contributed to the understanding of central principles of lymphangiogenesis also with relevance outside the eye. Grafting experiments at the cornea have been extensively used to analyze the role of lymphangiogenesis in transplant immunology. In this regard, we recently demonstrated the crucial role of lymphatic vessels in mediating corneal allograft rejection and could show that antilymphangiogenic therapy increases graft survival. In the field of cancer research, we recently detected tumor-associated lymphangiogenesis in the most common malignant tumors of the eye, such as conjunctival carcinoma and melanoma, and ciliochoroidal melanoma with extraocular extension. These neolymphatics correlate with an increased risk of local recurrence, metastasis and tumor related death, and may offer potential therapeutic targets for the treatment of these tumors. This review will focus on corneal and tumor-associated ocular lymphangiogenesis. First, we will describe common experimentally used corneal lymphangiogenesis models and will recapitulate recent findings regarding the involvement of lymphatic vessels in corneal diseases and transplant immunology. The second part of this article will summarize findings about the participation of tumor-associated lymphangiogenesis in ocular malignancies and their implications for the development of future therapeutic strategies.
Collapse
|
23
|
[Decellularized collagen matrix from tilapia fish scales for corneal reconstruction (BioCornea)]. Ophthalmologe 2014; 111:1027-32. [PMID: 25337678 DOI: 10.1007/s00347-013-3011-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
BACKGROUND The worldwide need for donor corneal tissue clearly exceeds the availability of transplantable human tissue; therefore, recent efforts aim to identify and characterize alternative tissues, such as decellularized collagen scaffolds. OBJECTIVES The transparent fish scales of tilapia (Oreochromis mossambicus) were analyzed as a potential alternative for corneal reconstruction ("BioCornea"). MATERIAL AND METHODS The article gives a review of the literature and own preliminary results. After decellularization the tissue characteristics of the fish scales, the repopulation with corneal epithelium and stromal cells, immunogenicity, the feasibility of corneal transplantation and the angiogenic properties were analyzed in vitro and in various animal models. RESULTS The fish scales mainly consist of collagen type I and show an architecture that is similar to the human cornea. Corneal epithelium and stromal cells are able to grow over and into the scaffold. It is possible to transplant fish scales in various animal models without severe inflammatory responses. Furthermore, in mice, less blood and lymphatic vessels grow into the xenograft when compared to conventional allogenic transplants. CONCLUSION Preliminary results with decellularized tilapia fish scales as an alternative for corneal reconstruction ("BioCornea") are promising.
Collapse
|
24
|
Kymionis GD, Mikropoulos DG, Portaliou DM, Boboridis KG, Voudouragkaki IC, Dragoumis ND, Konstas AGP. New perspectives on lamellar keratoplasty. Adv Ther 2014; 31:494-511. [PMID: 24846543 DOI: 10.1007/s12325-014-0121-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Indexed: 11/28/2022]
Abstract
Lamellar (anterior and posterior) keratoplasty entails the surgical replacement of diseased-only corneal tissue, while healthy host corneal tissue is preserved. Selective keratoplasty offers several advantages in comparison to penetrating keratoplasty such as a lower rate of graft rejection, less endothelial cell loss, faster/superior visual rehabilitation and enhanced resistance to closed injury. The surgical approach of "partial corneal transplantation" may be divided into anterior and posterior: techniques including superficial and deep anterior lamellar keratoplasty (SALK and DALK, respectively) and endothelial keratoplasty as well as Descemet stripping automated endothelial keratoplasty (DSAEK) and Descemet membrane endothelial keratoplasty (DMEK). These novel surgical procedures are rapidly becoming the preferred therapy option for specific corneal dysfunctions involving the corneal stroma (SALK, DALK), or corneal endothelium (DSAEK, DMEK). During the past decade, the continuing advancement of surgical techniques and the development of innovative surgical instruments have significantly enhanced corneal transplantation. Lamellar keratoplasty techniques facilitate corneal surgery, provide patients with superior outcomes and can successfully restore vision in corneal-related blindness. Nevertheless, more long-term evidence is needed to better evaluate these promising new techniques.
Collapse
Affiliation(s)
- George D Kymionis
- Vardinoyiannion Eye Institute of Crete (VEIC), Faculty of Medicine, University of Crete, Heraklion, Crete, Greece
| | | | | | | | | | | | | |
Collapse
|
25
|
Faraj LA, Elalfy MS, Said DG, Dua HS. Fine needle diathermy occlusion of corneal vessels. Br J Ophthalmol 2014; 98:1287-90. [DOI: 10.1136/bjophthalmol-2014-304891] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
|
26
|
Chauhan SK, Dohlman TH, Dana R. Corneal Lymphatics: Role in Ocular Inflammation as Inducer and Responder of Adaptive Immunity. ACTA ACUST UNITED AC 2014; 5. [PMID: 25580370 PMCID: PMC4287999 DOI: 10.4172/2155-9899.1000256] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The normal cornea is devoid of lymphatic and blood vessels, thus suppressing both the afferent (lymphatic) and efferent (vascular) arms of the immune response–contributing to its ‘immune privilege’. Inflammation, however, negates this unique ‘immune’ and ‘angiogenic’ privilege of the cornea. Abnormal blood vessel growth from pre-existing limbal vessels into the cornea has been studied for many years, but it is only recently that the significance of new lymphatic vessels (lymphangiogenesis) in ocular inflammatory diseases has been demonstrated. Whereas blood vessels in inflamed ocular surface provide a route of entry for immune effector cells to the cornea, lymphatics facilitate the exit of antigen-presenting cells and antigenic material from the cornea to regional lymph nodes, thus promoting induction of adaptive immune response. This review summarizes the current evidence for lymphangiogenesis in the cornea, and describes its molecular mediators; and discusses the interface between corneal lymphangiogenesis and adaptive immunity. Furthermore, the pathophysiologic implications of corneal lymphangiogenesis in the setting of allo- and autoimmune-mediated corneal inflammation are discussed.
