Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Cells ; 13(7)2024 Apr 04.
Article in English | MEDLINE | ID: mdl-38607062

ABSTRACT

Limbal epithelial progenitor cells (LEPC) rely on their niche environment for proper functionality and self-renewal. While extracellular vesicles (EV), specifically small EVs (sEV), have been proposed to support LEPC homeostasis, data on sEV derived from limbal niche cells like limbal mesenchymal stromal cells (LMSC) remain limited, and there are no studies on sEVs from limbal melanocytes (LM). In this study, we isolated sEV from conditioned media of LMSC and LM using a combination of tangential flow filtration and size exclusion chromatography and characterized them by nanoparticle tracking analysis, transmission electron microscopy, Western blot, multiplex bead arrays, and quantitative mass spectrometry. The internalization of sEV by LEPC was studied using flow cytometry and confocal microscopy. The isolated sEVs exhibited typical EV characteristics, including cell-specific markers such as CD90 for LMSC-sEV and Melan-A for LM-sEV. Bioinformatics analysis of the proteomic data suggested a significant role of sEVs in extracellular matrix deposition, with LMSC-derived sEV containing proteins involved in collagen remodeling and cell matrix adhesion, whereas LM-sEV proteins were implicated in other cellular bioprocesses such as cellular pigmentation and development. Moreover, fluorescently labeled LMSC-sEV and LM-sEV were taken up by LEPC and localized to their perinuclear compartment. These findings provide valuable insights into the complex role of sEV from niche cells in regulating the human limbal stem cell niche.


Subject(s)
Extracellular Vesicles , Mesenchymal Stem Cells , Humans , Proteomics/methods , Mesenchymal Stem Cells/metabolism , Stem Cells , Melanocytes , Extracellular Vesicles/metabolism
2.
Int J Mol Sci ; 24(23)2023 Nov 28.
Article in English | MEDLINE | ID: mdl-38069177

ABSTRACT

Organ culture storage techniques for corneoscleral limbal (CSL) tissue have improved the quality of corneas for transplantation and allow for longer storage times. Cultured limbal tissue has been used for stem cell transplantation to treat limbal stem cell deficiency (LSCD) as well as for research purposes to assess homeostasis mechanisms in the limbal stem cell niche. However, the effects of organ culture storage conditions on the quality of limbal niche components are less well described. Therefore, in this study, the morphological and immunohistochemical characteristics of organ-cultured limbal tissue are investigated and compared to fresh limbal tissues by means of light and electron microscopy. Organ-cultured limbal tissues showed signs of deterioration, such as edema, less pronounced basement membranes, and loss of the most superficial layers of the epithelium. In comparison to the fresh limbal epithelium, organ-cultured limbal epithelium showed signs of ongoing proliferative activity (more Ki-67+ cells) and exhibited an altered limbal epithelial phenotype with a loss of N-cadherin and desmoglein expression as well as a lack of precise staining patterns for cytokeratin ((CK)14, CK17/19, CK15). The analyzed extracellular matrix composition was mainly intact (collagen IV, fibronectin, laminin chains) except for Tenascin-C, whose expression was increased in organ-cultured limbal tissue. Nonetheless, the expression patterns of cell-matrix adhesion proteins varied in organ-cultured limbal tissue compared to fresh limbal tissue. A decrease in the number of melanocytes (Melan-A+ cells) and Langerhans cells (HLA-DR+, CD1a+, CD18+) was observed in the organ-cultured limbal tissue. The organ culture-induced alterations of the limbal epithelial stem cell niche might hamper its use in the treatment of LSCD as well as in research studies. In contrast, reduced numbers of donor-derived Langerhans cells seem associated with better clinical outcomes. However, there is a need to consider the preferential use of fresh CSL for limbal transplants and to look at ways of improving the limbal stem cell properties of stored CSL tissue.


Subject(s)
Epithelium, Corneal , Humans , Organ Culture Techniques , Epithelium, Corneal/metabolism , Stem Cells/metabolism , Stem Cell Niche , Limbal Stem Cells , Epithelial Cells , Cells, Cultured
3.
Int Ophthalmol ; 43(12): 4551-4562, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37684398

ABSTRACT

PURPOSE: Hypoxia-inducible factors (HIFs) are considered to play a significant role in the pathogenesis of pterygium. The aim of this study was to investigate the relative expression or immunoreactivity of HIF1α and HIF2α in the epithelium of primary pterygium, recurrences and healthy conjunctiva. METHODS: Immunohistochemical staining was performed with antibodies against HIF1α and HIF2α, respectively, on 55/84 primary pterygium specimens, 6/28 recurrences and 20/20 control tissues (healthy conjunctiva). RESULTS: Immunohistochemical staining revealed lower epithelial immunoreactivity of HIF1α and HIF2α in both primary pterygium (11% and 38%) and recurrences (18% and 21%) when compared to healthy conjunctival tissue (46% and 66%). Differences between immunoreactivity of HIF1α and of HIF2α in primary pterygium and controls were each highly significant (p < .001). Within the group of primary pterygium, epithelial immunoreactivity of HIF2α (38%) was significantly higher than that of HIF1α (11%). In recurrent pterygium and healthy conjunctiva, immunoreactivity levels of HIF2α were higher than those of HIF1α as well; however, differences between both isoforms were not significant. CONCLUSION: Our study shows evidence that the higher expressed epithelial HIF2α, rather than HIF1α, and the balance between both HIF isoforms might be relevant factors associated with pathogenesis of primary pterygium. Modulation of HIF2α levels and activity may thus offer a new therapeutic approach to the treatment of advancing pterygium where the initial stage with its HIF1-peak has already passed.


