Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.198
Filter
1.
Front Immunol ; 15: 1416181, 2024.
Article in English | MEDLINE | ID: mdl-39104531

ABSTRACT

Several blinding diseases affecting the retina and optic nerve are exacerbated by or caused by dysregulated inflammation and oxidative stress. These diseases include uveitis, age related macular degeneration, diabetic retinopathy and glaucoma. Consequently, despite their divergent symptoms, treatments that reduce oxidative stress and suppress inflammation may be therapeutic. The production of inflammatory cytokines and their activities are regulated by a class of proteins termed Suppressors of Cytokine Signaling (SOCS). SOCS1 and SOCS3 are known to dampen signaling via pathways employing Janus kinases and signal transducer and activator of transcription proteins (JAK/STAT), Toll-like Receptors (TLR), nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), mitogen activated kinase (MAPK) and NLR family pyrin domain containing 3 (NLRP3). We have developed cell-penetrating peptides from the kinase inhibitory region of the SOCS1 and SOCS3 (denoted as R9-SOCS1-KIR and R9-SOCS3-KIR) and tested them in retinal pigment epithelium (RPE) cells and in macrophage cell lines. SOCS-KIR peptides exhibited anti-inflammatory, anti-oxidant and anti-angiogenic properties. In cell culture, both Th1 and Th17 cells were suppressed together with the inhibition of other inflammatory markers. We also observed a decrease in oxidants and a simultaneous rise in neuroprotective and anti-oxidant effectors. In addition, treatment prevented the loss of gap junction proteins and the ensuing drop in transepithelial electrical resistance in RPE cells. When tested in mouse models by eye drop instillation, they showed protection against autoimmune uveitis, as a prophylactic as well as a therapeutic. Mice with endotoxin-induced uveitis were protected by eye drop administration as well. R9-SOCS3-KIR was particularly effective against the pathways acting through STAT3, e.g. IL-6 and VEGF-A mediated responses that lead to macular degeneration. Eye drop administration of R9-SOCS3-KIR stimulated production of antioxidant effectors and reduced clinical symptoms in mouse model of oxidative stress that replicates the RPE injury occurring in AMD. Because these peptides suppress multiple pathogenic stimuli and because they can be delivered topically to the cornea, they are attractive candidates for therapeutics for uveitis, macular degeneration, diabetic retinopathy and glaucoma.


Subject(s)
Oxidative Stress , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling 3 Protein , Animals , Oxidative Stress/drug effects , Suppressor of Cytokine Signaling 3 Protein/metabolism , Mice , Suppressor of Cytokine Signaling 1 Protein/metabolism , Humans , Inflammation/immunology , Inflammation/drug therapy , Cornea/metabolism , Cornea/immunology , Retinal Pigment Epithelium/metabolism , Eye Diseases/drug therapy , Eye Diseases/immunology , Eye Diseases/metabolism , Peptides/pharmacology , Peptides/therapeutic use , Disease Models, Animal , Mice, Inbred C57BL
2.
Front Cell Infect Microbiol ; 14: 1416105, 2024.
Article in English | MEDLINE | ID: mdl-39119293

ABSTRACT

Introduction: Herpes simplex keratitis (HSK) is a blinding disease caused by corneal infection of Herpes simplex virus type 1 (HSV-1). Effective clearance of HSV-1 from the infected cornea is crucial for HSK management. Macrophages play an important part in the innate immune defense against viral infections. This study investigates the immunomodulatory role of NLRP12 in macrophage immune response during HSV-1 infection. Methods: NLRP12 expression post-infection was assessed in various macrophage cell lines. Overexpression of NLRP12 was achieved by lentiviral transfection, and its effect on HSV-1 replication and immune responses were examined. Mechanistic insights into the role of NLRP12 were explored using immunofluorescence and Western Blot. For in vivo studies, ocular adoptive transfer of NLRP12-overexpressing bone marrow derived macrophages (BMDMs) was performed. HSV-1 viral loads, HSK symptoms, and macrophage-mediated immune responses were investigated. Results: A significant decrease in NLRP12 expression post-infection was observed in various macrophage cell lines. Overexpression of NLRP12 in macrophages reduced HSV-1 replication. Mechanistically, overexpression of NLRP12 triggered early and robust pyroptosis in response to HSV-1 infection, inducing interleukin (IL)-18 production and activating downstream antiviral responses through the JAK-STAT signaling pathway. In vivo, ocular adoptive transfer of NLRP12-overexpressing BMDMs to mouse corneas alleviated HSK damage and reduced HSV-1 viral loads. NLRP12-overexpressing BMDMs improved antiviral responses in the cornea and promoted the maturation of corneal-infiltrating macrophages and dendritic cells. Additionally, NLRP12-overexpressing BMDMs amplified the adaptive immune response in the submandibular draining lymph nodes. Discussion: These findings highlight the role of NLRP12 in macrophage-mediated immune response against HSV-1 infection and suggest its potential for possible immunotherapy for HSK.