Collapse
Affiliation(s)
- Sunil K Chauhan
- Schepens Eye Research Institute and Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, Massachusetts 02114, USA
| | - Thomas H Dohlman
- Schepens Eye Research Institute and Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, Massachusetts 02114, USA
| | - Reza Dana
- Schepens Eye Research Institute and Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, Massachusetts 02114, USA
| |
Collapse
|
27
|
Takiyama N, Mizuno T, Iwai R, Uechi M, Nakayama Y. In-body tissue-engineered collagenous connective tissue membranes (BIOSHEETs) for potential corneal stromal substitution. J Tissue Eng Regen Med 2013; 10:E518-E526. [PMID: 24668614 DOI: 10.1002/term.1859] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Revised: 09/07/2013] [Accepted: 11/10/2013] [Indexed: 01/15/2023]
Abstract
There is a severe shortage of donor cornea for transplantation in many countries. Collagenous connective tissue membranes, named BIOSHEETs, grown in vivo were successfully implanted in rabbit corneal stroma for in vivo evaluation of their suitability as a corneal stromal substitute to solve this global donor shortage. BIOSHEETs were prepared by embedding silicone moulds into dorsal subcutaneous pouches in rabbits for 1 month and stored in glycerol. After re-swelling in saline and trephining, disk-shaped BIOSHEETs (4 mm diameter) were allogeneically implanted into stromal pockets prepared in the right cornea of seven rabbits. Clinical tests for corneal thickness and transparency, and tissue analyses were performed. Because the BIOSHEETs (thickness, 131 ± 14 µm) obtained were opaque immediately after implantation, the transparency of the cornea decreased. The total thickness of the BIOSHEET-implanted cornea increased from 364 ± 21.0 µm to 726 ± 131 µm. After 4 weeks' implantation, the thickness of the cornea stabilized (493 ± 80 µm at 4 weeks and 447 ± 46 µm at 8 weeks). The transparency of the cornea increased progressively with time of implantation. The random orientation of collagen fibrils in the original BIOSHEETs tended to be homogeneous, similar to that of the native stroma. No inflammatory cells accumulated and fibroblast-like cells infiltrated the implant. The BIOSHEETs showed high biocompatibility with stromal tissues; however, further studies are needed to test its functional aspects. Although this research is only intended as a proof of concept, BIOSHEETs may be considered a feasible corneal stromal replacement, especially for treating visual impairment caused by stromal haze. Copyright © 2013 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Naoaki Takiyama
- Department of Veterinary Medicine, College of Bioresource Sciences, Nihon University, Kanagawa, Japan
| | - Takeshi Mizuno
- Department of Veterinary Medicine, College of Bioresource Sciences, Nihon University, Kanagawa, Japan.,Division of Medical Engineering and Materials, National Cerebral and Cardiovascular Centre Research Institute, Osaka, Japan
| | - Ryosuke Iwai
- Division of Medical Engineering and Materials, National Cerebral and Cardiovascular Centre Research Institute, Osaka, Japan
| | - Masami Uechi
- Department of Veterinary Medicine, College of Bioresource Sciences, Nihon University, Kanagawa, Japan.,Division of Medical Engineering and Materials, National Cerebral and Cardiovascular Centre Research Institute, Osaka, Japan
| | - Yasuhide Nakayama
- Division of Medical Engineering and Materials, National Cerebral and Cardiovascular Centre Research Institute, Osaka, Japan.
| |
Collapse
|
28
|
Heindl LM, Bucher F, Naumann GOH, Cursiefen C. Lack of ciliary body lymphatics in iridociliary melanocytoma. Graefes Arch Clin Exp Ophthalmol 2013; 252:169-71. [PMID: 24213841 DOI: 10.1007/s00417-013-2510-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Revised: 10/18/2013] [Accepted: 10/22/2013] [Indexed: 02/01/2023] Open
Affiliation(s)
- Ludwig M Heindl
- Department of Ophthalmology, University Hospital of Cologne, Kerpener Strasse 62, 50924, Cologne, Germany,
| | | | | | | |
Collapse
|
29
|
Abstract
Corneal transplantation is the most commonly performed organ transplantation. Immune privilege of the cornea is widely recognized, partly because of the relatively favorable outcome of corneal grafts. The first-time recipient of corneal allografts in an avascular, low-risk setting can expect a 90% success rate without systemic immunosuppressive agents and histocompatibility matching. However, immunologic rejection remains the major cause of graft failure, particularly in patients with a high risk for rejection. Corticosteroids remain the first-line therapy for the prevention and treatment of immune rejection. However, current pharmacological measures are limited in their side-effect profiles, repeated application, lack of targeted response, and short duration of action. Experimental ocular gene therapy may thus present new horizons in immunomodulation. From efficient viral vectors to sustainable alternative splicing, we discuss the progress of gene therapy in promoting graft survival and postulate further avenues for gene-mediated prevention of allogeneic graft rejection.
Collapse
Affiliation(s)
- Yureeda Qazi
- Cornea and Refractive Surgery Service, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, USA
| | - Pedram Hamrah
- Cornea and Refractive Surgery Service, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, USA
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
30
|
Ling S, Li W, Liu L, Zhou H, Wang T, Ye H, Liang L, Yuan J. Allograft survival enhancement using doxycycline in alkali-burned mouse corneas. Acta Ophthalmol 2013; 91:e369-78. [PMID: 23387987 DOI: 10.1111/aos.12070] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
PURPOSE To explore the inhibitory effects of doxycycline on allograft rejection in alkali-burned cornea beds. METHODS The corneas of BALB/c mice were injured using a 1 mol/l NaOH solution. Following the injury, the corneas from C57BL/6 mice were transplanted into the eyes of BALB/c mice after being randomized into three groups: allogeneic corneal transplantation (group A), topical use of doxycycline after allogeneic corneal transplantation (group B) and syngeneic corneal transplantation (group C). Corneal angiogenesis was examined using whole-mount immunofluorescence, and corneal inflammation was evaluated using inflammation index scoring. The immune rejection of the grafts was examined using a slit lamp. In addition, the expression of vascular endothelial growth factor A and interleukin-1β in the transplanted corneas was examined using a real-time polymerase chain reaction, immunohistochemistry and an enzyme-linked immunosorbent assay. RESULTS The outgrowth of the corneal blood vessels in the group A mice was faster than that in the group B and group C mice. The inflammation index levels were highest in the group A mice, intermediate in the group B mice and lowest in the group C mice. Vascular endothelial growth factor and the interleukin-1β protein and mRNA levels decreased dramatically in the group B mice compared with the group A mice (all p-values < 0.01). In addition, the mean survival time in the group B mice (27.00 ± 2.00 days) was significantly longer than that in the group A mice (11.67 ± 1.51 days; p < 0.05). CONCLUSIONS Doxycycline may have had a significant role in preventing corneal angiogenesis and inflammation in alkali-burned corneal beds, which resulted in higher allograft survival rates.
