1
|
Jammes M, Tabasi A, Bach T, Ritter T. Healing the Cornea: Exploring the Therapeutic Solutions Offered by MSCs and MSC-derived EVs. Prog Retin Eye Res 2024:101325. [PMID: 39709150 DOI: 10.1016/j.preteyeres.2024.101325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024]
Abstract
Affecting a large proportion of the population worldwide, corneal disorders constitute a concerning health hazard associated to compromised eyesight or blindness for most severe cases. In the last decades, mesenchymal stem/stromal cells (MSCs) demonstrated promising abilities in improving symptoms associated to corneal diseases or alleviating these affections, especially through their anti-inflammatory, immunomodulatory and pro-regenerative properties. More recently, MSC therapeutic potential was shown to be mediated by the molecules they release, and particularly by their extracellular vesicles (EVs; MSC-EVs). Consequently, using MSC-EVs emerged as a pioneering strategy to mitigate the risks related to cell therapy while providing MSC therapeutic benefits. Despite the promises given by MSC- and MSC-EV-based approaches, many improvements are considered to optimize the therapeutic significance of these therapies. This review aspires to provide a comprehensive and detailed overview of current knowledge on corneal therapies involving MSCs and MSC-EVs, the strategies currently under evaluation, and the gaps remaining to be addressed for clinical implementation. From encapsulating MSCs or their EVs into biomaterials to enhance the ocular retention time to loading MSC-EVs with therapeutic drugs, a wide range of ground-breaking strategies are currently contemplated to lead to the safest and most effective treatments. Promising research initiatives also include diverse gene therapies and the targeting of specific cell types through the modification of the EV surface, paving the way for future therapeutic innovations. As one of the most important challenges, MSC-EV large-scale production strategies are extensively investigated and offer a wide array of possibilities to meet the needs of clinical applications.
Collapse
Affiliation(s)
- Manon Jammes
- Regenerative Medicine Institute, School of Medicine, University of Galway, Galway, Ireland
| | - Abbas Tabasi
- Regenerative Medicine Institute, School of Medicine, University of Galway, Galway, Ireland
| | - Trung Bach
- Regenerative Medicine Institute, School of Medicine, University of Galway, Galway, Ireland
| | - Thomas Ritter
- Regenerative Medicine Institute, School of Medicine, University of Galway, Galway, Ireland; CURAM Centre for Research in Medical Devices, University of Galway, Galway, Ireland.
| |
Collapse
|
2
|
Lee M, Elbasiony E, Cho WJ, Pulimamidi VK, Folorunso OS, Mittal SK, Dana R, Chauhan SK. Hepatocyte growth factor upregulates MMP1 and MMP10 expression and resolves corneal fibrosis. Sci Rep 2024; 14:26421. [PMID: 39488561 PMCID: PMC11531534 DOI: 10.1038/s41598-024-75488-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 10/07/2024] [Indexed: 11/04/2024] Open
Abstract
Different therapeutic modalities, including steroids, have been used to treat corneal scarring. However, the ability of steroids to reduce corneal scarring is limited and associated with numerous side effects. Our previous studies have demonstrated that topical hepatocyte growth factor (HGF) after corneal injury suppresses the development of stromal scars. Here, we investigated whether HGF can re-establish corneal clarity and normalize tissue structure in corneas with pre-existing scars. Corneal scarring was induced by mechanically removing the corneal epithelium and the anterior third of the stroma using a hand-held Algerbrush II in C57BL/6 mice. Substantial scar tissue formed by day 10 post-injury, at which time the epithelium was debrided and treated with 0.1% recombinant mouse HGF, 0.1% dexamethasone (steroid) or 0.1% control protein thrice a day for 10 days. Corneal clarity was significantly restored in the HGF treatment group, compared to both the steroid and control protein treatment groups. Moreover, HGF treatment downregulated the expression of αSMA and upregulated the expression of extracellular matrix-remodeling matrix metalloproteinases 1 and 10 (MMP1 and MMP10), suggesting HGF upregulates tissue remodeling molecule MMP1 and 10 to promote tissue restoration. These findings offer novel insights into the mechanisms by which HGF re-establishes corneal clarity, and promotes epithelial regeneration in corneas with pre-existing stromal scarring.
Collapse
Affiliation(s)
- Mingshun Lee
- Schepens Eye Research Institute of Mass Eye & Ear, Harvard Medical School, 20 Staniford Street, Boston, MA, 02114, USA
| | - Elsayed Elbasiony
- Schepens Eye Research Institute of Mass Eye & Ear, Harvard Medical School, 20 Staniford Street, Boston, MA, 02114, USA
| | - Wonkyung J Cho
- Schepens Eye Research Institute of Mass Eye & Ear, Harvard Medical School, 20 Staniford Street, Boston, MA, 02114, USA
| | - Vinay K Pulimamidi
- Schepens Eye Research Institute of Mass Eye & Ear, Harvard Medical School, 20 Staniford Street, Boston, MA, 02114, USA
| | - Olufemi S Folorunso
- Schepens Eye Research Institute of Mass Eye & Ear, Harvard Medical School, 20 Staniford Street, Boston, MA, 02114, USA
| | - Sharad K Mittal
- Schepens Eye Research Institute of Mass Eye & Ear, Harvard Medical School, 20 Staniford Street, Boston, MA, 02114, USA
| | - Reza Dana
- Schepens Eye Research Institute of Mass Eye & Ear, Harvard Medical School, 20 Staniford Street, Boston, MA, 02114, USA
| | - Sunil K Chauhan
- Schepens Eye Research Institute of Mass Eye & Ear, Harvard Medical School, 20 Staniford Street, Boston, MA, 02114, USA.
| |
Collapse
|
3
|
Shen S, Zhang Y. Restoration of corneal epithelial barrier function: A possible target for corneal neovascularization. Ocul Surf 2024; 34:38-49. [PMID: 38901546 DOI: 10.1016/j.jtos.2024.06.003] [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: 03/13/2024] [Revised: 06/10/2024] [Accepted: 06/11/2024] [Indexed: 06/22/2024]
Abstract
Corneal neovascularization (CoNV) is the second leading common cause of vision impairment worldwide and is a blinding pathological alteration brought on by ocular trauma, infection, and other factors. There are some limitations in the treatment of CoNV, hence it's critical to look into novel therapeutic targets. The corneal epithelial barrier, which is the initial barrier of the ocular surface, is an important structure that shields the eye from changes in the internal environment or invasion by the external environment. This study sought to collate evidence on the regulation of corneal epithelial barrier injury on the activation of vascular endothelial cells (VECs), basement membrane (BM) degradation, differentiation, migration, and proliferation of VECs, vascular maturation and stability, and other key processes in CoNV, so as to provide a novel concept for CoNV therapy targeting corneal epithelial barrier repair.
Collapse
Affiliation(s)
- Sitong Shen
- Department of Ophthalmology, The Second Hospital of Jilin University, 218 Ziqiang Street, Nanguan District, Changchun, Jilin Province, 130041, China
| | - Yan Zhang
- Department of Ophthalmology, The Second Hospital of Jilin University, 218 Ziqiang Street, Nanguan District, Changchun, Jilin Province, 130041, China; Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China.
| |
Collapse
|
4
|
Barone V, Scirocco L, Surico PL, Micera A, Cutrupi F, Coassin M, Di Zazzo A. Mast cells and ocular surface: An update review. Exp Eye Res 2024; 245:109982. [PMID: 38942134 DOI: 10.1016/j.exer.2024.109982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 06/10/2024] [Accepted: 06/25/2024] [Indexed: 06/30/2024]
Abstract
Mast cells (MCs), traditionally viewed as key players in IgE-mediated allergic responses, are increasingly recognized for their versatile roles. Situated at critical barrier sites such as the ocular surface, these sentinel cells participate in a broad array of physiological and pathological processes. This review presents a comprehensive update on the immune pathophysiology of MCs, with a particular focus on the mechanisms underlying innate immunity. It highlights their roles at the ocular surface, emphasizing their participation in allergic reactions, maintenance of corneal homeostasis, neovascularization, wound healing, and immune responses in corneal grafts. The review also explores the potential of MCs as therapeutic targets, given their significant contributions to disease pathogenesis and their capacity to modulate immunity. Through a thorough examination of current literature, we aim to elucidate the immune pathophysiology and multifaceted roles of MCs in ocular surface health and disease, suggesting directions for future research and therapeutic innovation.