Subject(s)
Pterygium , Humans , Pterygium/metabolism , Epithelium/pathology , Conjunctiva/pathology , Protein Isoforms/metabolism
4.
Int J Mol Sci ; 24(8)2023 Apr 19.
Article in English | MEDLINE | ID: mdl-37108705

ABSTRACT

The porcine ocular surface is used as a model of the human ocular surface; however, a detailed characterization of the porcine ocular surface has not been documented. This is due, in part, to the scarcity of antibodies produced specifically against the porcine ocular surface cell types or structures. We performed a histological and immunohistochemical investigation on frozen and formalin-fixed, paraffin-embedded ocular surface tissue from domestic pigs using a panel of 41 different antibodies related to epithelial progenitor/differentiation phenotypes, extracellular matrix and associated molecules, and various niche cell types. Our observations suggested that the Bowman's layer is not evident in the cornea; the deep invaginations of the limbal epithelium in the limbal zone are analogous to the limbal interpalisade crypts of human limbal tissue; and the presence of goblet cells in the bulbar conjunctiva. Immunohistochemistry analysis revealed that the epithelial progenitor markers cytokeratin (CK)15, CK14, p63α, and P-cadherin were expressed in both the limbal and conjunctival basal epithelium, whereas the basal cells of the limbal and conjunctival epithelium did not stain for CK3, CK12, E-cadherin, and CK13. Antibodies detecting marker proteins related to the extracellular matrix (collagen IV, Tenascin-C), cell-matrix adhesion (ß-dystroglycan, integrin α3 and α6), mesenchymal cells (vimentin, CD90, CD44), neurons (neurofilament), immune cells (HLA-ABC; HLA-DR, CD1, CD4, CD14), vasculature (von Willebrand factor), and melanocytes (SRY-homeobox-10, human melanoma black-45, Tyrosinase) on the normal human ocular surface demonstrated similar immunoreactivity on the normal porcine ocular surface. Only a few antibodies (directed against N-cadherin, fibronectin, agrin, laminin α3 and α5, melan-A) appeared unreactive on porcine tissues. Our findings characterize the main immunohistochemical properties of the porcine ocular surface and provide a morphological and immunohistochemical basis useful to research using porcine models. Furthermore, the analyzed porcine ocular structures are similar to those of humans, confirming the potential usefulness of pig eyes to study ocular surface physiology and pathophysiology.


Subject(s)
Limbus Corneae , Swine , Humans , Animals , Cornea , Conjunctiva/metabolism , Extracellular Matrix , Sus scrofa , Epithelial Cells/metabolism
5.
Cells ; 11(12)2022 06 20.
Article in English | MEDLINE | ID: mdl-35741104

ABSTRACT

Interactions between limbal epithelial progenitor cells (LEPC) and surrounding niche cells, which include limbal mesenchymal stromal cells (LMSC) and melanocytes (LM), are essential for the maintenance of the limbal stem cell niche required for a transparent corneal surface. P-cadherin (P-cad) is a critical stem cell niche adhesion molecule at various epithelial stem cell niches; however, conflicting observations were reported on the presence of P-cad in the limbal region. To explore this issue, we assessed the location and phenotype of P-cad+ cells by confocal microscopy of human corneoscleral tissue. In subsequent fluorescence-activated cell sorting (FACS) experiments, we used antibodies against P-cad along with CD90 and CD117 for the enrichment of LEPC, LMSC and LM, respectively. The sorted cells were characterized by immunophenotyping and the repopulation of decellularized limbal scaffolds was evaluated. Our findings demonstrate that P-cad is expressed by epithelial progenitor cells as well as melanocytes in the human limbal epithelial stem cell niche. The modified flow sorting addressing P-cad as well as CD90 and CD117 yielded enriched LEPC (CD90-CD117-P-cad+) and pure populations of LMSC (CD90+CD117-P-cad-) and LM (CD90-CD117+P-cad+). The enriched LEPC showed the expression of epithelial progenitor markers and better colony-forming ability than their P-cad- counterparts. The cultured LEPC and LM exhibited P-cad expression at intercellular junctions and successfully repopulated decellularized limbal scaffolds. These data suggest that P-cad is a critical cell-cell adhesion molecule, connecting LEPC and LM, which may play an important role in the long-term maintenance of LEPC at the limbal stem cell niche; moreover, these findings led to further improvement of cell enrichment protocols to enhance the yield of LEPC.