Collapse
Affiliation(s)
- Shiqi Ling
- Department of Ophthalmology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Effect of glucocorticoid (triamcinolone acetonide) pretreatment in a murine penetrating keratoplasty and suture model. Cornea 2013; 31:1468-75. [PMID: 22677639 DOI: 10.1097/ico.0b013e3182473356] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
PURPOSE To evaluate the effect of glucocorticoid (triamcinolone acetonide injectable suspension) pretreatment on corneal neovascularization, lymphangiogenesis, and inflammation in a murine penetrating keratoplasty (PK) and corneal suture model. METHODS For the PK model, BALB/c mice were used as recipients and C57BL/6 mice were used as donors. A group pretreated with subconjunctival glucocorticoid and a combination of post-subconjunctival and topical glucocorticoids (group I) was compared with two groups that did not receive glucocorticoid pretreatment [one group received a combination of subconjunctival and topical glucocorticoids postoperatively (group II) and the other group received only topical glucocorticoid treatment postoperatively (group III)]. All groups were treated with subconjunctival glucocorticoid on the day of surgery. For the corneal suture model, BALB/c mice were used. A group receiving only pre-suture glucocorticoid treatment (group A) and a group receiving only post-suture glucocorticoid treatment (group C) were compared with a control group that did not receive glucocorticoid therapy (group B). The degree of neovascularization, lymphangiogenesis, and inflammatory infiltration was compared in each of these models. RESULTS In the PK model, the group receiving glucocorticoid pretreatment (group I) showed less neovascularization compared with the posttreatment-only groups (group II, P=0.043; group III, P=0.020) and less lymphangiogenesis compared with group III (P=0.005). In the corneal suture model, the glucocorticoid pretreatment group showed a similar level of neovascularization, lymphangiogenesis, and inflammatory infiltration as the posttreatment-only groups (P>0.05). CONCLUSIONS Glucocorticoid pretreatment before PK decreases neovascularization and lymphangiogenesis compared with posttransplant glucocorticoid treatment alone.
Collapse
|
32
|
Thanos S, Gatzioufas Z, Schallenberg M, König S, Meyer-Rüsenberg HW, Busse H. Clinical Transplantation of Individualized Recipient Serum-Adapted Cornea Reduces the Risk of Graft Rejection after Keratoplasty. Cell Transplant 2013; 22:477-91. [DOI: 10.3727/096368912x657459] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Corneal diseases cause severe visual impairment that necessitates corneal transplantation and frequently repetitive procedures due to graft rejection. We tested the hypothesis that exposure of donor corneas to recipient serum-derived factors during eye banking triggers a preoperative adaptation that is beneficial for postoperative tolerance. Donor corneas were incubated in a medium containing human serum (HS) obtained in each case from the prospective graft recipient in order to individually expose the donor cornea to the recipient's serum. All recipient serum-adapted corneas (RSACs) fulfilled the clinical criteria required by the national law and were transplanted successfully. The postoperative ophthalmological examination extended up to 8 years. All RSACs were tolerated by their recipients and did not cause postoperative complications and no rejection. Proteomic analysis of corneas cultivated in culture medium containing either fetal calf serum (FCS) that is routinely used for cornea banking or HS revealed different patterns of proteins. HS-cultured corneas showed a greater proteomic similarity with native human corneas than did the FCS-cultured corneas, indicating a differential nutrification of the cultured corneal tissue by HS-derived factors. The clinical results show for the first time that postoperative complications such as tissue intolerance and graft rejection might be managed if the corneal tissue is individually adapted to the recipient's serum trophic factors. This new donor tissue treatment procedure offers incontrovertible advantages and could be adapted for low-risk eyes as well as other transplantable tissues.