Collapse
Affiliation(s)
- Vincenzo Barone
- Ophthalmology Campus Bio-Medico University, Rome, Italy; Ophthalmology Operative Complex Unit, Campus Bio-Medico University Hospital Foundation, Rome, Italy
| | - Laura Scirocco
- Ophthalmology Campus Bio-Medico University, Rome, Italy; Ophthalmology Operative Complex Unit, Campus Bio-Medico University Hospital Foundation, Rome, Italy
| | - Pier Luigi Surico
- Ophthalmology Campus Bio-Medico University, Rome, Italy; Ophthalmology Operative Complex Unit, Campus Bio-Medico University Hospital Foundation, Rome, Italy; Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Alessandra Micera
- Research and Development Laboratory for Biochemical, Molecular and Cellular Applications in Ophthalmological Science, IRCCS - Fondazione Bietti, Rome, Italy
| | - Francesco Cutrupi
- Ophthalmology Campus Bio-Medico University, Rome, Italy; Ophthalmology Operative Complex Unit, Campus Bio-Medico University Hospital Foundation, Rome, Italy
| | - Marco Coassin
- Ophthalmology Campus Bio-Medico University, Rome, Italy; Ophthalmology Operative Complex Unit, Campus Bio-Medico University Hospital Foundation, Rome, Italy
| | - Antonio Di Zazzo
- Ophthalmology Campus Bio-Medico University, Rome, Italy; Ophthalmology Operative Complex Unit, Campus Bio-Medico University Hospital Foundation, Rome, Italy; Rare Corneal Diseases Center, Campus Bio-Medico University Hospital Foundation, Rome, Italy.
| |
Collapse
|
5
|
Ji Z, Li J, Tao S, Li H, Kong X, Huang B, Feng Z, Wei X, Zheng Z, Chen J, Chen B, Liu J, Zhao F. Mrgprb2-mediated mast cell activation exacerbates Modic changes by regulating immune niches. Exp Mol Med 2024; 56:1178-1192. [PMID: 38689089 PMCID: PMC11148035 DOI: 10.1038/s12276-024-01230-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 12/03/2023] [Accepted: 02/26/2024] [Indexed: 05/02/2024] Open
Abstract
Modic changes are radiographic features associated with microfracture, low-virulence organism infection and chronic inflammation with inflammatory cell infiltration in the vertebral endplate region. Mast cells, as innate immune cells similar to macrophages, are present in painful degenerated intervertebral discs. However, the involvement and mechanisms of mast cells in the development of Modic changes remain unclear. Herein, we found increased mast cell infiltration in samples from patients with Modic changes and in mouse models of Modic changes. To clarify the role of mast cells in the progression of Modic changes, we used mast cell-deficient (KITW-SH/W-SH) mice to construct a model of Modic changes and found that the severity of Modic changes in KITW-SH/W-SH mice was significantly lower than that in WT mice. These findings were further supported by the use of a mast cell-specific activator (compound 48/80) and a stabilizer (cromolyn). Furthermore, we found that mast cells were not activated via the classic IgE pathway in the Modic change models and that Mrgprb2 is the specific receptor for mast cell activation reported in recent studies. Then, we utilized Mrgprb2 knockout mice to demonstrate that Mrgprb2 knockout inhibited mast cell activation and thus reduced the degree of Modic changes. Transcriptomic sequencing revealed aberrant PI3K-AKT and MAPK pathway activation in the Mrgprb2-deficient mast cells. Additionally, Mrgpbrb2-activated mast cells regulate immune niches by recruiting macrophages, promoting M1 polarization and reducing M2 polarization, thereby promoting the progression of Modic changes. These findings suggest that mast cells may serve as a novel therapeutic target for addressing Modic changes.
Collapse
Affiliation(s)
- Zhongyin Ji
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3, Qingchun Road East, Hangzhou, 310016, P. R. China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, No. 3, Qingchun Road East, Hangzhou, 310016, P. R. China
| | - Jie Li
- Department of Orthopaedic Surgery, Ningbo Medical Center Li Huili Hospital, Ningbo, Zhejiang, China
| | - Siyue Tao
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3, Qingchun Road East, Hangzhou, 310016, P. R. China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, No. 3, Qingchun Road East, Hangzhou, 310016, P. R. China
| | - Hui Li
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3, Qingchun Road East, Hangzhou, 310016, P. R. China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, No. 3, Qingchun Road East, Hangzhou, 310016, P. R. China
| | - Xiangxi Kong
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3, Qingchun Road East, Hangzhou, 310016, P. R. China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, No. 3, Qingchun Road East, Hangzhou, 310016, P. R. China
| | - Bao Huang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3, Qingchun Road East, Hangzhou, 310016, P. R. China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, No. 3, Qingchun Road East, Hangzhou, 310016, P. R. China
| | - Zhenhua Feng
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3, Qingchun Road East, Hangzhou, 310016, P. R. China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, No. 3, Qingchun Road East, Hangzhou, 310016, P. R. China
| | - Xiaoan Wei
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3, Qingchun Road East, Hangzhou, 310016, P. R. China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, No. 3, Qingchun Road East, Hangzhou, 310016, P. R. China
| | - Zeyu Zheng
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3, Qingchun Road East, Hangzhou, 310016, P. R. China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, No. 3, Qingchun Road East, Hangzhou, 310016, P. R. China
| | - Jian Chen
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3, Qingchun Road East, Hangzhou, 310016, P. R. China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, No. 3, Qingchun Road East, Hangzhou, 310016, P. R. China
| | - Binhui Chen
- Department of Orthopaedic Surgery, Ningbo Medical Center Li Huili Hospital, Ningbo, Zhejiang, China.
| | - Junhui Liu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3, Qingchun Road East, Hangzhou, 310016, P. R. China.
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, No. 3, Qingchun Road East, Hangzhou, 310016, P. R. China.
| | - Fengdong Zhao
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3, Qingchun Road East, Hangzhou, 310016, P. R. China.