Subject(s)
Herpesvirus 1, Human , Keratitis, Herpetic , Macrophages , Virus Replication , Keratitis, Herpetic/immunology , Keratitis, Herpetic/virology , Keratitis, Herpetic/therapy , Animals , Macrophages/immunology , Mice , Herpesvirus 1, Human/immunology , Cornea/virology , Cornea/immunology , Immunity, Innate , Cell Line , Disease Models, Animal , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Pyroptosis , Mice, Inbred C57BL , Humans , Female , Viral Load
3.
Exp Eye Res ; 245: 109982, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38942134

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.


Subject(s)
Mast Cells , Humans , Mast Cells/physiology , Cornea/immunology , Immunity, Innate/physiology , Corneal Diseases/pathology , Animals , Wound Healing/physiology , Conjunctiva/immunology , Conjunctiva/pathology
4.
Exp Eye Res ; 245: 109971, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38871165

ABSTRACT

PURPOSE: This study aims to reveal the immunopathogenesis of the high-risk corneal transplantation using a comparative proteomic approach. METHODS: The immunological properties of ocular tissues (including corneal grafts, aqueous humour, and iris-ciliary body) were analysed using a high-risk rabbit corneal transplantation model employing a comparative proteomic approach. RESULTS: The corneal grafts revealed a dramatic increase in the immune response both at the early (postoperative day 7) and rejection stages, along with the appearance of transplantation stress-induced cellular senescence in the early stage. The aqueous humour (AH) displayed persistent pathological alterations, indicated by the significant enrichment of complement and coagulation cascades pathway in the early stage and interleukin (IL)-17 signalling pathway in the rejection stage. More surprisingly, the pronounced elevation of immune response was also observed in the iris-ciliary body (I-CB) tissues at the early and rejection stages. The enriched immune-related pathways were associated with antigen processing and presentation, complement and coagulation cascades, and IL-17 signalling pathway. Furthermore, proteomic analysis revealed that the implantation of Cyclosporine A drug delivery system (CsA-DDS) into the anterior chamber obviously mitigated corneal transplantation rejection by inhibiting immunoreaction both in the corneal grafts and I-CB tissues. CONCLUSION: The results highlighted the involvement of intraocular immunity both in the grafts and I-CB tissues during corneal transplantation rejection, further suggesting the anterior chamber as an optimal drug-delivery site for its treatment.


Subject(s)
Aqueous Humor , Corneal Transplantation , Graft Rejection , Proteomics , Animals , Graft Rejection/immunology , Graft Rejection/metabolism , Rabbits , Aqueous Humor/metabolism , Disease Models, Animal , Iris/immunology , Ciliary Body/immunology , Ciliary Body/metabolism , Male , Immunosuppressive Agents/therapeutic use , Cornea/immunology , Cornea/metabolism , Cornea/pathology , Eye Proteins/metabolism
5.
Int Immunopharmacol ; 137: 112428, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-38908077

ABSTRACT

Herpes simplex keratitis (HSK) is a blinding disease caused by herpes simplex virus type 1 (HSV-1) infection, and rapid eradication of the virus from the affected cornea is imperative. Nod-like receptors (NLRs) are intracellular innate immune sensors closely associated with cell death, inflammation and immune responses. In this study, we investigated the role of NLRP12 in the antiviral immunology in HSK and the underlying mechanisms. We found that NLRP12 expression was significantly decreased in HSV-1-infected human corneal epithelial cells (HCE-Ts) and HSK mouse corneas. Overexpression of NLRP12 significantly reduced viral replication in infected HCE-Ts and functioned through inflammasome-mediated pyroptosis and downstream IL-18-IFN-γ axis. In HSK mouse models, overexpression of NLRP12 reduced viral replication in the cornea and alleviated HSK symptoms. This resulted from enhanced antiviral immune responses including the activation of specific immune cells in both the cornea and the draining lymph nodes. Specifically, the NLRP12-IL-18-IFN-γ axis regulated the interaction between infected corneal epithelial cells and macrophages. In conclusion, our study identified a role of NLRP12 in mediating pyroptosis and regulating antiviral immune responses. This novel finding opens the possibilities of NLRP12 as a viable target in the therapeutic strategies for HSV-1 infection.