Collapse
Affiliation(s)
- Solon Thanos
- Institute of Experimental Ophthalmology, School of Medicine, University of Münster, Münster, Germany
| | - Zissis Gatzioufas
- Institute of Experimental Ophthalmology, School of Medicine, University of Münster, Münster, Germany
| | - Maurice Schallenberg
- Institute of Experimental Ophthalmology, School of Medicine, University of Münster, Münster, Germany
| | - Simone König
- Integrated Functional Genomics (IFG), Interdisciplinary Center for Clinical Research, Medical Faculty, Westphalian Wilhelms-University, Münster, Germany
| | | | - Holger Busse
- University Eye Hospital, Clinical Department, Münster, Germany
| |
Collapse
|
33
|
Bock F, Maruyama K, Regenfuss B, Hos D, Steven P, Heindl LM, Cursiefen C. Novel anti(lymph)angiogenic treatment strategies for corneal and ocular surface diseases. Prog Retin Eye Res 2013; 34:89-124. [PMID: 23348581 DOI: 10.1016/j.preteyeres.2013.01.001] [Citation(s) in RCA: 130] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Revised: 12/17/2012] [Accepted: 01/04/2013] [Indexed: 12/21/2022]
Abstract
The cornea is one of the few tissues which actively maintain an avascular state, i.e. the absence of blood and lymphatic vessels (corneal [lymph]angiogenic privilege). Nonetheless do several diseases interfere with this privilege and cause pathologic corneal hem- and lymphangiogenesis. The ingrowths of pathologic blood and lymphatic vessels into the cornea not only reduce transparency and thereby visual acuity up to blindness, but also significantly increases the rate of graft rejections after subsequent corneal transplantation. Therefore great interest exists in new strategies to target pathologic corneal (lymph)angiogenesis to promote graft survival. This review gives an overview on the vascular anatomy of the normal ocular surface, on the molecular mechanisms contributing to the corneal (lymph)angiogenic privilege and on the cellular and molecular mechanisms occurring during pathological neovascularization of the cornea. In addition we summarize the current preclinical and clinical evidence for three novel treatment strategies against ocular surface diseases based on targeting pathologic (lymph)angiogenesis: (a) modulation of the immune responses after (corneal) transplantation by targeting pathologic (lymph)angiogenesis prior to and after transplantation, (b) novel concepts against metastasis and recurrence of ocular surface tumors such as malignant melanoma of the conjunctiva by anti(lymph)angiogenic therapy and (c) new ideas on how to target ocular surface inflammatory diseases such as dry eye by targeting conjunctival and corneal lymphatic vessels. Based on compelling preclinical evidence and early data from clinical trials the novel therapeutic concepts of promoting graft survival, inhibiting tumor metastasis and dampening ocular surface inflammation and dry eye disease by targeting (lymph)angiogenesis are on their way to translation into the clinic.
Collapse
Affiliation(s)
- Felix Bock
- Department of Ophthalmology, University of Cologne, Cologne, Germany
| | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
Corneal transplantation is among the most successful solid organ transplants. However, despite low rejection rates of grafts in the ‘low-risk’ setting, rejection can be as high as 70% when grafted into ‘high-risk’ recipient beds. Under normal homeostatic conditions, the avascular cornea provides a unique environment that facilitates immune and angiogenic privilege. An imbalance in pro-inflammatory, angiogenic and lymphangiogenic mediators leads to a breakdown in corneal immune privilege with a consequent host response against the donor graft. Recent developments in lamellar and endothelial keratoplasties have reduced the rates of graft rejection even more, while providing improved visual outcomes. The corneal layer against which an immune response is initiated, largely determines reversibility of the acute episode. While epithelial and stromal graft rejection may be treated with topical corticosteroids with higher success, acute endothelial rejection mandates a more aggressive approach to therapy due to the lack of regenerative capacity of this layer. However, current immunosuppressive regimens come with the caveat of ocular and systemic side effects, making prolonged aggressive treatment undesirable. With the advent of biologics, efficacious therapies with a superior side effect profile are on the horizon. In our review we discuss the mediators of ocular immune privilege, the roles of cellular and molecular immune players in graft rejection, with a focus on human leukocyte antigen and antigen presenting cells. Furthermore, we discuss the clinical risk factors for graft rejection and compare rates of rejection in lamellar and endothelial keratoplasties to traditional penetrating keratoplasty. Lastly, we present the current and upcoming measures of therapeutic strategies to manage and treat graft rejection, including an overview of biologics and small molecule therapy.
Collapse
Affiliation(s)
- Yureeda Qazi
- Ocular Surface and Imaging Center & Cornea Service Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Pedram Hamrah
- Ocular Surface and Imaging Center & Cornea Service Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA ; Schepens Eye Research Institute, Massachusetts Eye & Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
35
|
Sel S, Schlaf G, Schurat O, Altermann WW. A novel ELISA-based crossmatch procedure to detect donor-specific anti-HLA antibodies responsible for corneal allograft rejections. J Immunol Methods 2012; 381:23-31. [DOI: 10.1016/j.jim.2012.04.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Revised: 04/05/2012] [Accepted: 04/11/2012] [Indexed: 10/28/2022]
|
36
|
Zhang H, Grimaldo S, Yuen D, Chen L. Combined blockade of VEGFR-3 and VLA-1 markedly promotes high-risk corneal transplant survival. Invest Ophthalmol Vis Sci 2011; 52:6529-35. [PMID: 21715348 DOI: 10.1167/iovs.11-7454] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
PURPOSE. High-risk corneal transplantation refers to grafting performed on inflamed and highly vascularized host beds. It represents a clinical dilemma because the rejection rate can be as high as 90%, irrespective of current treatment modalities. This study was conducted to investigate whether combined blockade of VEGFR-3 (vascular endothelial growth factor receptor-3) and VLA-1 (very late antigen-1) promotes high-risk transplant survival and how it correlates with corneal lymphangiogenesis and hemangiogenesis before and after transplantation. METHODS. High-risk corneal transplantation was performed between normal C57BL/6 (donor) and inflamed BALB/c (recipient) mice. The recipients were randomized to receive intraperitoneal injections of VEGFR-3 and VLA-1-neutralizing antibodies or their controls twice a week for up to 8 weeks after transplantation. Corneal grafts were evaluated by ophthalmic slit-lamp biomicroscopy and analyzed by Kaplan-Meier survival curve. Additionally, whole-mount corneas before and after transplantation were examined by immunofluorescent microscopic assays, and the correlation between lymphatic or blood vessel distribution and transplant outcome was analyzed. RESULTS. The combined blockade markedly promotes 90% survival of high-risk transplants. This strategy specifically modified host beds by selective inhibition of lymphangiogenesis but not hemangiogenesis. A strong correlation was also identified between high-risk transplant rejection and severe lymphatic invasion reaching the donor-graft border. CONCLUSIONS. These novel findings not only provide a new and potentially powerful strategy to promote high-risk transplant survival, they also confirm a critical role of high-degree lymphangiogenesis in mediating high-risk transplant rejection. Results from this study may also shed new light on our understanding and management of other lymphatic- and immune-related diseases in general.