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, No. 3, Qingchun Road East, Hangzhou, 310016, P. R. China.
| |
Collapse
|
6
|
Mishra N, Kant R, Goswami DG, Petrash JM, Agarwal C, Tewari-Singh N, Agarwal R. Metabolomics for identifying pathways involved in vesicating agent lewisite-induced corneal injury. Exp Eye Res 2023; 236:109672. [PMID: 37797797 PMCID: PMC10843384 DOI: 10.1016/j.exer.2023.109672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 09/11/2023] [Accepted: 10/02/2023] [Indexed: 10/07/2023]
Abstract
Lewisite (LEW) is an arsenical vesicant that can be a potentially dangerous chemical warfare agent (CWA). Eyes are particularly susceptible to vesicant induced injuries and ocular LEW exposure can act swiftly, causing burning of eyes, edema, inflammation, cell death and even blindness. In our previous studies, we developed a LEW exposure-induced corneal injury model in rabbit and showed increased inflammation, neovascularization, cell death, and structural damage to rabbit corneas upon LEW exposure. In the present study, we further assessed the metabolomic changes to delineate the possible mechanisms underlying the LEW-induced corneal injuries. This information is vital and could help in the development of effective targeted therapies against ocular LEW injuries. Thus, the metabolomic changes associated with LEW exposures in rabbit corneas were assessed as a function of time, to delineate pathways from molecular perturbations at the genomic and proteomic levels. New Zealand white rabbit corneas (n = 3-6) were exposed to LEW vapor (0.2 mg/L; flow rate: 300 ml/min) for 2.5 min (short exposure; low dose) or 7.5 min (long-exposure; high dose) and then collected at 1, 3, 7, or 14 days post LEW exposure. Samples were prepared using the automated MicroLab STAR® system, and proteins precipitated to recover the chemically diverse metabolites. Metabolomic analysis was carried out by reverse phase UPLC-MS/MS and gas chromatography (GC)-MS. The data obtained were analyzed using Metabolon's software. The results showed that LEW exposures at high doses were more toxic, particularly at the day 7 post exposure time point. LEW exposure was shown to dysregulate metabolites associated with all the integral functions of the cornea and cause increased inflammation and immune response, as well as generate oxidative stress. Additionally, all important metabolic functions of the cells were also affected: lipid and nucleotide metabolism, and energetics. The high dose LEW exposures were more toxic, particularly at day 7 post LEW exposure (>10-fold increased levels of histamine, quinolinate, N-acetyl-β-alanine, GMP, and UPM). LEW exposure dysregulated integral functions of the cornea, caused inflammation and heightened immune response, and generated oxidative stress. Lipid and nucleotide metabolism, and energetics were also affected. The novel information about altered metabolic profile of rabbit cornea following LEW exposure could assist in delineating complex molecular events; thus, aid in identifying therapeutic targets to effectively ameliorate ocular trauma.
Collapse
Affiliation(s)
- Neha Mishra
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado-Anschutz Medical Campus, Aurora, CO, USA
| | - Rama Kant
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado-Anschutz Medical Campus, Aurora, CO, USA
| | - Dinesh G Goswami
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado-Anschutz Medical Campus, Aurora, CO, USA
| | - J Mark Petrash
- Department of Ophthalmology, School of Medicine, University of Colorado-Anschutz Medical Campus, Aurora, CO, USA
| | - Chapla Agarwal
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado-Anschutz Medical Campus, Aurora, CO, USA
| | - Neera Tewari-Singh
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado-Anschutz Medical Campus, Aurora, CO, USA
| | - Rajesh Agarwal
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado-Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
7
|
Cho WJ, Elbasiony E, Singh A, Mittal SK, Chauhan SK. IL-36γ Augments Ocular Angiogenesis by Promoting the Vascular Endothelial Growth Factor-Vascular Endothelial Growth Factor Receptor Axis. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:1740-1749. [PMID: 36740182 PMCID: PMC10616713 DOI: 10.1016/j.ajpath.2023.01.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 12/05/2022] [Accepted: 01/06/2023] [Indexed: 02/05/2023]
Abstract
Prevention of inflammatory angiogenesis is critical for suppressing chronic inflammation and inhibiting inflammatory tissue damage. Angiogenesis is particularly detrimental to the cornea because pathologic growth of new blood vessels can lead to marked vision impairment and even loss of vision. The expression of proinflammatory cytokines by injured tissues exacerbates the inflammatory cascade, including angiogenesis. IL-36 cytokine, a subfamily of the IL-1 superfamily, consists of three proinflammatory agonists, IL-36α, IL-36β, and IL-36γ, and an IL-36 receptor antagonist (IL-36Ra). Data from the current study indicate that human vascular endothelial cells constitutively expressed the cognate IL-36 receptor. The current investigation, for the first time, characterized the direct contribution of IL-36γ to various angiogenic processes. IL-36γ up-regulated the expression of vascular endothelial growth factors (VEGFs) and their receptors VEGFR2 and VEGFR3 by human vascular endothelial cells, suggesting that IL-36γ mediates the VEGF-VEGFR signaling by endothelial cells. Moreover, by using a naturally occurring antagonist IL-36Ra in a murine model of inflammatory angiogenesis, this study demonstrated that blockade of endogenous IL-36γ signaling results in significant retardation of inflammatory angiogenesis. The current investigation on the proangiogenic function of IL-36γ provides novel evidence of the development of IL-36γ-targeting strategies to hamper inflammatory angiogenesis.
Collapse
Affiliation(s)
- WonKyung J Cho
- Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, Massachusetts
| | - Elsayed Elbasiony
- Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, Massachusetts
| | - Aastha Singh
- Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, Massachusetts
| | - Sharad K Mittal
- Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, Massachusetts
| | - Sunil K Chauhan
- Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
8
|
Singh A, Cho WJ, Pulimamidi VK, Mittal SK, Chauhan SK. Interleukin-11 Suppresses Ocular Surface Inflammation and Accelerates Wound Healing. Invest Ophthalmol Vis Sci 2023; 64:1. [PMID: 37910094 PMCID: PMC10627293 DOI: 10.1167/iovs.64.14.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 09/26/2023] [Indexed: 11/03/2023] Open
Abstract
Purpose Regulation of inflammation is critical for achieving favorable outcomes in wound healing. In this study, we determine the functional role and mechanism of action of IL-11, an immunomodulatory cytokine, in regulating inflammatory response at the ocular surface. Methods Corneal injury was induced by mechanical removal of the epithelium and anterior stroma using an AlgerBrush II. Transcript and protein levels of IL-11 in injured cornea were quantified using real-time PCR and ELISA analysis. Corneal inflammation was assessed by measuring frequencies of total CD45+ inflammatory cells, CD11b+Ly6G+ polymorphonuclear cells (neutrophils), and CD11b+Ly6G- mononuclear cells (macrophages, monocytes) at the ocular surface using flow cytometry. To assess the effect of IL-11 on innate immune cell function, cell activation marker and inflammatory cytokines including major histocompatibility complex (MHC) class II, myeloperoxidase (MPO), TNFα, and inducible nitric oxide synthase (iNOS) were measured following recombinant IL-11 treatment (1 µg/mL). Injured corneas were topically treated with IL-11 (1 µg/mL), and wound healing was evaluated using corneal fluorescein staining. Results Corneal injury resulted in increased levels of IL-11 in the cornea, particularly in the stroma. Neutrophils and CD11b+ mononuclear cells (macrophages, monocytes) substantially expressed IL-11 receptor. Interestingly, IL-11 significantly downregulated the activation of immune cells, as evidenced by the lower expression of MHC II and TNFα by CD11b+ mononuclear cells and lower levels of MPO by neutrophils. Topical administration of IL-11 to injured corneas led to faster wound healing and better retention of tissue architecture. Conclusions Our findings demonstrate IL-11 is a key modulator of ocular surface inflammation and provide novel evidence of IL-11 as a potential therapeutic to control inflammatory damage and accelerate wound repair following injury.