Subject(s)
Herpesvirus 1, Human , Interferon-gamma , Interleukin-18 , Keratitis, Herpetic , Mice, Inbred C57BL , Pyroptosis , Signal Transduction , Animals , Keratitis, Herpetic/immunology , Keratitis, Herpetic/virology , Humans , Interleukin-18/metabolism , Interleukin-18/immunology , Interferon-gamma/metabolism , Interferon-gamma/immunology , Herpesvirus 1, Human/immunology , Herpesvirus 1, Human/physiology , Mice , Cornea/virology , Cornea/immunology , Cornea/pathology , Female , Virus Replication , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Disease Models, Animal , Inflammasomes/metabolism , Inflammasomes/immunology , Immunity, Innate
6.
Front Cell Infect Microbiol ; 14: 1346821, 2024.
Article in English | MEDLINE | ID: mdl-38694515

ABSTRACT

Background: Microbial keratitis is one of the leading causes of blindness globally. An overactive immune response during an infection can exacerbate damage, causing corneal opacities and vision loss. This study aimed to identify the differentially expressed genes between corneal infection patients and healthy volunteers within the cornea and conjunctiva and elucidate the contributing pathways to these conditions' pathogenesis. Moreover, it compared the corneal and conjunctival transcriptomes in corneal-infected patients to cytokine levels in tears. Methods: Corneal and conjunctival swabs were collected from seven corneal infection patients and three healthy controls under topical anesthesia. RNA from seven corneal infection patients and three healthy volunteers were analyzed by RNA sequencing (RNA-Seq). Tear proteins were extracted from Schirmer strips via acetone precipitation from 38 cases of corneal infection and 14 healthy controls. The cytokines and chemokines IL-1ß, IL-6, CXCL8 (IL-8), CX3CL1, IL-10, IL-12 (p70), IL-17A, and IL-23 were measured using an antibody bead assay. Results: A total of 512 genes were found to be differentially expressed in infected corneas compared to healthy corneas, with 508 being upregulated and four downregulated (fold-change (FC) <-2 or > 2 and adjusted p <0.01). For the conjunctiva, 477 were upregulated, and 3 were downregulated (FC <-3 or ≥ 3 and adjusted p <0.01). There was a significant overlap in cornea and conjunctiva gene expression in patients with corneal infections. The genes were predominantly associated with immune response, regulation of angiogenesis, and apoptotic signaling pathways. The most highly upregulated gene was CXCL8 (which codes for IL-8 protein). In patients with corneal infections, the concentration of IL-8 protein in tears was relatively higher in patients compared to healthy controls but did not show statistical significance. Conclusions: During corneal infection, many genes were upregulated, with most of them being associated with immune response, regulation of angiogenesis, and apoptotic signaling. The findings may facilitate the development of treatments for corneal infections that can dampen specific aspects of the immune response to reduce scarring and preserve sight.


Subject(s)
Conjunctiva , Cornea , Cytokines , Keratitis , Tears , Transcriptome , Humans , Tears/metabolism , Cytokines/metabolism , Cytokines/genetics , Cornea/metabolism , Cornea/immunology , Female , Male , Middle Aged , Adult , Conjunctiva/metabolism , Conjunctiva/immunology , Keratitis/genetics , Keratitis/immunology , Keratitis/metabolism , Aged , Gene Expression Profiling
7.
Ophthalmologie ; 121(7): 565-570, 2024 Jul.
Article in German | MEDLINE | ID: mdl-38639889

ABSTRACT

BACKGROUND: The aim of this study was to assess the impact of the ratio between the graft and host corneal size (RGH) on postoperative complications, such as immune reactions, re-bubbling rate and endothelial cell loss (ECL) after Descemet membrane endothelial keratoplasty (DMEK). PATIENTS AND METHODS: Retrospectively, 457 patient eyes were included which had undergone surgery between 2016 and 2019 in the Department of Ophthalmology, Saarland University Medical Center in Homburg/Saar using DMEK or triple DMEK, diagnosed as Fuchs' endothelial dystrophy (n = 431), pseudophakic bullous keratopathy (n = 9) and others (n = 17). The follow-up period extended until the end of 2020. Main outcome measures included immune reaction (IR), re-bubbling rate and the postoperative endothelial cell loss (ECL) at 6 weeks, 6 months and 12 months and whether these measures depended on the RGH. RESULTS: The RGH in this study ranged from 0.35 to 0.62 (0.46 ± 0.04). There were 33 (7.2%) postoperative IRs (DMEK n = 25; triple DMEK n = 8). The average RGH without IR (0.46 ± 0.04) was significantly (p = 0.038) smaller than in the group with IR (0.47 ± 0.05). Re-bubbling was necessary in 159 of 457 (34.8%) patient eyes. The RGH in patient eyes with re-bubbling (0.47 ± 0.04) was significantly (p = 0.014) higher than that in eyes without re-bubbling (0.45 ± 0.04). The mean preoperative endothelial cell count (ECD) was 2603 ± 251 cells/mm2 (min: 2161, max: 3500 cells/mm2). It was shown that a larger RGH had no positive influence on endothelial cell loss (r = 0.001; p = 0.974). CONCLUSION: Our results suggest that a larger graft diameter compared to host corneal size is associated with an increased rate of immune reactions and a higher re-bubbling rate after DMEK. Otherwise, a larger RGH had no positive influence on endothelial cell loss after DMEK. Accordingly, the graft size for DMEK should not be unnecessarily large, especially in eyes with Fuchs' endothelial dystrophy.