Collapse
Affiliation(s)
- Hui Zhang
- Center for Eye Disease and Development, Program in Vision Science and School of Optometry, University of California, Berkeley, USA
| | | | | | | |
Collapse
|
37
|
Jin Y, Chauhan SK, El Annan J, Annan JEI, Sage PT, Sharpe AH, Dana R. A novel function for programmed death ligand-1 regulation of angiogenesis. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 178:1922-9. [PMID: 21435468 DOI: 10.1016/j.ajpath.2010.12.027] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 05/07/2010] [Revised: 11/15/2010] [Accepted: 12/17/2010] [Indexed: 02/01/2023]
Abstract
Programmed death ligand-1 (PD-L1) plays a critical role in T-cell regulatory function. Here, we report a newly discovered effect of PD-L1 on angiogenesis. We demonstrate that PD-L1 and its receptor CD80, but not PD-1, are expressed by primary murine lung and heart vascular endothelial cells and the miscrovascular endothelial cell line (MS1) at both the mRNA and protein levels in vitro. The inhibition of PD-L1 or CD80 expression in MS1 cells, by small-interfering RNA transfection, led to a significant up-regulation of vascular endothelial growth factor receptor 2 expression and cell proliferation levels in MS1 cells. Furthermore, MS1 cells were found to have a significantly lower proliferation and vascular endothelial growth factor receptor 2 expression levels when they were co-cultured with PD-L1-expressing normal corneal epithelial cells, as compared to MS1 cells co-cultured with PD-L1(-/-) corneal epithelial cells. In a suture-induced corneal angiogenesis model, we observed a significantly higher level of angiogenic response in PD-L1(-/-) knockout mice as compared to wild-type mice, although there was no significant difference in the expression of inflammatory cytokines (interleukin-1α, interleukin-1β, or tumor necrosis factor-α) or the infiltration of innate immune cells (neutrophils and macrophages) between the two groups. We conclude that the expression of PD-L1 in both vascular endothelial cells and corneal epithelial cells regulates corneal angiogenesis.
Collapse
Affiliation(s)
- Yiping Jin
- Department of Ophthalmology, Schepens Eye Research Institute and Massachusetts Eye and Ear Infirmary, Boston, MA 02114, USA
| | | | | | | | | | | | | |
Collapse
|
38
|
Gharebaghi R, Heidary F, Heidary R. Possible contribution of intraocular tumour-associated lymphangiogenesis to the novel uveolymphatic pathway. Med Hypotheses 2011; 76:144. [DOI: 10.1016/j.mehy.2010.08.031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2010] [Revised: 08/11/2010] [Accepted: 08/12/2010] [Indexed: 11/25/2022]
|
39
|
Intraocular Tumor-Associated Lymphangiogenesis. Ophthalmology 2010; 117:334-42. [DOI: 10.1016/j.ophtha.2009.06.057] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2009] [Revised: 06/15/2009] [Accepted: 06/23/2009] [Indexed: 11/24/2022] Open
|
40
|
|
41
|
Zhang X, Shen L, Jin Y, Saban DR, Chauhan SK, Dana R. Depletion of passenger leukocytes from corneal grafts: an effective means of promoting transplant survival? Invest Ophthalmol Vis Sci 2009; 50:3137-44. [PMID: 19136708 PMCID: PMC2739689 DOI: 10.1167/iovs.08-1899] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
PURPOSE To develop and compare effective strategies for depleting graft-derived passenger leukocytes that include antigen-presenting cells from corneal buttons and to assess the effectiveness of this strategy in promoting graft survival using a high-risk (HR) model of corneal transplantation. METHODS Corneal buttons harvested from C57BL/6 mice were used in three ex vivo strategies of passenger leukocyte depletion. Two strategies involved storage in medium at different temperatures for prolonged periods. A third strategy used complement-dependent cytotoxicity (CDC) by treating the buttons with anti-CD45 mAb plus complement. Wholemount corneal buttons or cells from enzyme-digested corneas were analyzed using confocal microscopy or flow cytometry, respectively, for the pan-leukocyte surface marker CD45. HR host beds were created and used to evaluate the efficacy of passenger leukocyte depletion on transplant survival. RESULTS Passenger leukocyte numbers in the buttons were significantly reduced by all three treatments. CDC was the most efficient strategy for passenger leukocyte depletion with 39% reduction (P < 0.00005) of CD45(+) cells, and negligible damage to the endothelial layer, achievable within 24 hours. However, passenger leukocyte depletion failed to improve HR graft longevity. CONCLUSIONS Anti-CD45 antibody plus complement-mediated targeting of donor tissue is the most efficient way to deplete corneal passenger leukocytes and can considerably reduce the time required for cell depletion. However, depletion of graft passenger leukocytes does not have a significant effect on promoting graft survival even in the HR setting.
Collapse
Affiliation(s)
- Xian Zhang
- Schepens Eye Research Institute, Boston, Massachusetts
- Massachusetts Eye and Ear Infirmary, Boston, Massachusetts
- Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Linling Shen
- Schepens Eye Research Institute, Boston, Massachusetts
- Massachusetts Eye and Ear Infirmary, Boston, Massachusetts
- Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Yiping Jin
- Schepens Eye Research Institute, Boston, Massachusetts
- Massachusetts Eye and Ear Infirmary, Boston, Massachusetts
- Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Daniel R. Saban
- Schepens Eye Research Institute, Boston, Massachusetts
- Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Sunil K. Chauhan
- Schepens Eye Research Institute, Boston, Massachusetts
- Massachusetts Eye and Ear Infirmary, Boston, Massachusetts
- Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Reza Dana
- Schepens Eye Research Institute, Boston, Massachusetts
- Massachusetts Eye and Ear Infirmary, Boston, Massachusetts
- Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
42
|
Chong EM, Dana MR. Graft failure IV. Immunologic mechanisms of corneal transplant rejection. Int Ophthalmol 2008; 28:209-22. [PMID: 17673946 DOI: 10.1007/s10792-007-9099-9] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2007] [Accepted: 05/08/2007] [Indexed: 12/16/2022]
Abstract
Corneal transplantation is the oldest and the most common form of solid tissue transplantation in humans. Immunologic graft rejection is one of the main causes of short and long-term graft failure. Rejection involves donor tissue recognition and destruction by allo-specific immune cells of the recipient. This review outlines (1) the immunobiology of transplantation, with reference to ocular immune privilege, (2) factors that confer "high-risk" status to a graft and (3) the pathophysiologic mechanisms of corneal transplant rejection.