Collapse
Affiliation(s)
- Aastha Singh
- Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, Massachusetts, United States
| | - WonKyung J Cho
- Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, Massachusetts, United States
| | - Vinay K Pulimamidi
- Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, Massachusetts, United States
| | - Sharad K Mittal
- Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, Massachusetts, United States
| | - Sunil K Chauhan
- Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, Massachusetts, United States
| |
Collapse
|
9
|
Clahsen T, Hadrian K, Notara M, Schlereth SL, Howaldt A, Prokosch V, Volatier T, Hos D, Schroedl F, Kaser-Eichberger A, Heindl LM, Steven P, Bosch JJ, Steinkasserer A, Rokohl AC, Liu H, Mestanoglu M, Kashkar H, Schumacher B, Kiefer F, Schulte-Merker S, Matthaei M, Hou Y, Fassbender S, Jantsch J, Zhang W, Enders P, Bachmann B, Bock F, Cursiefen C. The novel role of lymphatic vessels in the pathogenesis of ocular diseases. Prog Retin Eye Res 2023; 96:101157. [PMID: 36759312 DOI: 10.1016/j.preteyeres.2022.101157] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 12/13/2022] [Accepted: 12/17/2022] [Indexed: 02/10/2023]
Abstract
Historically, the eye has been considered as an organ free of lymphatic vessels. In recent years, however, it became evident, that lymphatic vessels or lymphatic-like vessels contribute to several ocular pathologies at various peri- and intraocular locations. The aim of this review is to outline the pathogenetic role of ocular lymphatics, the respective molecular mechanisms and to discuss current and future therapeutic options based thereon. We will give an overview on the vascular anatomy of the healthy ocular surface and the molecular mechanisms contributing to corneal (lymph)angiogenic privilege. In addition, we present (i) current insights into the cellular and molecular mechanisms occurring during pathological neovascularization of the cornea triggered e.g. by inflammation or trauma, (ii) the role of lymphatic vessels in different ocular surface pathologies such as dry eye disease, corneal graft rejection, ocular graft versus host disease, allergy, and pterygium, (iii) the involvement of lymphatic vessels in ocular tumors and metastasis, and (iv) the novel role of the lymphatic-like structure of Schlemm's canal in glaucoma. Identification of the underlying molecular mechanisms and of novel modulators of lymphangiogenesis will contribute to the development of new therapeutic targets for the treatment of ocular diseases associated with pathological lymphangiogenesis in the future. The preclinical data presented here outline novel therapeutic concepts for promoting transplant survival, inhibiting metastasis of ocular tumors, reducing inflammation of the ocular surface, and treating glaucoma. Initial data from clinical trials suggest first success of novel treatment strategies to promote transplant survival based on pretransplant corneal lymphangioregression.
Collapse
Affiliation(s)
- Thomas Clahsen
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany
| | - Karina Hadrian
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany
| | - Maria Notara
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany
| | - Simona L Schlereth
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany
| | - Antonia Howaldt
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Verena Prokosch
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Thomas Volatier
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Deniz Hos
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany
| | - Falk Schroedl
- Center for Anatomy and Cell Biology, Institute of Anatomy and Cell Biology - Salzburg, Paracelsus Medical University, Salzburg, Austria
| | - Alexandra Kaser-Eichberger
- Center for Anatomy and Cell Biology, Institute of Anatomy and Cell Biology - Salzburg, Paracelsus Medical University, Salzburg, Austria
| | - Ludwig M Heindl
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Philipp Steven
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Cluster of Excellence: Cellular Stress Responses in Ageing-Associated Diseases, CECAD, University of Cologne, Cologne, Germany
| | - Jacobus J Bosch
- Centre for Human Drug Research and Leiden University Medical Center, Leiden, the Netherlands
| | | | - Alexander C Rokohl
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Hanhan Liu
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Mert Mestanoglu
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Hamid Kashkar
- Institute for Molecular Immunology, Center for Molecular Medicine Cologne (CMMC), CECAD Research Center, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Björn Schumacher
- Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany; Cluster of Excellence: Cellular Stress Responses in Ageing-Associated Diseases, CECAD, University of Cologne, Cologne, Germany
| | - Friedemann Kiefer
- European Institute for Molecular Imaging (EIMI), University of Münster, 48149, Münster, Germany
| | - Stefan Schulte-Merker
- Institute for Cardiovascular Organogenesis and Regeneration, Faculty of Medicine, WWU Münster, Münster, Germany
| | - Mario Matthaei
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Yanhong Hou
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, 83 Fenyang Road, Xuhui District, Shanghai, China
| | - Sonja Fassbender
- IUF‒Leibniz Research Institute for Environmental Medicine, Duesseldorf, Germany; Immunology and Environment, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Jonathan Jantsch
- Institute for Medical Microbiology, Immunology and Hygiene, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Wei Zhang
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Philip Enders
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Björn Bachmann
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Felix Bock
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany
| | - Claus Cursiefen
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany; Cluster of Excellence: Cellular Stress Responses in Ageing-Associated Diseases, CECAD, University of Cologne, Cologne, Germany.
| |
Collapse
|
10
|
Chia SL, Kapoor S, Carvalho C, Bajénoff M, Gentek R. Mast cell ontogeny: From fetal development to life-long health and disease. Immunol Rev 2023; 315:31-53. [PMID: 36752151 PMCID: PMC10952628 DOI: 10.1111/imr.13191] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Mast cells (MCs) are evolutionarily ancient innate immune cells with important roles in protective immunity against bacteria, parasites, and venomous animals. They can be found in most organs of the body, where they also contribute to normal tissue functioning, for example by engaging in crosstalk with nerves. Despite this, they are most widely known for their detrimental roles in allergy, anaphylaxis, and atopic disease. Just like macrophages, mast cells were conventionally thought to originate from the bone marrow. However, they are already present in fetal tissues before the onset of bone marrow hematopoiesis, questioning this dogma. In recent years, our view of myeloid cell ontogeny has been revised. We now know that the first mast cells originate from progenitors made in the extra-embryonic yolk sac, and later get supplemented with mast cells produced from subsequent waves of hematopoiesis. In most connective tissues, sizeable populations of fetal-derived mast cells persist into adulthood, where they self-maintain largely independently from the bone marrow. These developmental origins are highly reminiscent of macrophages, which are known to have critical functions in development. Mast cells too may thus support healthy development. Their fetal origins and longevity also make mast cells susceptible to genetic and environmental perturbations, which may render them pathological. Here, we review our current understanding of mast cell biology from a developmental perspective. We first summarize how mast cell populations are established from distinct hematopoietic progenitor waves, and how they are subsequently maintained throughout life. We then discuss what functions mast cells may normally have at early life stages, and how they may be co-opted to cause, worsen, or increase susceptibility to disease.
Collapse
Affiliation(s)
- Shin Li Chia
- Institute for Regeneration and Repair, Centre for Inflammation Research & Centre for Reproductive HealthThe University of EdinburghEdinburghUK
| | - Simran Kapoor
- Institute for Regeneration and Repair, Centre for Inflammation Research & Centre for Reproductive HealthThe University of EdinburghEdinburghUK
| | - Cyril Carvalho
- Institute for Regeneration and Repair, Centre for Inflammation Research & Centre for Reproductive HealthThe University of EdinburghEdinburghUK
| | - Marc Bajénoff
- Centre d'Immunologie de Marseille‐Luminy (CIML)MarseilleFrance
| | - Rebecca Gentek
- Institute for Regeneration and Repair, Centre for Inflammation Research & Centre for Reproductive HealthThe University of EdinburghEdinburghUK
| |
Collapse
|
11
|
Cho WJ, Mittal SK, Chauhan SK. Mesenchymal Stromal Cells Suppress T-Cell-Mediated Delayed-Type Hypersensitivity via ALCAM-CD6 Interaction. Stem Cells Transl Med 2023; 12:221-233. [PMID: 36972356 PMCID: PMC10108723 DOI: 10.1093/stcltm/szad012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 02/06/2023] [Indexed: 03/29/2023] Open
Abstract
Mounting evidence suggests mesenchymal stromal cells (MSCs) suppress CD4+ T-cell activation, but whether MSCs directly regulate activation and expansion of allogeneic T cells has not been fully deciphered. Here, we identified that both human and murine MSCs constitutively express ALCAM, a cognate ligand for CD6 receptors on T cells, and investigated its immunomodulatory function using in vivo and in vitro experiments. Our controlled coculture assays demonstrated that ALCAM-CD6 pathway is critical for MSCs to exert its suppressive function on early CD4+CD25- T-cell activation. Moreover, neutralizing ALCAM or CD6 results in the abrogation of MSC-mediated suppression of T-cell expansion. Using a murine model of delayed-type hypersensitivity response to alloantigen, we show that ALCAM-silenced MSCs lose the capacity to suppress the generation of alloreactive IFNγ-secreting T cells. Consequently, MSCs, following ALCAM knockdown, failed to prevent allosensitization and alloreactive T-cell-mediated tissue damage.