Subject(s)
Limbus Corneae , Cadherins/metabolism , Cell Adhesion Molecules/metabolism , Humans , Melanocytes/metabolism , Stem Cell Niche , Stem Cells
6.
Z Naturforsch C J Biosci ; 77(11-12): 493-500, 2022 Nov 25.
Article in English | MEDLINE | ID: mdl-35767310

ABSTRACT

Diabetes is characterized by hyperglycemia and a significant risk of vascular complications. Vascular endothelial growth factor (VEGF) and its main receptor VEGFR2 (KDR), which is highly expressed in vascular endothelial cells, are essential mediators of vascular maintenance and angiogenesis. During glycolysis after high calorie food intake, methylglyoxal (MGO) is formed and MGO blood levels are elevated in diabetes. MGO reacts with arginine residues to generate MG-H1 or with lysine residues to carboxyethyl lysine which are common components of advanced glycation end-products. Therefore, the question arises whether hyperglycemic conditions affect VEGF signaling via a ligand-independent direct modification of signaling components. As a first step, the effect of MGO on VEGFR2 activation was investigated in cultured endothelial cells from human umbilical vein by determination of VEGFR2 phosphorylation at selected tyrosine residues by ELISA and immunoblotting using phospho-specific antibodies. Phosphorylation of VEGFR2-Y996, VEGFR2-Y1054, or VEGFR2-Y1175 reached a maximum 5 min after stimulation of endothelial cells with VEGF. Phosphorylation was significantly inhibited by 100 µM MGO and to a lesser extent by high glucose treatment. 2,3-Pentanedione and glyoxal were investigated for comparison. In summary, VEGFR2 phosphorylation is sensitive to MGO or high glucose concentrations which may be relevant in the pathophysiology of microvascular disease in diabetes.


Subject(s)
Diabetes Mellitus , Pyruvaldehyde , Humans , Pyruvaldehyde/chemistry , Pyruvaldehyde/pharmacology , Phosphorylation , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/pharmacology , Endothelial Cells/metabolism , Tyrosine/metabolism , Tyrosine/pharmacology , Lysine/metabolism , Lysine/pharmacology , Magnesium Oxide/pharmacology , Glucose/pharmacology
7.
Invest Ophthalmol Vis Sci ; 63(5): 17, 2022 05 02.
Article in English | MEDLINE | ID: mdl-35579905

ABSTRACT

Purpose: Proliferative vitreoretinopathy (PVR) remains an unresolved clinical challenge and can lead to frequent revision surgery and blindness vision loss. The aim of this study was to characterize the microenvironment of epiretinal PVR tissue, in order to shed more light on the complex pathophysiology and to unravel new treatment options. Methods: A total of 44 tissue samples were analyzed in this study, including 19 epiretinal PVRs, 13 epiretinal membranes (ERMs) from patients with macular pucker, as well as 12 internal limiting membranes (ILMs). The cellular and molecular microenvironment was assessed by cell type deconvolution analysis (xCell), RNA sequencing data and single-cell imaging mass cytometry. Candidate drugs for PVR treatment were identified in silico via a transcriptome-based drug-repurposing approach. Results: RNA sequencing of tissue samples demonstrated distinct transcriptional profiles of PVR, ERM, and ILM samples. Differential gene expression analysis revealed 3194 upregulated genes in PVR compared with ILM, including FN1 and SPARC, which contribute to biological processes, such as extracellular matrix (ECM) organization. The xCell and IMC analyses showed that PVR membranes were composed of macrophages, retinal pigment epithelium, and α-SMA-positive myofibroblasts, the latter predominantly characterized by the co-expression of immune cell signature markers. Finally, 13 drugs were identified as potential therapeutics for PVR, including aminocaproic acid and various topoisomerase-2A inhibitors. Conclusions: Epiretinal PVR membranes exhibit a unique and complex transcriptional and cellular profile dominated by immune cells and myofibroblasts, as well as a variety of ECM components. Our findings provide new insights into the pathophysiology of PVR and suggest potential targeted therapeutic options.


Subject(s)
Epiretinal Membrane , Vitreoretinopathy, Proliferative , Epiretinal Membrane/metabolism , Humans , RNA/genetics , Retina/metabolism , Retinal Pigment Epithelium/metabolism , Vitreoretinopathy, Proliferative/metabolism
8.
Sci Rep ; 11(1): 19140, 2021 09 27.
Article in English | MEDLINE | ID: mdl-34580409

ABSTRACT

Despite the reported low expression of the primary SARS-CoV-2 receptor ACE2 in distinct ocular tissues, some clinical evidence suggests that SARS-CoV-2 can infect the eye. In this study, we explored potential entry sites for SARS-CoV-2 by viral S protein histochemistry on various ocular tissues and compared the staining patterns with RNA and protein expression of TMPRSS2 and ACE2. Potential viral entry sites were investigated by histochemistry using tagged recombinant viral S protein on 52 ocular tissue samples including specimens of the cornea, conjunctiva, lid margin, lacrimal gland tissue, retina, choroid, and RPE. In addition, ACE2 and TMPRSS2 immunohistochemistry were performed on the same ocular tissue, each with distinct antibodies binding to different epitopes. Lung tissue samples were used as positive controls. Finally, bulk RNA sequencing (RNA-Seq) was used to determine the expression of ACE2 and its auxiliary factors in the tissues mentioned above. S protein histochemistry revealed a positive staining in lung tissue but absent staining in the cornea, the conjunctiva, eye lid samples, the lacrimal glands, the retina and the optic nerve which was supported by hardly any immunoreactivity for ACE2 and TMPRSS2 and scarce ACE2 and TMPRSS2 RNA expression. Negligible staining with antibodies targeting ACE2 or TMPRSS2 was seen in the main and accessory lacrimal glands. In contrast, ocular staining (S protein, ACE2, TMPRSS2) was distinctly present in pigmented cells of the RPE and choroid, as well as in the ciliary body and the iris stroma. S protein histochemistry revealed hardly any SARS-CoV-2 entry sites in all ocular tissues examined. Similarly, no significant ACE2 or TMPRSS2 expression was found in extra- and intraocular tissue. While this study suggest a rather low risk of ocular infection with SARS-CoV-2, it should be noted, that potential viral entry sites may increase in response to inflammation or in certain disease states.