Collapse
Affiliation(s)
- Eva-Marie Chong
- Cornea Service, Massachusetts Eye and Ear Infirmary and Department of Ophthalmology, Harvard Medical School, 243 Charles Street, Boston, MA 02114, USA
| | | |
Collapse
|
43
|
Lu P, Li L, Kuno K, Wu Y, Baba T, Li YY, Zhang X, Mukaida N. Protective roles of the fractalkine/CX3CL1-CX3CR1 interactions in alkali-induced corneal neovascularization through enhanced antiangiogenic factor expression. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2008; 180:4283-4291. [PMID: 18322241 DOI: 10.4049/jimmunol.180.6.4283] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Macrophages accumulate during the course of corneal neovascularization, but its mechanisms and roles still remain elusive. To address these points, we herein examined corneal neovascularization after alkali injury in mice deficient in fractalkine receptor/CX3CR1, which is normally expressed by macrophages. After alkali injury, the mRNA expression of CX3CR1 was augmented along with accumulation of F4/80-positive macrophages and Gr-1-positive neutrophils in the corneas. Compared with wild-type mice, CX3CR1-deficient mice exhibited enhanced corneal neovascularization 2 wk after injury, as evidenced by enlarged CD31-positive areas. Concomitantly, the accumulation of F4/80-positive macrophages, but not Gr-1-positive neutrophils, was markedly attenuated in CX3CR1-deficient mice compared with wild-type mice. The intraocular mRNA expression of vascular endothelial growth factor (VEGF) was enhanced to similar extents in wild-type and CX3CR1-deifient mice after the injury. However, the mRNA expression of antiangiogenic factors, thrombospondin (TSP) 1, TSP-2, and a disintegrin and metalloprotease with thrombospondin (ADAMTS) 1, was enhanced to a greater extent in wild-type than CX3CR1-deificient mice. A double-color immunofluorescence analysis demonstrated that F4/80-positive cells also expressed CX3CR1 and ADAMTS-1 and that TSP-1 and ADAMTS-1 were detected in CX3CR1-positive cells. CX3CL1 enhanced TSP-1 and ADAMTS-1, but not VEGF, expression by peritoneal macrophages. Moreover, topical application of CX3CL1 inhibited corneal neovascularization at 2 wk, along with enhanced intraocular expression of TSP-1 and ADAMTS-1 but not VEGF. Thus, these observations indicate that accumulation of CX3CR1-positive macrophages intraocularly can dampen alkali-induced corneal neovascularization by producing antiangiogenic factors such as TSP-1 and ADAMTS-1 and suggest the potential therapeutic efficacy of using CX3CL1 against alkali-induced corneal neovascularization.
Collapse
Affiliation(s)
- Peirong Lu
- Clinical Immunology Key Laboratory of Jiangsu Province, First Affiliated Hospital of Suzhou University, Suzhou, China
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Birnbaum F, Schwartzkopff J, Scholz C, Reis A, Reinhard T. The new malononitrilamide immunosuppressant FK778 prolongs corneal allograft survival in the rat keratoplasty model. Eye (Lond) 2007; 21:1516-23. [PMID: 17401326 DOI: 10.1038/sj.eye.6702727] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
PURPOSE Aim of this study was to prove the efficacy and safety of the new malononitrilamide immunosuppressive FK778 in prolonging clear graft survival following allogeneic orthotopic keratoplasty in rats. METHODS Sixty-seven penetrating keratoplasties were performed using Fisher and Lewis rats as donors and recipients, respectively: group 1 (n=11), allogeneic control without therapy; group 2 (n=12), syngeneic control; group 3 (n=11), mycophenolate mofetil (MMF) 40 mg/kg bodyweight; group 4 (n=12), FK778 5 mg/kg bodyweight; group 5 (n=12), FK778 10 mg/kg bodyweight; and group 6 (n=9), FK778 20 mg/kg bodyweight. Four animals in each group were killed for immunohistological evaluation on day 14. Therapy was administered orally for 18 days. The grafts were evaluated every three days by means of a scoring system including opacity, oedema, and vascularization. Time to rejection was analysed with the Kaplan-Meier survival analysis and compared with the log-rank test. The densities of infiltrating immune cells were compared statistically using the non-parametric Mann-Whitney test. RESULTS Mean rejection-free graft survival was 11.4 days in group 1 (allogeneic control), 100 days (total follow-up time) in group 2 (syngeneic control), 24.0 days in group 3 (MMF 40 mg/kg), 15.7 days in group 4 (FK778 5 mg/kg), 19.1 days in group 5 (FK778 10 mg/kg), and 25.4 days in group 6 (FK778 20 mg/kg) (P<0.005). CONCLUSIONS Systemic immunosuppression with FK778 prolongs graft survival in the rat keratoplasty model. FK778's efficacy is comparable with that of MMF in preventing immunologic graft rejection.