Collapse
Affiliation(s)
- WonKyung J Cho
- Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Sharad K Mittal
- Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Sunil K Chauhan
- Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
12
|
Msallam R, Redegeld FA. Mast cells-fetal mast cells crosstalk with maternal interfaces during pregnancy: Friend or foe? Pediatr Allergy Immunol 2023; 34:e13943. [PMID: 37102389 DOI: 10.1111/pai.13943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 03/04/2023] [Accepted: 03/08/2023] [Indexed: 04/28/2023]
Abstract
Mast cells (MC) are hematopoietic immune cells that play a major role during allergic reactions in adults by releasing a myriad of vasoactive and inflammatory mediators. MC seed all vascularized tissues and are most prominent in organs with a barrier function such as skin, lungs, and intestines. These secreted molecules cause mild symptoms such as localized itchiness and sneezing to life-threatening symptoms (i.e., anaphylactic shock). Presently, despite the extensive research on Th2-mediated immune responses in allergic diseases in adults, we are still unable to determine the mechanisms of the role of MC in developing pediatric allergic (PA) disorders. In this review, we will summarize the most recent findings on the origin of MC and discuss the underappreciated contribution of MC in the sensitization phase to maternal antibodies during pregnancy in allergic reactions and other diseases such as infectious diseases. Then, we will lay out potential MC-dependent therapeutic strategies to be considered in future investigations to understand the remaining gaps in MC research for a better quality of life for these young patients.
Collapse
Affiliation(s)
- Rasha Msallam
- Next Gen of Immunology (NGIg) Consultancy, Dubai, UAE
| | - Frank A Redegeld
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
13
|
Elbasiony E, Cho WJ, Singh A, Mittal SK, Zoukhri D, Chauhan SK. Increased activity of lacrimal gland mast cells are associated with corneal epitheliopathy in aged mice. NPJ AGING 2023; 9:2. [PMID: 36849524 PMCID: PMC9971332 DOI: 10.1038/s41514-023-00099-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 02/13/2023] [Indexed: 03/01/2023]
Abstract
The lacrimal gland undergoes significant structural and functional deterioration with aging. Marked with increased inflammation and fibrosis, the aged lacrimal gland is unable to perform its protective function. As a result, the ocular surface becomes highly susceptible to various ocular surface pathologies, including corneal epitheliopathy. We and others have previously shown that mast cells mediate tissue inflammation by recruiting other immune cells. However, despite their well-known characteristics of secreting various inflammatory mediators, whether mast cells contribute to the immune cell aggregation and activation, and acinar dystrophy of the aged lacrimal gland has not been investigated. Here, we demonstrate the role of mast cells in age-related lacrimal gland pathophysiology using mast cell-deficient (cKitw-sh) mice. Our data demonstrated a significant increase in mast cell frequencies and immune cell infiltration in the lacrimal gland of aged mice. Interestingly, mast cell deficiency resulted in a substantial reduction in inflammation and preservation of lacrimal gland structure, suggesting that mast cells mediate the aging process of the lacrimal gland.
Collapse
Affiliation(s)
- Elsayed Elbasiony
- Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - WonKyung J Cho
- Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Aastha Singh
- Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Sharad K Mittal
- Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Driss Zoukhri
- Department of Ophthalmology, Tufts University School of Medicine, Boston, MA, USA
- Department of Comprehensive Care, Tufts University School of Dental Medicine, Boston, MA, USA
| | - Sunil K Chauhan
- Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
14
|
Bonini S, Leonardi A. The multifaceted aspects of ocular allergies: Phenotypes and endotypes. Ocul Surf 2022; 26:174-183. [PMID: 36067980 DOI: 10.1016/j.jtos.2022.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 08/26/2022] [Accepted: 08/29/2022] [Indexed: 12/01/2022]
Abstract
Like the lung, skin, and nose, the external eye is a common target of allergic inflammation. Ocular allergy (OA) represents a collection of underestimated diseases of the eye observed in children and adults. The ocular manifestations are the expression of multifactorial immune mechanisms that generally have a good prognosis, but for a few patients, long term inflammation may remarkably reduce the visual function. Evidence suggests that other co-participant systems, including epigenetic, genetic, environmental, individual factors, sex hormones, and the central and autonomic nervous systems may influence the ocular response from distant sites. This is consistent with the concept that the eye is an organ fully integrated with the rest of the body and that the therapeutic approach should be holistic, dynamic, and personalized. For instance, androgens and estrogens binding to receptors on the ocular surface and the continuous cross-talking of neuromediators and growth factors with immune cells act to maintain the ocular surface homeostasis in response to environmental challenges. The immune system links and regulates the response of the ocular surface. Complex and incompletely understood mechanisms influence the innate and adaptive immune responses and generate different OA phenotypes and endotypes discussed in the present review.
Collapse
Affiliation(s)
- Stefano Bonini
- Ophthalmology Operative Complex Unit, Campus Bio-Medico University Hospital, 00128, Rome, Italy
| | - Andrea Leonardi
- Department of Neuroscience, Ophthalmology Unit, University of Padova, Padova, Italy.
| |
Collapse
|
15
|
Shukla S, Cho W, Elbasiony E, Singh RB, Mittal SK, Chauhan SK. Non-immune and immune functions of interleukin-36γ suppress epithelial repair at the ocular surface. FASEB J 2022; 36:10.1096/fj.202200174RR. [PMID: 35781326 PMCID: PMC9924024 DOI: 10.1096/fj.202200174rr] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 05/16/2022] [Accepted: 06/03/2022] [Indexed: 02/05/2023]
Abstract
Regulation of innate inflammation is critical for maintaining tissue homeostasis and barrier function, especially in those interfacing the external environments such as the skin and cornea. Expression of pro-inflammatory cytokines by injured tissues has been shown to exacerbate the inflammatory cascade, causing tissue damage. Interleukin 36, a subfamily of the IL-1 superfamily, consists of three pro-inflammatory agonists-IL36α, IL36β, and IL36γ and an IL36 receptor antagonist (IL36Ra). The current investigation, for the first time, reports that IL36γ is the primary agonist expressed by the corneal epithelium, which is significantly upregulated following corneal injury. The function of IL36γ on non-immune cells, in addition to innate inflammatory cells, in regulating tissue homeostasis has not been well investigated. Using a loss-of-function approach via neutralizing antibody treatment, our data demonstrate that blocking endogenously expressed IL36γ in epithelial cells promotes rapid re-epithelialization in in vitro wound closure assay. Finally, by utilizing a naturally occurring antagonist IL36Ra in a well-established murine model of ocular injury, our study demonstrates that inhibition of IL36γ accelerates epithelial regeneration and suppresses tissue inflammation. Given rapid wound healing is critical for re-establishing normal tissue structure and function, our investigation on the function of IL36γ provides evidence for the development of novel IL36γ-targeting strategies to promote tissue repair.