Subject(s)
COVID-19/prevention & control , Conjunctiva/metabolism , Cornea/metabolism , SARS-CoV-2/metabolism , Spike Glycoprotein, Coronavirus/metabolism , Angiotensin-Converting Enzyme 2/genetics , Angiotensin-Converting Enzyme 2/metabolism , COVID-19/virology , Conjunctiva/virology , Cornea/virology , Gene Expression Profiling/methods , Humans , Immunohistochemistry/methods , RNA-Seq/methods , SARS-CoV-2/physiology , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Virus Internalization
9.
Ophthalmologe ; 118(Suppl 1): 85-88, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33141332

ABSTRACT

Recent studies have described conjunctivitis in approximately 1% of COVID-19 patients and speculated that SARS-CoV­2 can be transmitted via the conjunctiva. In this article we recapitulate the molecular mechanisms of host cell entry of SARS-CoV­2 and discuss the current evidence for a potential conjunctival transmission of SARS-CoV­2. The current body of evidence indicates that SARS-CoV­2 requires the membrane-bound angiotensin-converting enzyme 2 (ACE2) and the membrane-bound serine protease TMPRSS2 to enter cells. Recent studies suggest that COVID-19 patients rarely exhibit viral RNA in tear film and conjunctival smears and that, ACE2 and TMPRSS2 are only expressed in small amounts in the conjunctiva, making conjunctival infection with SARS-CoV­2 via these mediators unlikely. Nevertheless, we consider the current evidence to be still too limited to provide a conclusive statement and recommend appropriate protective measures for healthcare personnel who are in close contact with suspected and confirmed COVID-19 patients.


Subject(s)
COVID-19 , SARS-CoV-2 , Conjunctiva , Humans
10.
Mol Vis ; 26: 731-741, 2020.
Article in English | MEDLINE | ID: mdl-33273800

ABSTRACT

Purpose: Retinal vein occlusions (RVOs) are a common disease, but there are no animal models for spontaneous RVO formation. The critical sites of predilection, especially for branch RVO (BRVO), are the arteriovenous crossing sites in the inner retina. To gain more insight into possible animal models, the anatomic structure of retinal arteriovenous crossings was investigated in mice, rats, and pigs and compared to the human situation. Methods: Retinal flat mounts and paraffin sections of eyes from mice, rats, pigs, and humans were stained with GS lectin, Masson's trichrome, or immunohistochemistry for ACTA2 and GFAP. Serial sections of arteriovenous crossing sites were investigated. Results: Mice usually do not show retinal arteriovenous crossings. Rats have a mean of 2.8±1.4 crossings per eye at a mean distance from the optic nerve head of 2.79±0.53 mm, though the diameters of the crossing vessels are small. The situation in pigs is similar to that in humans, with many arteriovenous crossings of vessels and with similar diameters as found in humans. A mean of 28.4±3.5 crossings per retina was found, and 23% of these were arterial overcrossings. Serial paraffin sections showed that the tunica media of the artery touched that of the vein, but they did not fuse. Conclusions: While the retinal arteriovenous crossings of mice and rats are absent or comprised of rather thin vessels, those in the porcine retina are similar to adult humans. Therefore, the porcine retinal vascular bed may serve as a model to assess early steps in the formation of RVOs.


Subject(s)
Retinal Artery/abnormalities , Retinal Vein/abnormalities , Aged , Animals , Female , Humans , Mice, Inbred C57BL , Rats, Sprague-Dawley , Species Specificity , Swine
11.
Ophthalmologe ; 117(7): 626-630, 2020 Jul.
Article in German | MEDLINE | ID: mdl-32572552

ABSTRACT

Recent studies have described conjunctivitis in approximately 1% of COVID-19 patients and speculated that SARS-CoV­2 can be transmitted via the conjunctiva. In this article we recapitulate the molecular mechanisms of host cell entry of SARS-CoV­2 and discuss the current evidence for a potential conjunctival transmission of SARS-CoV­2. The current body of evidence indicates that SARS-CoV­2 requires the membrane-bound angiotensin-converting enzyme 2 (ACE2) and the membrane-bound serine protease TMPRSS2 to enter cells. Recent studies suggest that COVID-19 patients rarely exhibit viral RNA in tear film and conjunctival smears and that, ACE2 and TMPRSS2 are only expressed in very small amounts in the conjunctiva, making conjunctival infection with SARS-CoV­2 via these mediators unlikely. Nevertheless, we consider the current evidence to be still too limited to provide a conclusive statement and recommend appropriate protective measures for healthcare personnel who are in close contact with suspected and confirmed COVID-19 patients.