Collapse
Affiliation(s)
- F Birnbaum
- Eye Hospital, Albert-Ludwigs University, Killianstr. 5, Freiburg, Germany.
| | | | | | | | | |
Collapse
|
45
|
Cursiefen C, Maruyama K, Jackson DG, Streilein JW, Kruse FE. Time course of angiogenesis and lymphangiogenesis after brief corneal inflammation. Cornea 2006; 25:443-7. [PMID: 16670483 DOI: 10.1097/01.ico.0000183485.85636.ff] [Citation(s) in RCA: 135] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE To study the time course of angiogenesis and lymphangiogenesis in the cornea after a short inflammatory insult. This might be helpful for the timing of corneal transplantation in high-risk eyes. METHODS The mouse model of suture-induced inflammatory corneal neovascularization was used. After placement of 3 interrupted 11-0 sutures into the corneal stroma of BALB/c mice (left in place for 14 days), corneas were excised 2, 3, 5, 7, 14, and 21 days as well as 1, 2, 3, 6, and 8 months after surgery. Hem- and lymphangiogenesis were evaluated using double immunohistochemistry of corneas with CD31/PECAM1 as panendothelial and LYVE-1 as lymphatic endothelial marker. RESULTS Both blood and lymphatic vessels grew into the cornea as early as day 2 after suture placement. The outgrowth was initially parallel. Hem- and lymphangiogenesis peaked around day 14. Thereafter, both vessel types started to regress. Regression of lymphatic vessels started earlier and was more pronounced than that of blood vessels. Whereas at 6 and 8 months (partly) perfused CD31+++/LYVE-1(-) blood vessels and (nonperfused) ghost vessels could still be observed, there were no CD31+/LYVE-1+++ lymphatic vessels detectable beyond 6 months after this short inflammation. CONCLUSIONS After a temporary inflammatory insult to the cornea, there is initially parallel outgrowth of both blood and lymphatic vessels. But thereafter, lymphatic vessels regress earlier than blood vessels and are completely regressed by 6 months. Earlier regression of pathologic corneal lymph versus blood vessels suggests that corneal graft survival in high-risk eyes might best be delayed for a prolonged interval following an inflammatory insult.
Collapse
Affiliation(s)
- Claus Cursiefen
- Department of Ophthalmology, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany.
| | | | | | | | | |
Collapse
|
46
|
Maruyama K, Ii M, Cursiefen C, Jackson DG, Keino H, Tomita M, Van Rooijen N, Takenaka H, D'Amore PA, Stein-Streilein J, Losordo DW, Streilein JW. Inflammation-induced lymphangiogenesis in the cornea arises from CD11b-positive macrophages. J Clin Invest 2005; 115:2363-72. [PMID: 16138190 PMCID: PMC1193872 DOI: 10.1172/jci23874] [Citation(s) in RCA: 540] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2004] [Accepted: 06/07/2005] [Indexed: 12/12/2022] Open
Abstract
In the inflamed cornea, there is a parallel outgrowth of blood and lymphatic vessels into the normally avascular cornea. We tested whether adaptive and/or innate immune cells were actively involved in the genesis of new lymphatic vessels. Our results indicate that innate immune cells (CD11b+ macrophages, but not CD11c+ dendritic cells) physically contributed to lymphangiogenesis under pathological conditions and that bone marrow-derived CD11b+ macrophages expressed lymphatic endothelial markers such as LYVE-1 and Prox-1 under inflamed conditions in the corneal stromata of mice. Furthermore, blood vascular endothelial cells that expressed the Tie2 promoter did not contribute to newly formed lymphatic vessels under inflamed conditions. Our in vitro experiments demonstrated that CD11b+ macrophages alone were capable of forming tube-like structures that expressed markers of lymphatic endothelium such as LYVE-1 and podoplanin. The novel finding that CD11b+ macrophages are critical for the development of inflammation-dependent lymphangiogenesis in the eye suggests a new mechanism of lymphangiogenesis.
Collapse
Affiliation(s)
- Kazuichi Maruyama
- Ocular Immunology Group, The Schepens Eye Research Institute, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts 02114, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Yamada J. Thiol redox and immune regulation in corneal transplantation. Cornea 2005; 24:S59-S65. [PMID: 16227826 DOI: 10.1097/01.ico.0000178734.50544.82] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Penetrating keratoplasty (PKP) is the most common type of clinical grafting performed in humans. Although PKP has emerged as the most successful form of transplantation, PKP in "high-risk" eyes shows high incidence of allograft rejection. The incidence of epithelial rejection after limbal transplantation (LT) is extremely higher and swifter than PKP rejection, and even intensive systemic immunosuppressive therapy is often of no avail. Because failure of corneal grafts is an important cause of blindness, developing new strategies for suppressing graft rejection is a worthy goal for research. Corneal allograft rejection is mainly mediated by the TH1-type immune response, which leads to a delayed-type hypersensitivity reaction. Because the TH2-type immune response regulates the TH1-type immune response, we have successfully elicited allograft survival after both PKP and LT by inducing systemic TH2-type immune responses. Because intracellular thiol redox status of antigen-presenting cells (APC) reportedly regulates TH1/TH2 balance via distinctive cytokine production by APC, we also investigated the effect of modulating macrophage intracellular thiol redox status on corneal allograft survival. These strategies are quite effective in major histocompatibility complex (MHC) matching in mice, although it is believed that MHC matching has no effect on corneal allograft survival according to many rodent studies. Recently, many laboratories are reconsidering HLA matching for allograft survival in human corneal transplantation. It may be possible that MHC matching improves corneal allograft survival in the context of TH1 suppression. We propose that the suppression of the TH1-type immune response and MHC matching together may promote allograft survival in humans.