Collapse
Affiliation(s)
- Sachin Shukla
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts
- L.V. Prasad Eye Institute, Hyderabad, India
| | - WonKyung Cho
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts
| | - Elsayed Elbasiony
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts
| | - Rohan Bir Singh
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts
| | - Sharad K. Mittal
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts
| | - Sunil K. Chauhan
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
16
|
Corneal stromal repair and regeneration. Prog Retin Eye Res 2022; 91:101090. [DOI: 10.1016/j.preteyeres.2022.101090] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 05/17/2022] [Accepted: 05/19/2022] [Indexed: 01/02/2023]
|
17
|
Li F, Yu R, Sun X, Chen X, Xu P, Huang Y, Huang S, Xue Y, Fu T, Liu J, Li Z. Autonomic nervous system receptor-mediated regulation of mast cell degranulation modulates the inflammation after corneal epithelial abrasion. Exp Eye Res 2022; 219:109065. [PMID: 35421396 DOI: 10.1016/j.exer.2022.109065] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 03/17/2022] [Accepted: 03/30/2022] [Indexed: 01/24/2023]
Abstract
Mast cells (MCs) regulate wound healing and are influenced by the autonomic nervous system (ANS). However, the underlying mechanisms affecting wound healing outcomes remain elusive. Here, we explored the specific role of the ANS by regulating MC degranulation following corneal epithelium abrasion. A mouse model of corneal abrasion was established by mechanically removing a 2-mm central epithelium. Wound closure, neutrophil infiltration, and transcription of injured corneas were investigated using whole-mount immunostaining, flow cytometry, and RNA-sequencing analysis, respectively. Inhibition of MC degranulation by the MC stabilizers cromolyn sodium and lodoxamide tromethamine increased the infiltration of neutrophils and delayed healing of abraded corneas. Moreover, transcriptomic profiling analysis showed that purified MCs from the limbus expressed adrenergic and cholinergic receptors. Pharmacological manipulation and sympathectomy with 6-hydroxydopamine confirmed that sympathetic nervous system signaling inhibited MC degranulation after corneal abrasion, whereas parasympathetic nervous system signaling enhanced MC degranulation. We conclude that normal degranulation of MCs in the corneal limbus and crosstalk between the ANS and MCs are crucial for the appropriate control of inflammation and the repair progress of wounded corneas. This suggests a potential approach for improving defective corneal wound healing by the administration of clinically available autonomic activity-modulating agents.
Collapse
Affiliation(s)
- Fanying Li
- International Ocular Surface Research Center, Institute of Ophthalmology, Key Laboratory for Regenerative Medicine, Jinan University, Guangzhou, China; Department of Microbiology and Immunology, School of Medicine, Jinan University, Guangzhou, China
| | - Ruoxun Yu
- International Ocular Surface Research Center, Institute of Ophthalmology, Key Laboratory for Regenerative Medicine, Jinan University, Guangzhou, China; Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xin Sun
- International Ocular Surface Research Center, Institute of Ophthalmology, Key Laboratory for Regenerative Medicine, Jinan University, Guangzhou, China; Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xinwei Chen
- International Ocular Surface Research Center, Institute of Ophthalmology, Key Laboratory for Regenerative Medicine, Jinan University, Guangzhou, China; Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Pengyang Xu
- International Ocular Surface Research Center, Institute of Ophthalmology, Key Laboratory for Regenerative Medicine, Jinan University, Guangzhou, China
| | - Yijia Huang
- International Ocular Surface Research Center, Institute of Ophthalmology, Key Laboratory for Regenerative Medicine, Jinan University, Guangzhou, China; Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Shuoya Huang
- International Ocular Surface Research Center, Institute of Ophthalmology, Key Laboratory for Regenerative Medicine, Jinan University, Guangzhou, China; Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yunxia Xue
- International Ocular Surface Research Center, Institute of Ophthalmology, Key Laboratory for Regenerative Medicine, Jinan University, Guangzhou, China
| | - Ting Fu
- International Ocular Surface Research Center, Institute of Ophthalmology, Key Laboratory for Regenerative Medicine, Jinan University, Guangzhou, China
| | - Jun Liu
- International Ocular Surface Research Center, Institute of Ophthalmology, Key Laboratory for Regenerative Medicine, Jinan University, Guangzhou, China; Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, China.
| | - Zhijie Li
- International Ocular Surface Research Center, Institute of Ophthalmology, Key Laboratory for Regenerative Medicine, Jinan University, Guangzhou, China; Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, China.
| |
Collapse
|
18
|
Chen Y, Wang S, Alemi H, Dohlman T, Dana R. Immune regulation of the ocular surface. Exp Eye Res 2022; 218:109007. [PMID: 35257715 PMCID: PMC9050918 DOI: 10.1016/j.exer.2022.109007] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 02/10/2022] [Accepted: 02/20/2022] [Indexed: 01/01/2023]
Abstract
Despite constant exposure to various environmental stimuli, the ocular surface remains intact and uninflamed while maintaining the transparency of the cornea and its visual function. This 'immune privilege' of the ocular surface is not simply a result of the physical barrier function of the mucosal lining but, more importantly, is actively maintained through a variety of immunoregulatory mechanisms that prevent the disruption of immune homeostasis. In this review, we focus on essential molecular and cellular players that promote immune quiescence in steady-state conditions and suppress inflammation in disease-states. Specifically, we examine the interactions between the ocular surface and its local draining lymphoid compartment, by encompassing the corneal epithelium, corneal nerves and cornea-resident myeloid cells, conjunctival goblet cells, and regulatory T cells (Treg) in the context of ocular surface autoimmune inflammation (dry eye disease) and alloimmunity (corneal transplantation). A better understanding of the immunoregulatory mechanisms will facilitate the development of novel, targeted immunomodulatory strategies for a broad range of ocular surface inflammatory disorders.
Collapse
Affiliation(s)
- Yihe Chen
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA.
| | - Shudan Wang
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA
| | - Hamid Alemi
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA
| | - Thomas Dohlman
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA
| | - Reza Dana
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA
| |
Collapse
|
19
|
Kempuraj D, Mohan RR. Autophagy in Extracellular Matrix and Wound Healing Modulation in the Cornea. Biomedicines 2022; 10:biomedicines10020339. [PMID: 35203548 PMCID: PMC8961790 DOI: 10.3390/biomedicines10020339] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 12/24/2021] [Accepted: 12/28/2021] [Indexed: 12/18/2022] Open
Abstract
Autophagy is a robust cellular mechanism for disposing of harmful molecules or recycling them to cells, which also regulates physiopathological processes in cornea. Dysregulated autophagy causes inefficient clearance of unwanted proteins and cellular debris, mitochondrial disorganization, defective inflammation, organ dysfunctions, cell death, and diseases. The cornea accounts for two-thirds of the refraction of light that occurs in the eyes, but is prone to trauma/injury and infection. The extracellular matrix (ECM) is a noncellular dynamic macromolecular network in corneal tissues comprised of collagens, proteoglycans, elastin, fibronectin, laminins, hyaluronan, and glycoproteins. The ECM undergoes remodeling by matrix-degrading enzymes and maintains corneal transparency. Autophagy plays an important role in the ECM and wound healing maintenance. Delayed/dysregulated autophagy impacts the ECM and wound healing, and can lead to corneal dysfunction. Stromal wound healing involves responses from the corneal epithelium, basement membrane, keratocytes, the ECM, and many cytokines and chemokines, including transforming growth factor beta-1 and platelet-derived growth factor. Mild corneal injuries self-repair, but greater injuries lead to corneal haze/scars/fibrosis and vision loss due to disruptions in the ECM, autophagy, and normal wound healing processes. Presently, the precise role of autophagy and ECM remodeling in corneal wound healing is elusive. This review discusses recent trends in autophagy and ECM modulation in the context of corneal wound healing and homeostasis.