Subject(s)
Betacoronavirus , Conjunctiva , Coronavirus Infections , Pandemics , Pneumonia, Viral , COVID-19 , Humans , SARS-CoV-2
12.
J Med Virol ; 92(10): 2081-2086, 2020 10.
Article in English | MEDLINE | ID: mdl-32374427

ABSTRACT

SARS-CoV-2 is assumed to use angiotensin-converting enzyme 2 (ACE2) and other auxiliary proteins for cell entry. Recent studies have described conjunctival congestion in 0.8% of patients with laboratory-confirmed severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), and there has been speculation that SARS-CoV-2 can be transmitted through the conjunctiva. However, it is currently unclear whether conjunctival epithelial cells express ACE2 and its cofactors. In this study, a total of 38 conjunctival samples from 38 patients, including 12 healthy conjunctivas, 12 melanomas, seven squamous cell carcinomas, and seven papilloma samples, were analyzed using high-throughput RNA sequencing to assess messenger RNA (mRNA) expression of the SARS-CoV-2 receptor ACE2 and its cofactors including TMPRSS2, ANPEP, DPP4, and ENPEP. ACE2 protein expression was assessed in eight healthy conjunctival samples using immunohistochemistry. Our results show that the SARS-CoV-2 receptor ACE2 is not substantially expressed in conjunctival samples on the mRNA (median: 0.0 transcripts per million [TPM], min: 0.0 TPM, max: 1.7 TPM) and protein levels. Similar results were obtained for the transcription of other auxiliary molecules. In conclusion, this study finds no evidence for a significant expression of ACE2 and its auxiliary mediators for cell entry in conjunctival samples, making conjunctival infection with SARS-CoV-2 via these mediators unlikely.


Subject(s)
COVID-19/virology , Carcinoma, Squamous Cell/virology , Eye Neoplasms/virology , Melanoma/virology , Papilloma/virology , Receptors, Virus/genetics , Adult , Aged , Aged, 80 and over , Angiotensin-Converting Enzyme 2/genetics , Angiotensin-Converting Enzyme 2/metabolism , COVID-19/complications , COVID-19/pathology , COVID-19/surgery , Carcinoma, Squamous Cell/complications , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/surgery , Case-Control Studies , Conjunctiva/pathology , Conjunctiva/surgery , Dipeptidyl Peptidase 4/genetics , Dipeptidyl Peptidase 4/metabolism , Eye Neoplasms/complications , Eye Neoplasms/pathology , Eye Neoplasms/surgery , Gene Expression , Glutamyl Aminopeptidase/genetics , Glutamyl Aminopeptidase/metabolism , Humans , Immunohistochemistry , Male , Melanoma/complications , Melanoma/pathology , Melanoma/surgery , Middle Aged , Papilloma/complications , Papilloma/pathology , Papilloma/surgery , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Virus/metabolism , SARS-CoV-2/genetics , SARS-CoV-2/metabolism , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism
13.
Macromol Rapid Commun ; 41(3): e1900352, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31894619

ABSTRACT

The authors describe a process that may eventually reduce the risk of scar formation after glaucoma surgery. For this, a thin hydrogel coating is photochemically generated and linked to the sclera surface at the surgical site. This coating is generated from a photoreactive prepolymer containing anthraquinone groups, which is administered as a thin pad to the sclera surface. Short UV irradiation leads to a reaction of the photogroups with neighboring chains via C-H insertion crosslinking, thus transforming the precursor polymer into a hydrogel. Simultaneously, a reaction between the photogroups and the underlying sclera tissue occurs, so that the hydrogel patch becomes covalently linked to the tissue. The authors show that the resulting thin coating is strongly cell repellent and hinders tenon fibroblasts to form tenon tissue at the site of the coating and is suitable for inclusion into a surgical procedure.


Subject(s)
Cicatrix/prevention & control , Glaucoma/surgery , Sclera/surgery , Animals , Cell Adhesion , Epithelial Cells , Fibroblasts , Humans , Hydrogels/administration & dosage , Hydrogels/chemistry , Retina/surgery , Risk , Swine , Tenon Capsule/surgery , Ultraviolet Rays
14.
Ophthalmic Res ; 61(3): 174-182, 2019.
Article in English | MEDLINE | ID: mdl-29050001

ABSTRACT

PURPOSE: Anti-angiogenic treatment is well established in the management of exudative age-related macular degeneration (AMD), but not sufficient in all patients. The characterisation of factors driving this chronic disease could serve to identify additional treatment options. The purpose of this study was to assess gene expression patterns and distinct changes in cells derived from surgically extracted choroidal neovascularisation (CNV) membranes. MATERIALS AND METHODS: The expression of >11,000 genes was analysed by means of a microarray in cells cultured from 2 late-stage CNV membranes compared to primary human retinal pigment epithelium (RPE) and ARPE-19 cells. A pathway analysis was performed to identify gene expression patterns associated with exudative AMD. RESULTS: The analysis revealed significant alterations in gene sets associated with inflammatory processes in CNV-derived cells, involving the upregulation of pro-inflammatory factors IL6, C3, and C5, and downregulation of anti-inflammatory complement factor B and complement factor I. Factors associated with angiogenesis, such as VEGFA or ANGPT2, were not significantly regulated in the 2 RPE-derived cell lines. CONCLUSION: In late-stage CNV membrane-derived RPE, gene expression was shifted towards a pro-inflammatory state. Angiogenesis-associated factors were regulated differently in the 2 CNV-derived RPE membranes. While inflammation seems to be continuously stimulated by RPE associated with late exudative AMD, this appears not to be the case with regard to angioregulatory mechanisms.