Subject(s)
Corneal Endothelial Cell Loss , Descemet Stripping Endothelial Keratoplasty , Graft Rejection , Postoperative Complications , Humans , Retrospective Studies , Male , Aged , Female , Middle Aged , Postoperative Complications/immunology , Postoperative Complications/pathology , Postoperative Complications/epidemiology , Graft Rejection/pathology , Graft Rejection/immunology , Corneal Endothelial Cell Loss/pathology , Corneal Endothelial Cell Loss/etiology , Cornea/pathology , Cornea/immunology , Cornea/surgery , Aged, 80 and over , Endothelium, Corneal/pathology , Endothelium, Corneal/immunology , Fuchs' Endothelial Dystrophy/surgery , Fuchs' Endothelial Dystrophy/pathology , Visual Acuity , Organ Size , Adult
8.
Front Biosci (Landmark Ed) ; 29(4): 146, 2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38682193

ABSTRACT

The ocular surface microenvironment, containing the cornea, conjunctiva, and lacrimal gland, constitutes the mucosal frontline of the eye and houses a myriad of immune cells. As a part of unconventional T cells, gamma delta (γδ) T cells differ in the development and functions from canonical alpha beta (αß) T cells. They are predominantly situated in mucosal sites throughout the body, including ocular surface tissues. Recent research has elucidated that γδ T cells serve as the primary interleukin-17A (IL-17A) source in the conjunctiva. They play a pivotal role in preserving ocular surface homeostasis and exhibit both protective and pathogenic roles in ocular surface diseases. This review delves into the general profiles of γδ T cells, their distribution in ocular surface tissues, and consolidates current insights into their functions in different conditions including dry eye disease, infectious keratitis, corneal wound healing, anterior chamber-associated immune deviation, allergic conjunctival disease, and diabetic ocular surface disease. The aim is to provide a systemic perspective on γδ T cells in the ocular surface microenvironment and outline potential directions for future studies.


Subject(s)
Homeostasis , Humans , Homeostasis/immunology , Conjunctiva/immunology , Animals , Eye Diseases/immunology , Intraepithelial Lymphocytes/immunology , Cornea/immunology , Dry Eye Syndromes/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , Receptors, Antigen, T-Cell, gamma-delta/metabolism
9.
Mucosal Immunol ; 17(3): 323-345, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38428739

ABSTRACT

Sleep deprivation (SD) has a wide range of adverse health effects. However, the mechanisms by which SD influences corneal pathophysiology and its post-wound healing remain unclear. This study aimed to examine the basic physiological characteristics of the cornea in mice subjected to SD and determine the pathophysiological response to injury after corneal abrasion. Using a multi-platform water environment method as an SD model, we found that SD leads to disturbances of corneal proliferative, sensory, and immune homeostasis as well as excessive inflammatory response and delayed repair after corneal abrasion by inducing hyperactivation of the sympathetic nervous system and hypothalamic-pituitary-adrenal axis. Pathophysiological changes in the cornea mainly occurred through the activation of the IL-17 signaling pathway. Blocking both adrenergic and glucocorticoid synthesis and locally neutralizing IL-17A significantly improved corneal homeostasis and the excessive inflammatory response and delay in wound repair following corneal injury in SD-treated mice. These results indicate that optimal sleep quality is essential for the physiological homeostasis of the cornea and its well-established repair process after injury. Additionally, these observations provide potential therapeutic targets to ameliorate SD-induced delays in corneal wound repair by inhibiting or blocking the activation of the stress system and its associated IL-17 signaling pathway.