Collapse
Affiliation(s)
- Duraisamy Kempuraj
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO 65212, USA;
- One-Health Vision Research Program, Departments of Veterinary Medicine & Surgery and Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Rajiv R. Mohan
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO 65212, USA;
- One-Health Vision Research Program, Departments of Veterinary Medicine & Surgery and Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO 65212, USA
- Mason Eye Institute, School of Medicine, University of Missouri, Columbia, MO 65212, USA
- Correspondence:
| |
Collapse
|
20
|
Suppression of lipopolysaccharide-induced corneal opacity by hepatocyte growth factor. Sci Rep 2022; 12:494. [PMID: 35017561 PMCID: PMC8752742 DOI: 10.1038/s41598-021-04418-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 12/22/2021] [Indexed: 12/21/2022] Open
Abstract
Keratitis induced by bacterial toxins, including lipopolysaccharide (LPS), is a major cause of corneal opacity and vision loss. Our previous study demonstrates hepatocyte growth factor (HGF) promotes epithelial wound healing following mechanical corneal injury. Here, we investigated whether HGF has the capacity to suppress infectious inflammatory corneal opacity using a new model of LPS-induced keratitis. Keratitis, induced by two intrastromal injections of LPS on day 1 and 4 in C57BL/6 mice, resulted in significant corneal opacity for up to day 10. Following keratitis induction, corneas were topically treated with 0.1% HGF or PBS thrice daily for 5 days. HGF-treated mice showed a significantly smaller area of corneal opacity compared to PBS-treated mice, thus improving corneal transparency. Moreover, HGF treatment resulted in suppression of α-SMA expression, compared to PBS treatment. HGF-treated corneas showed normalized corneal structure and reduced expression of pro-inflammatory cytokine, demonstrating that HGF restores corneal architecture and immune quiescence in corneas with LPS-induced keratitis. These findings offer novel insight into the potential application of HGF-based therapies for the prevention and treatment of infection-induced corneal opacity.
Collapse
|
21
|
Cho W, Mittal SK, Elbasiony E, Chauhan SK. Ocular surface mast cells promote inflammatory lymphangiogenesis. Microvasc Res 2022; 141:104320. [PMID: 35031275 PMCID: PMC8923954 DOI: 10.1016/j.mvr.2022.104320] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 11/16/2021] [Accepted: 01/06/2022] [Indexed: 12/01/2022]
Abstract
Mast cells, sentinel immune cells, are most abundantly expressed in vascularized tissues that interface the external environment, such as the skin and ocular surface. Our previous reports have shown mast cells reside closely with vascular endothelial cells and mediate the pathogenic angiogenic response. However, the contribution of mast cells and their underlying mechanisms on lymphangiogenesis have not been fully deciphered. Using a murine model of inflammatory corneal angiogenesis, we observed adjacent migration of activated mast cells with new lymph vessel growth. Our in vitro co-culture assays demonstrate that mast cells express high levels of of VEGF-D and directly promote lymphatic endothelial cell tube formation and proliferation. Moreover, our loss-of-function approaches, using mast cell knockout mice and cromolyn-mediated mast cell inhibition, showed mast cell deficiency suppresses the induction of inflammatory lymphangiogenesis and VEGF-D expression at the ocular surface following corneal tissue insult. Our findings suggest blockade of mast cells as a potential therapeutic strategy to inhibit pathological lymphangiogenesis.
Collapse
Affiliation(s)
- WonKyung Cho
- Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Sharad K Mittal
- Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Elsayed Elbasiony
- Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Sunil K Chauhan
- Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
22
|
Huang C, Qi X, Chen H, Chao W, Qi X, Wang H, Gao H. Monolith/Hydrogel composites as triamcinolone acetonide carriers for curing corneal neovascularization in mice by inhibiting the fibrinolytic system. Drug Deliv 2021; 29:18-30. [PMID: 34962228 PMCID: PMC8725936 DOI: 10.1080/10717544.2021.2014603] [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: 11/17/2022] Open
Abstract
Corneal neovascularization is a serious corneal pathological change caused by various factors. The drug delivery system is of great significance for the effective treatment of corneal neovascularization. Herein, we developed and characterized a monolith/hydrogel composite as the triamcinolone acetonide (TA) carrier for curing corneal neovascularization. The composite was prepared by photo-initiated free radical polymerization of multi-methacrylate substituted dodecamine organic molecular cage and post-modified by the sequential photo-initiated free radical polymerization of acrylated gelatin. The globular morphology and structural property of as-prepared composites were evaluated by scanning electron microscopy, Fourier-transform infrared spectroscopy and solid-state cross polarization magic angle spinning carbon-13 nuclear magnetic resonance. Then swelling ratio and the TA loading capacity were investigated then. Compared with gelatin hydrogel, the composites exhibited a decreased swelling ratio and an improved loading capacity. With good biocompatibility, the composite can sustainedly release TA for up to 28 days, and effectively inhibit corneal neovascularization with an alkali burn-induced corneal neovascularization model. Additionally, tandem mass tags-labeled quantitative proteomics were performed to identify differentially expressed proteins between vascularized and devascularized corneas. The Kyoto Encyclopedia of Genes and Genomes enrichment analysis revealed that the inhibition process could be primarily linked to the fibrinolytic system. These results demonstrated the potential of monolith/hydrogel composites as delivery systems in the therapy for biomedical diseases.
Collapse
Affiliation(s)
- Cixin Huang
- Medical College, Qingdao University, Qingdao, China.,State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China.,Eye Hospital of Shandong First Medical University, Jinan, China
| | - Xia Qi
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China.,School of Ophthalmology, Shandong First Medical University, Jinan, China
| | - Huilin Chen
- Medical College, Qingdao University, Qingdao, China.,State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China.,Eye Hospital of Shandong First Medical University, Jinan, China
| | - Wei Chao
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China.,School of Ophthalmology, Shandong First Medical University, Jinan, China
| | - Xiaolin Qi
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China.,Eye Hospital of Shandong First Medical University, Jinan, China.,School of Ophthalmology, Shandong First Medical University, Jinan, China
| | - Hongwei Wang
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China.,School of Ophthalmology, Shandong First Medical University, Jinan, China
| | - Hua Gao
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China.,Eye Hospital of Shandong First Medical University, Jinan, China.,School of Ophthalmology, Shandong First Medical University, Jinan, China
| |
Collapse
|
23
|
Fukase S, Ando T, Matsuzawa M, Kimura M, Sone Y, Izawa K, Kaitani A, Kamei A, Kojima M, Nakano N, Maeda K, Shimizu T, Ogawa H, Okumura K, Nishiyama M, Murakami A, Ebihara N, Kitaura J. Pollen shells and soluble factors play non-redundant roles in the development of allergic conjunctivitis in mice. Ocul Surf 2021; 22:152-162. [PMID: 34428578 DOI: 10.1016/j.jtos.2021.08.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 08/12/2021] [Accepted: 08/16/2021] [Indexed: 11/26/2022]
Abstract
PURPOSE We aimed to clarify the role of particulate allergen exposure to the conjunctiva in the development of allergic conjunctivitis. METHODS We administered ragweed pollen suspension, pollen extract, pollen shell, particulate air pollutants, and their combinations to the mouse conjunctiva five days a week without prior sensitization. Clinical signs were scored. Histological changes, cellular infiltrations, mRNA expressions, lymph node cell recall responses, and serum immunoglobulin levels were assessed. Immune cell-depleting antibodies and ST2 knockout mice were used to investigate the cellular and molecular requirements. RESULTS Pollen suspension, but not the extract or shell alone, induced robust eosinophilic conjunctivitis, accompanied by a proliferative response of epithelial cells. A combination of pollen extract and shell completely restored eosinophil accumulation. In addition, eosinophilic conjunctivitis was induced by a mixture of particulate air pollutants and pollen extract. Mechanistically, eosinophil accumulation was ameliorated by deficiency of the IL-33 receptor ST2 and abolished by depleting CD4+ T cells. Pollen shells, but not the extract, induced IL-33 release from conjunctival epithelial cells in vivo. CONCLUSIONS Our results indicate the non-redundant roles for the allergens' particulate properties and soluble factors in the development of allergic conjunctivitis.