Subject(s)
Angiogenic Proteins/genetics , Choroidal Neovascularization/genetics , Gene Expression/physiology , Inflammation Mediators/metabolism , Retinal Pigment Epithelium/metabolism , Aged, 80 and over , Angiogenic Proteins/metabolism , Cells, Cultured , Female , Gene Expression Profiling , Humans , Male , Real-Time Polymerase Chain Reaction , Retinal Pigment Epithelium/pathology
15.
PLoS One ; 13(3): e0191338, 2018.
Article in English | MEDLINE | ID: mdl-29529099

ABSTRACT

PURPOSE: Retinal vein occlusion (RVO) has been investigated in several laser-induced animal models using pigs, rabbits and rats. However, laser-induced RVO has been rarely reported in mice, despite the impressive number of available mutants, ease of handling and cost effectiveness. The aim of this study was to further assess the feasibility of a RVO mouse model for gene expression analysis and its possible use to investigate effects of hypoxia. METHODS: C57Bl/6J mice were injected with eosin Y for photo-sensitization. Subsequently, large retinal veins were laser-treated in one eye to induce vascular occlusion. Contralateral control eyes received non-occlusive retinal laser treatment sparing large vessels. The animals were followed for up to eight days and assessed by funduscopy, angiography, hypoxyprobe staining, histopathology and gene expression analysis by qPCR and RNA sequencing (RNAseq). Another group of mice was left untreated and studied at a single time point to determine baseline characteristics. RESULTS: Laser-induced RVO persisted in half of the treated veins for three days, and in a third of the veins for the whole observation period of 8 days. Funduscopy revealed large areas of retinal swelling in all laser-treated eyes, irrespective of vascular targeting or occlusion status. Damage of the outer retina, retinal pigment epithelium (RPE), and even choroid and sclera at the laser site was observed in histological sections. Genes associated with inflammation or cell damage were highly up-regulated in all laser-treated eyes as detected by RNAseq and qPCR. Retinal hypoxia was observed by hypoxyprobe staining in all RVO eyes for up to 5 days with a maximal extension at days 2 and 3, but no significant RVO-dependent changes in gene expression were detected for angiogenesis- or hypoxia-related genes. CONCLUSION: The laser-induced RVO mouse model is characterized by a predominant general inflammatory and tissue damage response, which may obscure distinct hypoxia- and angiogenesis-related effects. A non-occlusive laser treatment control is essential to allow for proper data interpretation and should be mandatory in animal studies of laser-induced RVO to dissect laser-induced tissue damage from vascular occlusion effects.


Subject(s)
Retinal Vein Occlusion/metabolism , Animals , Disease Models, Animal , Disease Progression , Gene Expression Profiling , Lasers , Mice, Inbred C57BL , RNA, Messenger/metabolism , Retinal Vein Occlusion/pathology , Time Factors
16.
BMC Ophthalmol ; 16(1): 159, 2016 Sep 05.
Article in English | MEDLINE | ID: mdl-27596098

ABSTRACT

BACKGROUND: Omega-3 polyunsaturated fatty acids (PUFAs) have a highly anti-angiogenic effect in animal models. However, the clinical relevance of omega-3PUFAs in human retinal pathologies remains unclear. The ARED 2 study found no effect of omega-3 PUFA supplementation on progression of age related macular degeneration (AMD). The aim of this study was to compare serum levels of omega-3- and omega-6 PUFAs between patients with diabetic retinopathy (DR), AMD and retinal vein occlusion (RVO), and to identify potential confounders of serum level measurements. METHODS: Venous blood samples were collected from 44 patients with DR, 25 with AMD, 12 with RVO and 27 controls. The lipid phase was extracted and analyzed using mass spectrometry. Retinal disease staging was done by indirect funduscopy and FAG where appropriate. Patient demographics and medical history including current medication and fasting state were acquired. Tukey contrasts for multiple comparisons of the mean and linear regression analysis were used for statistical analysis. RESULTS: Our data revealed no significant differences in omega-6 PUFA serum levels between patients with AMD, DR, RVO and controls (p > 0.858). Uncorrected omega-3 PUFA levels were significantly higher in patients with AMD compared to DR but not compared to controls (p = 0.004). However, after correcting for possible confounders such as body mass index (BMI), age, sex, fasting and use of statins, no statistically significant difference remained for serum omega-3 PUFA levels. Fasting was identified as an independent confounder of total omega-6 PUFAs, three individual omega-6 PUFAs and one omega-3 PUFA(p < 0.0427). Statin use was identified as an independent confounder of α-linolenic acid (an omega-3PUFA; p = 0.0210). CONCLUSION: In this pilot study with relatively low patient numbers, we report significant differences in serum levels of omega-3PUFAs among patients with different types of retinal diseases. However, these differences were not robust for disease specificity after correction for possible confounders in our cohort. Our results demonstrate that serum lipid profiles need to be interpreted with caution since they are significantly altered by variables like fasting and medication use independent from the underlying disease. Correcting for respective confounders is thus necessary to compare serum lipid profiles in clinical studies.