Subject(s)
Corneal Injuries , Disease Models, Animal , Interleukin-17 , Signal Transduction , Sleep Deprivation , Wound Healing , Animals , Mice , Interleukin-17/metabolism , Sleep Deprivation/immunology , Corneal Injuries/metabolism , Corneal Injuries/etiology , Male , Cornea/metabolism , Cornea/immunology , Cornea/pathology , Inflammation/immunology , Hypothalamo-Hypophyseal System/metabolism , Pituitary-Adrenal System/metabolism , Mice, Inbred C57BL , Stress, Physiological
10.
J Immunol ; 211(3): 474-485, 2023 08 01.
Article in English | MEDLINE | ID: mdl-37326494

ABSTRACT

Herpetic stromal keratitis (HSK) is a painful and vision-impairing disease caused by recurrent HSV-1 infection of the cornea. The virus replication in the corneal epithelium and associated inflammation play a dominant role in HSK progression. Current HSK treatments targeting inflammation or virus replication are partially effective and promote HSV-1 latency, and long-term use can cause side effects. Thus, understanding molecular and cellular events that control HSV-1 replication and inflammation is crucial for developing novel HSK therapies. In this study, we report that ocular HSV-1 infection induces the expression of IL-27, a pleiotropic immunoregulatory cytokine. Our data indicate that HSV-1 infection stimulates IL-27 production by macrophages. Using a primary corneal HSV-1 infection mouse model and IL-27 receptor knockout mice, we show that IL-27 plays a critical role in controlling HSV-1 shedding from the cornea, the optimum induction of effector CD4+ T cell responses, and limiting HSK progression. Using in vitro bone marrow-derived macrophages, we show that IL-27 plays an antiviral role by regulating macrophage-mediated HSV-1 killing, IFN-ß production, and IFN-stimulated gene expression after HSV-1 infection. Furthermore, we report that IL-27 is critical for macrophage survival, Ag uptake, and the expression of costimulatory molecules involved in the optimum induction of effector T cell responses. Our results indicate that IL-27 promotes endogenous antiviral and anti-inflammatory responses and represents a promising target for suppressing HSK progression.


Subject(s)
Cornea , Interleukins , Keratitis, Herpetic , Animals , Female , Male , Mice , Cornea/immunology , Cornea/virology , Herpesvirus 1, Human , Interferon-beta/immunology , Interleukins/immunology , Keratitis, Herpetic/immunology , Macrophages/immunology , Mice, Knockout , Virus Shedding , Th1 Cells/immunology , Immunity, Innate
11.
Sci Rep ; 12(1): 494, 2022 01 11.
Article in English | MEDLINE | ID: mdl-35017561

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.


Subject(s)
Corneal Opacity/drug therapy , Corneal Opacity/etiology , Hepatocyte Growth Factor/administration & dosage , Keratitis/drug therapy , Lipopolysaccharides/adverse effects , Actins/genetics , Actins/immunology , Animals , Cornea/drug effects , Cornea/immunology , Corneal Opacity/genetics , Corneal Opacity/immunology , Cytokines/genetics , Cytokines/immunology , Disease Models, Animal , Humans , Keratitis/etiology , Keratitis/genetics , Keratitis/immunology , Mice , Mice, Inbred C57BL
12.
Exp Eye Res ; 216: 108950, 2022 03.
Article in English | MEDLINE | ID: mdl-35065982

ABSTRACT

Manually quantifying immune cells (ICs), commonly considered dendritic cells, in the corneal epithelium from in vivo confocal microscopy (IVCM) images can be influenced by observer bias. This study sought to evaluate the repeatability of manual IC quantification. Cell counts were first performed for 184 non-overlapping IVCM images by a single observer. Quantifications were undertaken to establish the total cell numbers per image, and the numbers of three cell morphological subtypes: mature ICs (with elongated dendrites), immature ICs (with short- or non-discernible dendrites) and globular cells (with large bodies and no visible dendrites). Cell counts were then repeated by the same observer, and independently undertaken by a second observer. Prior to these counts, both observers undertook an agreement 'training' process to define IC appearance and delineate the morphological subtypes. Total IC counts demonstrated excellent intra- and inter-observer reliability (intraclass correlation coefficients (ICC) > 0.90). Bland-Altman plots showed that interobserver measurement bias increased as a function of the total IC number in the image prior to consensus training. For total IC counts after the observer training process, there was no significant interobserver measurement bias. For IC morphological subtypes, there was a positive relationship between the mean inter-observer difference and average cell count for mature ICs and globular cells, but not immature ICs. In conclusion, higher variability in manual corneal IC counts exists when more cells are present in an IVCM image. Implementing an observer training process reduced inter-observer variability and minimised systematic measurement error.