Collapse
Affiliation(s)
- Saaya Fukase
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan; Department of Ophthalmology, Juntendo University Urayasu Hospital, Urayasu, Chiba, 279-0021, Japan; Department of Ophthalmology, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
| | - Tomoaki Ando
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan.
| | - Moe Matsuzawa
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan; Department of Ophthalmology, Juntendo University Urayasu Hospital, Urayasu, Chiba, 279-0021, Japan; Department of Ophthalmology, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
| | - Meiko Kimura
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan; Department of Ophthalmology, Juntendo University Urayasu Hospital, Urayasu, Chiba, 279-0021, Japan; Department of Ophthalmology, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
| | - Yusuke Sone
- Laboratory of Cell Biotechnology, Biotechnology Research Center, The University of Tokyo, Tokyo, 113-8657, Japan
| | - Kumi Izawa
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
| | - Ayako Kaitani
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
| | - Anna Kamei
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan; Department of Science of Allergy and Inflammation, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
| | - Mayuki Kojima
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan; Department of Pediatrics and Adolescent Medicine, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
| | - Nobuhiro Nakano
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
| | - Keiko Maeda
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan; Department of Immunological Diagnosis, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
| | - Toshiaki Shimizu
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan; Department of Pediatrics and Adolescent Medicine, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
| | - Hideoki Ogawa
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
| | - Ko Okumura
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
| | - Makoto Nishiyama
- Laboratory of Cell Biotechnology, Biotechnology Research Center, The University of Tokyo, Tokyo, 113-8657, Japan
| | - Akira Murakami
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
| | - Nobuyuki Ebihara
- Department of Ophthalmology, Juntendo University Urayasu Hospital, Urayasu, Chiba, 279-0021, Japan; Department of Ophthalmology, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
| | - Jiro Kitaura
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan; Department of Science of Allergy and Inflammation, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan.
| |
Collapse
|
24
|
Lee SJ, Im ST, Wu J, Cho CS, Jo DH, Chen Y, Dana R, Kim JH, Lee SM. Corneal lymphangiogenesis in dry eye disease is regulated by substance P/neurokinin-1 receptor system through controlling expression of vascular endothelial growth factor receptor 3. Ocul Surf 2021; 22:72-79. [PMID: 34311077 DOI: 10.1016/j.jtos.2021.07.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 07/20/2021] [Accepted: 07/21/2021] [Indexed: 12/17/2022]
Abstract
PURPOSE To evaluate the role of substance P (SP)/neurokinin-1 receptor (NK1R) system in the regulation of pathologic corneal lymphangiogenesis in dry eye disease (DED). METHODS Immunocytochemistry, angiogenesis assay, and Western blot analysis of human dermal lymphatic endothelial cells (HDLECs) were conducted to assess the involvement of SP/NK1R system in lymphangiogenesis. DED was induced in wild-type C57BL/6 J mice using controlled-environment chamber without scopolamine. Immunohistochemistry, corneal fluorescein staining, and phenol red thread test were used to evaluate the effect of SP signaling blockade in the corneal lymphangiogenesis. The expression of lymphangiogenic factors in the corneal and conjunctival tissues of DED mouse model was quantified by real-time polymerase chain reaction. RESULTS NK1R expression and pro-lymphangiogenic property of SP/NK1R system in HDLECs were confirmed by Western blot analysis and angiogenesis assay. Blockade of SP signaling with L733,060, an antagonist of NK1R, or NK1R-targeted siRNA significantly inhibited lymphangiogenesis and expression of vascular endothelial growth factor (VEGF) receptor 3 stimulated by SP in HDLECs. NK1R antagonist also suppressed pathological corneal lymphangiogenesis and ameliorated the clinical signs of dry eye in vivo. Furthermore, NK1R antagonist effectively suppressed the lymphangiogenic factors, including VEGF-C, VEGF-D, and VEGF receptor 3 in the corneal and conjunctival tissues of DED. CONCLUSIONS SP/NK1R system promotes lymphangiogenesis in vitro and NK1R antagonism suppresses pathologic corneal lymphangiogenesis in DED in vivo.
Collapse
Affiliation(s)
- Seok Jae Lee
- Fight Against Angiogenesis-Related Blindness (FARB) Laboratory, Clinical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Sang-Taek Im
- Fight Against Angiogenesis-Related Blindness (FARB) Laboratory, Clinical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Jun Wu
- Fight Against Angiogenesis-Related Blindness (FARB) Laboratory, Clinical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Chang Sik Cho
- Fight Against Angiogenesis-Related Blindness (FARB) Laboratory, Clinical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Dong Hyun Jo
- Department of Anatomy & Cell Biology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yihe Chen
- Schepens Eye Research Institute of Massachusetts Eye and Ear, MA, USA; Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Reza Dana
- Schepens Eye Research Institute of Massachusetts Eye and Ear, MA, USA; Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Jeong Hun Kim
- Fight Against Angiogenesis-Related Blindness (FARB) Laboratory, Clinical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea; Department of Ophthalmology, College of Medicine, Seoul National University, Seoul, Republic of Korea; Advanced Biomedical Research Center, Korea Research Institute of Bioscience & Biotechnology, Daejeon, Republic of Korea.
| | - Sang-Mok Lee
- Department of Cornea, External Disease & Refractive Surgery, HanGil Eye Hospital, Incheon, Republic of Korea; Department of Ophthalmology, Catholic Kwandong University College of Medicine, Gangneung-si, Republic of Korea.
| |
Collapse
|
25
|
Spatial Distribution of Mast Cells Regulates Asymmetrical Angiogenesis at the Ocular Surface. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:1108-1117. [PMID: 33705754 DOI: 10.1016/j.ajpath.2021.02.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 02/10/2021] [Accepted: 02/17/2021] [Indexed: 11/23/2022]
Abstract
Mast cells, historically known for their function as effector cells in the induction of allergic diseases, reside in all vascularized tissues of the body, particularly, in proximity to blood and lymphatic vessels. Despite being neighboring sentinel cells to blood vessels, whether the spatial distribution of mast cells regulates the degree of angiogenesis remains to be investigated. Herein, an asymmetrical distribution of mast cells was shown at the murine ocular surface, with the higher number of mast cells distributed along the nasal limbus of the cornea compared with the temporal side. Using a well-characterized murine model of suture-induced corneal neovascularization, insult to the nasal side was shown to result in more extensive angiogenesis compared with that to the temporal side. To directly assess the impact of the spatial distribution of mast cell on angiogenesis, neovascularization was induced in mast cell-deficient mice (cKitw-sh). Unlike the wild-type (C57BL/6) mice, cKitw-sh mice did not show disproportionate growth of corneal blood vessels following the temporal and nasal insult. Moreover, cromolyn-mediated pharmacologic blockade of mast cells at the ocular surface attenuated the asymmetrical nasal and temporal neovascularization, suggesting that spatial distribution of mast cells significantly contributes to angiogenic response at the ocular surface.
Collapse
|