Collapse
Affiliation(s)
- Jun Yamada
- Department of Ophthalmology, Meiji University of Oriental Medicine, Kyoto, Japan.
| |
Collapse
|
48
|
Reinhard T, Mayweg S, Sokolovska Y, Seitz B, Mittelviefhaus H, Engelmann K, Voiculescu A, Godehardt E, Sundmacher R. Systemic mycophenolate mofetil avoids immune reactions in penetrating high-risk keratoplasty: preliminary results of an ongoing prospectively randomized multicentre study. Transpl Int 2005; 18:703-8. [PMID: 15910297 DOI: 10.1111/j.1432-2277.2005.00126.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Recently, in a monocentre study mycophenolate mofetil (MMF) was demonstrated to be efficacious and safe in penetrating high-risk keratoplasty. Here, preliminary results of a randomized multicentre trial are presented. To date, 86 of 140 scheduled patients undergoing high-risk penetrating keratoplasty have already been randomized into the two study groups: 48 into the MMF group and 38 into the control group. All 86 patients received fluocortolon 1 mg/kg body weight/day, tapered within 3 weeks, and topical prednisolone acetate 1% tapered within 5 months. MMF was administered at a daily oral dose of 2 x 1000 mg for the first 6 postoperative months. Thereafter, MMF was tapered within 2 weeks. The proportion of grafts with immune reactions and side-effects were the main outcome measures. Within an average follow up of 9.2 +/- 6.6 months two patients developed reversible endothelial immune reactions in the MMF group after cessation of MMF application. In the control group, five reversible and three irreversible immune reactions were observed within an average follow up of 10.1 +/- 7.6 months. According to Kaplan and Meier analysis, the ratio of grafts without immune reactions was estimated 89% 1 year postoperatively in the MMF group, in contrast to only 67% in the control group (P = 0.03; log-rank test). Fifteen patients experienced side-effects, especially gastroenterotoxicity, tachycardia, arthralgia or systemic infections. All attributable side-effects were reversible. Systemic MMF may be an effective and safe immune modulating drug in the prophylaxis of immune reactions after penetrating high-risk keratoplasty.
Collapse
|
49
|
Yamada J, Hamuro J, Sano Y, Maruyama K, Kinoshita S. Allogeneic Corneal Tolerance in Rodents with Long-Term Graft Survival. Transplantation 2005; 79:1362-9. [PMID: 15912105 DOI: 10.1097/01.tp.0000159869.55962.94] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Healthy C57BL/6 orthotopic corneal allografts in place for more than 8 weeks in BALB/c mice (acceptor8w+) can survive indefinitely due to active suppression of the donor-specific delayed-type hypersensitivity (DTH) response. This suggests a state of tolerance in the acceptor mice, however, the mechanism(s) underlying this acceptance remains to be demonstrated. We investigated the relationship between tolerance-induction and the DTH response using murine re-grafting models to explore the possibility of promoting allogeneic corneal regraft acceptance in high-risk graft beds. METHODS Acceptor8w+ BALB/c mice received C57BL/6- or C3H corneal regrafts onto the same eye. Re-grafting models were prepared by inducing corneal neovascularization in the graft beds of naive BALB/c mice 2 weeks before corneal allografting. These mice were intravenously (iv) injected with purified splenic T cells or T-cell-depleted splenocytes from acceptor8w+ mice at the time they received re-grafts of C57BL/6 corneas. We also iv injected acceptor8w+ splenocytes into mice bearing healthy primary corneal allografts for 4 weeks (acceptor4w) and assessed their DTH response to C57BL/6 alloantigen(s). In those experiments, acceptor4w mice received a C57BL/6 corneal regraft onto the same eye. RESULTS In all acceptor8w+ mice there was indefinite survival of C57BL/6-, but not of C3H regrafts. The iv injection of T cells, but not of T-cell-depleted populations, from acceptor8w+ splenocytes promoted allograft survival. Acceptor4w mice iv injected with acceptor8w+ splenocytes manifested a reduced C57BL/6-specific DTH response and the survival rate of C57BL/6 regrafts was increased from 0% to 87.5%. CONCLUSION As donor-specific T cells from acceptor8w+ mice induced prolonged regraft survival, we posit that the active suppression of DTH responses by T cells may have contributed to indefinite allogeneic regraft survival via the induction of corneal allograft tolerance.
Collapse
Affiliation(s)
- Jun Yamada
- Department of Ophthalmology, Meiji University of Oriental Medicine, Kyoto, Japan.
| | | | | | | | | |
Collapse
|
50
|
Cursiefen C, Chen L, Borges LP, Jackson D, Cao J, Radziejewski C, D'Amore PA, Dana MR, Wiegand SJ, Streilein JW. VEGF-A stimulates lymphangiogenesis and hemangiogenesis in inflammatory neovascularization via macrophage recruitment. J Clin Invest 2004; 113:1040-50. [PMID: 15057311 PMCID: PMC379325 DOI: 10.1172/jci20465] [Citation(s) in RCA: 793] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2003] [Accepted: 01/28/2004] [Indexed: 12/27/2022] Open
Abstract
Lymphangiogenesis, an important initial step in tumor metastasis and transplant sensitization, is mediated by the action of VEGF-C and -D on VEGFR3. In contrast, VEGF-A binds VEGFR1 and VEGFR2 and is an essential hemangiogenic factor. We re-evaluated the potential role of VEGF-A in lymphangiogenesis using a novel model in which both lymphangiogenesis and hemangiogenesis are induced in the normally avascular cornea. Administration of VEGF Trap, a receptor-based fusion protein that binds and neutralizes VEGF-A but not VEGF-C or -D, completely inhibited both hemangiogenesis and the outgrowth of LYVE-1(+) lymphatic vessels following injury. Furthermore, both lymphangiogenesis and hemangiogenesis were significantly reduced in mice transgenic for VEGF-A(164/164) or VEGF-A(188/188) (each of which expresses only one of the three principle VEGF-A isoforms). Because VEGF-A is chemotactic for macrophages and we demonstrate here that macrophages in inflamed corneas release lymphangiogenic VEGF-C/VEGF-D, we evaluated the possibility that macrophage recruitment plays a role in VEGF-A-mediated lymphangiogenesis. Either systemic depletion of all bone marrow-derived cells (by irradiation) or local depletion of macrophages in the cornea (using clodronate liposomes) prior to injury significantly inhibited both hemangiogenesis and lymphangiogenesis. We conclude that VEGF-A recruitment of monocytes/macrophages plays a crucial role in inducing inflammatory neovascularization by supplying/amplifying signals essential for pathological hemangiogenesis and lymphangiogenesis.
Collapse
Affiliation(s)
- Claus Cursiefen
- The Schepens Eye Research Institute, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts 02114, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|