Subject(s)
Diabetic Retinopathy/blood , Fatty Acids, Omega-3/blood , Fatty Acids, Omega-6/blood , Macular Degeneration/blood , Retinal Vein Occlusion/blood , Case-Control Studies , Female , Humans , Male , Mass Spectrometry , Pilot Projects , Regression Analysis
17.
Invest Ophthalmol Vis Sci ; 57(10): 4017-26, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27494343

ABSTRACT

PURPOSE: Retinal vascular disease represents a major cause for vision loss in the Western world. Recent research has shown that neuronal and vascular damage are closely related in retinal disease. Ciliary neurotrophic factor (CNTF) is a well-studied neurotrophic factor that is currently being tested in clinical trials for the treatment of retinal degenerative diseases and macular telangiectasia. However, little is known about its effect on retinal vasculature. In this study, we investigate the effects of CNTF in retinal neovascular disease using the mouse model of oxygen-induced retinopathy (OIR). METHODS: Newborn pups were exposed to 75% oxygen from postnatal day (P)7 to P12 and subsequently returned to room air. Ciliary neurotrophic factor was injected intravitreally at OIR P12 and the vaso-obliterated and neovascular areas were quantified at OIR P17. Immunohistochemistry, RNA, and protein analysis were used to identify CNTF-responsive cells. In vitro experiments were performed to analyze the effect of CNTF on endothelial and astroglial cells. RESULTS: In the OIR model, CNTF facilitated capillary regrowth and attenuated preretinal neovascularization in a dose-dependent manner. The protective effect of CNTF was mediated via activation of the JAK/STAT3/SOCS3 signaling pathway. Immunohistochemical studies identified endothelial cells among others as CNTF-responsive cells in the retina. In vitro studies confirmed the anti-angiogenic effect of CNTF on endothelial cell sprouting. CONCLUSIONS: This study provides evidence for a therapeutic potential of CNTF beyond degenerative retinal disease. Vasoproliferative retinopathies may benefit from a CNTF-dependent and SOCS3-mediated angiomodulatory effect.


Subject(s)
Ciliary Neurotrophic Factor/pharmacology , Gene Expression Regulation , Retinal Degeneration/drug therapy , Retinal Neovascularization/drug therapy , Retinal Vessels/pathology , Suppressor of Cytokine Signaling 3 Protein/genetics , Up-Regulation , Animals , Animals, Newborn , Cells, Cultured , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Humans , Immunohistochemistry , Mice, Inbred C57BL , Oxygen/toxicity , Polymerase Chain Reaction , RNA/genetics , Recombinant Proteins/pharmacology , Retinal Degeneration/chemically induced , Retinal Degeneration/diagnosis , Retinal Neovascularization/genetics , Retinal Neovascularization/metabolism , Retinal Vessels/drug effects , Signal Transduction , Suppressor of Cytokine Signaling 3 Protein/biosynthesis
18.
PLoS One ; 10(5): e0126859, 2015.
Article in English | MEDLINE | ID: mdl-25978399

ABSTRACT

BACKGROUND: Central retinal vein occlusion (CRVO) is a common disease characterized by a disrupted retinal blood supply and a high risk of subsequent vision loss due to retinal edema and neovascular disease. This study was designed to assess the concentrations of selected signaling proteins in the vitreous and blood of patients with ischemic CRVO. METHODS: Vitreous and blood samples were collected from patients undergoing surgery for ischemic CRVO (radial optic neurotomy (RON), n = 13), epiretinal gliosis or macular hole (control group, n = 13). Concentrations of 40 different proteins were determined by an ELISA-type antibody microarray. RESULTS: Expression of proteins enriched in the vitreous (CCL2, IGFBP2, MMP10, HGF, TNFRSF11B (OPG)) was localized by immunohistochemistry in eyes of patients with severe ischemic CRVO followed by secondary glaucoma. Vitreal expression levels were higher in CRVO patients than in the control group (CRVO / control; p < 0.05) for ADIPOQ (13.6), ANGPT2 (20.5), CCL2 (MCP1) (3.2), HGF (4.7), IFNG (13.9), IGFBP1 (14.7), IGFBP2 (1.8), IGFBP3 (4.1), IGFBP4 (1.7), IL6 (10.8), LEP (3.4), MMP3 (4.3), MMP9 (3.6), MMP10 (5.4), PPBP (CXCL7 or NAP2) (11.8), TIMP4 (3.8), and VEGFA (85.3). In CRVO patients, vitreal levels of CCL2 (4.2), HGF (23.3), IGFBP2 (1.23), MMP10 (2.47), TNFRSF11B (2.96), and VEGFA (29.2) were higher than the blood levels (vitreous / blood, p < 0.05). Expression of CCL2, IGFBP2, MMP10, HGF, and TNFRSF11B was preferentially localized to the retina and the retinal pigment epithelium (RPE). CONCLUSION: Proteins related to hypoxia, angiogenesis, and inflammation were significantly elevated in the vitreous of CRVO patients. Moreover, some markers known to indicate atherosclerosis may be related to a basic vascular disease underlying RVO. This would imply that local therapeutic targeting might not be sufficient for a long term therapy in a systemic disease but hypothetically reduce local changes as an initial therapeutic approach.