Subject(s)
Cornea/immunology , Dendritic Cells/cytology , Microscopy, Confocal , Cell Count , Cornea/diagnostic imaging , Humans , Observer Variation , Professional Competence , Reproducibility of Results
13.
Am J Pathol ; 192(2): 270-280, 2022 02.
Article in English | MEDLINE | ID: mdl-34774519

ABSTRACT

Corneal transplantation is the most common form of tissue transplantation. The success of corneal transplantation mainly relies on the integrity of corneal endothelial cells (CEnCs), which maintain tissue transparency by pumping out excess water from the cornea. After transplantation, the rate of CEnC loss far exceeds that seen with normal aging, which can threaten sight. The underlying mechanisms are poorly understood. Alpha-melanocyte-stimulating hormone (α-MSH) is a neuropeptide that is constitutively found in the aqueous humor with both cytoprotective and immunomodulatory effects. The curent study found high expression of melanocortin 1 receptor (MC1R), the receptor for α-MSH, on CEnCs. The effect of α-MSH/MC1R signaling on endothelial function and allograft survival in vitro and in vivo was investigated using MC1R signaling-deficient mice (Mc1re/e mice with a nonfunctional MC1R). Herein, the results indicate that in addition to its well-known immunomodulatory effect, α-MSH has cytoprotective effects on CEnCs after corneal transplantation, and the loss of MC1R signaling significantly decreases long-term graft survival in vivo. In conclusion, α-MSH/MC1R signaling is critical for CEnC function and graft survival after corneal transplantation.


Subject(s)
Cornea/immunology , Corneal Transplantation , Endothelial Cells/immunology , Graft Survival/immunology , Signal Transduction/immunology , alpha-MSH/immunology , Animals , Cell Line, Transformed , Cornea/pathology , Female , Graft Survival/genetics , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Receptor, Melanocortin, Type 1/genetics , Receptor, Melanocortin, Type 1/immunology , Signal Transduction/genetics , alpha-MSH/genetics
14.
Front Immunol ; 12: 789454, 2021.
Article in English | MEDLINE | ID: mdl-34868077

ABSTRACT

Herpes simplex virus type-1 (HSV-1) ocular infection is one of the leading causes of infectious blindness in developed countries. The resultant herpetic keratitis (HK) is caused by an exacerbated reaction of the adaptive immune response that persists beyond virus clearance causing substantial damage to the cornea. Intramuscular immunization of mice with the HSV-1(VC2) live-attenuated vaccine strain has been shown to protect mice against lethal ocular challenge. Herein, we show that following ocular challenge, VC2 vaccinated animals control ocular immunopathogenesis in the absence of neutralizing antibodies on ocular surfaces. Ocular protection is associated with enhanced intracorneal infiltration of γδ T cells compared to mock-vaccinated animals. The observed γδ T cellular infiltration was inversely proportional to the infiltration of neutrophils, the latter associated with exacerbated tissue damage. Inhibition of T cell migration into ocular tissues by the S1P receptors agonist FTY720 produced significant ocular disease in vaccinated mice and marked increase in neutrophil infiltration. These results indicate that ocular challenge of mice immunized with the VC2 vaccine induce a unique ocular mucosal response that leads into the infiltration of γδ T cells resulting in the amelioration of infection-associated immunopathogenesis.


Subject(s)
Chemotaxis, Leukocyte , Cornea/immunology , Herpes Simplex Virus Vaccines/administration & dosage , Herpesvirus 1, Human/immunology , Intraepithelial Lymphocytes/immunology , Keratitis, Herpetic/prevention & control , Vaccination , Animals , Cornea/pathology , Cornea/virology , Cytokines/metabolism , Disease Models, Animal , Female , Herpes Simplex Virus Vaccines/immunology , Herpesvirus 1, Human/pathogenicity , Host-Pathogen Interactions , Injections, Intramuscular , Intraepithelial Lymphocytes/virology , Keratitis, Herpetic/immunology , Keratitis, Herpetic/pathology , Keratitis, Herpetic/virology , Lymphangiogenesis , Mice, Inbred BALB C , Neovascularization, Pathologic , Neutrophil Infiltration , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology
15.
Sci Rep ; 11(1): 22884, 2021 11 24.
Article in English | MEDLINE | ID: mdl-34819589

ABSTRACT

Immune cell infiltration has been implicated in neurotoxic chemotherapy for cancer treatment. However, our understanding of immune processes is still incomplete and current methods of observing immune cells are time consuming or invasive. Corneal dendritic cells are potent antigen-presenting cells and can be imaged with in-vivo corneal confocal microscopy. Corneal dendritic cell densities and nerve parameters in patients treated with neurotoxic chemotherapy were investigated. Patients treated for cancer with oxaliplatin (n = 39) or paclitaxel (n = 48), 3 to 24 months prior to assessment were recruited along with 40 healthy controls. Immature (ImDC), mature (MDC) and total dendritic cell densities (TotalDC), and corneal nerve parameters were analyzed from in-vivo corneal confocal microscopy images. ImDC was increased in the oxaliplatin group (Median, Md = 22.7 cells/mm2) compared to healthy controls (Md = 10.1 cells/mm2, p = 0.001), but not in the paclitaxel group (Md = 10.6 cells/mm2). ImDC was also associated with higher oxaliplatin cumulative dose (r = 0.33, p = 0.04) and treatment cycles (r = 0.40, p = 0.01). There was no significant difference in MDC between the three groups (p > 0.05). Corneal nerve parameters were reduced in both oxaliplatin and paclitaxel groups compared to healthy controls (p < 0.05). There is evidence of elevation of corneal ImDC in oxaliplatin-treated patients. Further investigation is required to explore this potential link through longitudinal studies and animal or laboratory-based immunohistochemical research.