Subject(s)
Retinal Vein Occlusion/immunology , Retinal Vein Occlusion/metabolism , Vitreous Body/metabolism , Aged , Aged, 80 and over , Chemokine CCL2/metabolism , Female , Humans , Immunohistochemistry , Insulin-Like Growth Factor Binding Protein 2/metabolism , Male , Matrix Metalloproteinase 10/metabolism , Osteoprotegerin/metabolism , Vitreous Body/immunology
19.
Mol Cell Neurosci ; 56: 225-33, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23756201

ABSTRACT

The mouse model of oxygen-induced retinopathy (OIR) is commonly used to investigate various aspects of the pathogenesis of the retinopathy of prematurity (ROP) as well as angiogenesis in general. Retinal astrocytes were suggested to be involved in retinal angiogenesis. This study aimed to describe their localization and cell density during the course of physiological vascularization and pathological revascularization. Mice expressing H2B-GFP (green fluorescent protein fused to histone 2B) from the endogenous Pdgfra promoter were kept in 75% oxygen from P7 (post natal day 7) to P12 (mouse model of OIR). Retinal flatmounts or cryosections were immunostained for glial fibrillary acidic protein (Gfap), glutamine synthetase (Glul), collagen IV (Col IV), desmin (Des), caspase 3 (Casp3), paired box 2 (Pax2), or Ki67. Astrocytic nuclei were counted with the ImageJ macro AuTOCellQuant. The hypoxic state of the retina was investigated by Hypoxyprobe. The GFP signal of the Pdgfra reporter mice co-localized with Pax2, a nuclear marker for retinal astrocytes. This bright label was much easier to quantify than Gfap or Pax2 staining. Quantification of the cell density of astrocytes during physiological development specified the spreading of astrocytes in a concentrical wave from the optic nerve head towards the periphery. Astrocyte density was reduced during the remodelling of the primary vascular plexus into a hierarchical vascular tree (maximal astrocyte density at P1: 2800 astrocytes/mm2, final astrocyte density: 800 astrocytes/mm2). In the OIR model, cell density of astrocytes was elevated in the peripheral vascularized zone. In contrast, astrocyte density dropped to a half (400 astrocytes/mm2) of the normal value in the central avascular zone during the hyperoxic phase between P8 and P10 by apoptosis and rose only after P17 as the retinal network normalized. An additional drop of astrocyte density was observed within the angles between the large vessels of the central avascular zone during hypoxia between P12 and P17. Astrocyte density was not altered at vascular tufts. The hyperoxia effect on astrocytes including the reduced astrocyte density is not the reason for vascular tuft formation. Hypoxia-affected astrocytes in combination with a reduced astrocytic network in the central avascular zone during the hypoxic phase are important determinants in the formation of pathological features during retinal revascularization.


Subject(s)
Astrocytes/metabolism , Hyperoxia/complications , Neovascularization, Pathologic , Retina/pathology , Retinopathy of Prematurity/pathology , Animals , Astrocytes/pathology , Caspase 3/genetics , Caspase 3/metabolism , Collagen Type IV/genetics , Collagen Type IV/metabolism , Desmin/genetics , Desmin/metabolism , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Glutamate-Ammonia Ligase/genetics , Glutamate-Ammonia Ligase/metabolism , Histones/genetics , Histones/metabolism , Hyperoxia/metabolism , Hyperoxia/pathology , Ki-67 Antigen/genetics , Ki-67 Antigen/metabolism , Mice , Mice, Inbred C57BL , Neovascularization, Physiologic , Oxygen/toxicity , PAX2 Transcription Factor/genetics , PAX2 Transcription Factor/metabolism , Retina/metabolism , Retinopathy of Prematurity/etiology , Retinopathy of Prematurity/metabolism
20.
FEBS Lett ; 587(11): 1650-5, 2013 Jun 05.
Article in English | MEDLINE | ID: mdl-23603393

ABSTRACT

Semaphorins are known modulators of axonal sprouting and angiogenesis. In the retina, we identified a distinct and almost exclusive expression of Semaphorin 3F in the outer layers. Interestingly, these outer retinal layers are physiologically avascular. Using functional in vitro models, we report potent anti-angiogenic effects of Semaphorin 3F on both retinal and choroidal vessels. In addition, human retinal pigment epithelium isolates from patients with pathologic neovascularization of the outer retina displayed reduced Semaphorin 3F expression in 10 out of 15 patients. Combined, these results elucidate a functional role for Semaphorin 3F in the outer retina where it acts as a vasorepulsive cue to maintain physiologic avascularity.


Subject(s)
Angiostatic Proteins/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Retinal Photoreceptor Cell Outer Segment/metabolism , Animals , Cells, Cultured , Humans , Mice , Mice, Inbred C57BL , Retinal Pigment Epithelium/metabolism , Retinal Vessels/metabolism , Spheroids, Cellular , Vascular Endothelial Growth Factor A/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...