Subject(s)
Antineoplastic Agents/adverse effects , Cornea/drug effects , Dendritic Cells/drug effects , Nerve Fibers/drug effects , Neurotoxicity Syndromes/etiology , Oxaliplatin/adverse effects , Paclitaxel/adverse effects , Aged , Case-Control Studies , Cornea/immunology , Cornea/innervation , Cornea/pathology , Cross-Sectional Studies , Dendritic Cells/immunology , Dendritic Cells/pathology , Female , Humans , Male , Microscopy, Confocal , Middle Aged , Nerve Fibers/pathology , Neurotoxicity Syndromes/immunology , Neurotoxicity Syndromes/pathology , Time Factors , Treatment Outcome
16.
J Immunol ; 207(11): 2868-2877, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34686582

ABSTRACT

The IL-36 cytokines are known to play various roles in mediating the immune response to infection in a tissue- and pathogen-dependent manner. The present study seeks to investigate the role of IL-36R signaling in C57BL/6 mouse corneas in response to Pseudomonas aeruginosa infection. IL-36α-/-, IL-36γ-/-, and IL-36R-/- mice had significantly more severe keratitis than wild-type mice. At six hours postinfection, IL-36α pretreatment augmented P. aeruginosa-induced expression of IL-1Ra, IL-36γ, LCN2, and S100A8/A9. At one day postinfection, exogenous IL-36α suppressed, whereas IL-36α deficiency promoted, the expression of IL-1ß. At three days postinfection, exogenous IL-36α suppressed Th1 but promoted Th2 immune response. IL-36α stimulated the infiltration of IL-22-expressing immune cells, and IL-22 neutralization resulted in more severe keratitis. IL-36α alone stimulated dendritic cell infiltration in B6 mouse corneas. Taken together, our study suggests that IL-36R signaling plays a protective role in the pathogenesis of P. aeruginosa keratitis by promoting the innate immune defense, Th2, and/or Th22/IL-22 immune responses. Exogenous IL-36α might be a potential therapy for improving the outcome of P. aeruginosa keratitis.


Subject(s)
Cornea/immunology , Interleukin-1/immunology , Pseudomonas Infections/immunology , Pseudomonas aeruginosa/immunology , Animals , Interleukin-1/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout
17.
J Immunol Res ; 2021: 5372090, 2021.
Article in English | MEDLINE | ID: mdl-34642632

ABSTRACT

Cornea is one of the most commonly transplanted tissues worldwide. However, it is usually omitted in the field of transplantology. Transplantation of the cornea is performed to treat many ocular diseases. It restores eyesight significantly improving the quality of life. Advancements in banking of explanted corneas and progressive surgical techniques increased availability and outcomes of transplantation. Despite the vast growth in the field of transplantation laboratory testing, standards for corneal transplantation still do not include HLA typing or alloantibody detection. This standard practice is based on immune privilege dogma that accounts for high success rates of corneal transplantation. However, the increasing need for retransplantation in high-risk patients with markedly higher risk of rejection causes ophthalmology transplantation centers to reevaluate their standard algorithms. In this review we discuss immune privilege mechanisms influencing the allograft acceptance and factors disrupting the natural immunosuppressive environment of the eye. Current developments in testing and immunosuppressive treatments (including cell therapies), when applied in corneal transplantation, may give very good results, decrease the possibility of rejection, and reduce the need for retransplantation, which is fairly frequent nowadays.


Subject(s)
Allografts/immunology , Cornea/immunology , Corneal Transplantation/adverse effects , Graft Rejection/prevention & control , Immune Privilege , Immunosuppression Therapy/methods , Animals , Corneal Transplantation/standards , Disease Models, Animal , Graft Rejection/immunology , Graft Survival/immunology , Histocompatibility Testing/standards , Humans , Practice Guidelines as Topic , Transplantation, Homologous/adverse effects , Transplantation, Homologous/standards
18.
Cells ; 10(9)2021 08 26.
Article in English | MEDLINE | ID: mdl-34571859

ABSTRACT

The kinetics of antigen-presenting cells (APCs) vary depending on their resident tissues and the manner of immunization. We investigated the long-term changes in mature APC and T-cell subsets over 4 weeks in the ocular surface in murine models of corneal quiescent or potent sterile inflammation, and allosensitization using partial (PT), syngeneic (Syn), and allogeneic (Allo) corneal transplantation. In PT, CD11bintCD11chiMHCIIhiCD86hi cells increased until 4 weeks with an increase in IFNγhi T cells. In Syn, both CD11bintCD11chiMHCIIhiCD86hi and CD11bhiCD11chiMHCIIhiCD86hi APC subsets increased until 4 weeks with a brief increase in CD69hi T cells at 2 weeks. In Allo, CD11bintCD11chiMHCIIhiCD86hi and CD11bhiCD11chiMHCIIhiCD86hi APC subsets increased until 4 weeks, and an early increase in CD69hi T cells was observed at 2 weeks followed by a late increase in IFNγhi T cells at 4 weeks. The frequency of the IFNγhi T cell subset was positively correlated with the frequency of the CD11bintCD11chiMHCIIhiCD86hi subset, indicating the existence of APC-T cell interaction in the ocular surface. Together, the results indicate that allosensitization in mature APCs leads to T-cell activation in the ocular surface, whereas sterile inflammation merely induces a brief and non-specific T-cell activation in the ocular surface.


Subject(s)
Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Cornea/immunology , Allografts/immunology , Animals , Antigen-Presenting Cells/physiology , Cell Movement , Corneal Transplantation/methods , Dendritic Cells/immunology , Female , Inflammation/metabolism , Lymphocyte Activation/immunology , Macrophages/immunology , Mice , Mice, Inbred BALB C , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
19.
Cells ; 10(8)2021 07 22.
Article in English | MEDLINE | ID: mdl-34440626

ABSTRACT

We investigated whether aging-dependent changes in dendritic cell (DC) distributions are distinct in autoimmune dry eye compared with an aging-related murine model. Corneal staining and tear secretion were evaluated in young and aged C57BL/6 (B6) and NOD.B10.H2b mice (NOD). In the corneolimbus, lacrimal gland (LG), and mesenteric lymph node (MLN), CD11b- and CD11b+ DCs, CD103+ DCs and MHC-IIhi B cells were compared between young and aged B6 and NOD mice. With increased corneal staining, tear secretion decreased in both aged B6 and NOD mice (p < 0.001). In both aged B6 and NOD mice, the percentages of corneolimbal CD11b+ DCs were higher (p < 0.05) than those in young mice. While, the percentages of lymph nodal CD103+ DCs were higher in aged B6 and NOD mice (p < 0.05), the percentages of corneolimbal CD103+ DCs were only higher in aged NOD mice (p < 0.05). In aged NOD mice, the proportions of lacrimal glandial and lymph nodal MHC-IIhi B cells were also higher than those in young mice (p < 0.05). It indicates that corneolimbal or lacrimal glandial distribution of CD103+ DCs or MHC-IIhi B cells may be distinct in aged autoimmune dry eye models compared to those in aged immune competent murine models.


Subject(s)
Autoimmune Diseases/immunology , Autoimmunity , Cornea/immunology , Dendritic Cells/immunology , Dry Eye Syndromes/immunology , Lacrimal Apparatus/immunology , Age Factors , Animals , Antigens, CD/metabolism , Autoimmune Diseases/metabolism , Autoimmune Diseases/pathology , CD11b Antigen/metabolism , Cornea/metabolism , Cornea/pathology , Dendritic Cells/metabolism , Disease Models, Animal , Dry Eye Syndromes/metabolism , Dry Eye Syndromes/pathology , Histocompatibility Antigens Class II/metabolism , Integrin alpha Chains/metabolism , Lacrimal Apparatus/metabolism , Lacrimal Apparatus/pathology , Male , Mice, Inbred C57BL , Mice, Inbred NOD , Phenotype , Tears/metabolism
20.
Front Immunol ; 12: 701935, 2021.
Article in English | MEDLINE | ID: mdl-34220866

ABSTRACT

The eye and the brain have limited capacities for regeneration and as such, immune-mediated inflammation can produce devastating consequences in the form of neurodegenerative diseases of the central nervous system or blindness as a result of ocular inflammatory diseases such as uveitis. Accordingly, both the eye and the brain are designed to limit immune responses and inflammation - a condition known as "immune privilege". Immune privilege is sustained by physiological, anatomical, and regulatory processes that conspire to restrict both adaptive and innate immune responses.


Subject(s)
CD11c Antigen/immunology , Cornea/immunology , Dendritic Cells/immunology , Immune Privilege/immunology , Animals , Humans , Immunity, Innate/immunology , Inflammation/immunology
SELECTION OF CITATIONS
SEARCH DETAIL