Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.189
Filter
1.
Exp Eye Res ; 244: 109930, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38750782

ABSTRACT

Corneal neovascularization (CoNV) is a vision-threatening ocular disease commonly secondary to infectious, inflammatory, and traumatic etiologies. Slit lamp photography, in vivo confocal microscopy, angiography, and optical coherence tomography angiography (OCTA) are the primary diagnostic tools utilized in clinical practice to evaluate the vasculature of the ocular surface. However, there is currently a dearth of comprehensive literature that reviews the advancements in imaging technology for CoNV administration. Initially designed for retinal vascular imaging, OCTA has now been expanded to the anterior segment and has shown promising potential for imaging the conjunctiva, cornea, and iris. This expansion allows for the quantitative monitoring of the structural and functional changes associated with CoNV. In this review, we emphasize the impact of algorithm optimization in anterior segment-optical coherence tomography angiography (AS-OCTA) on the diagnostic efficacy of CoNV. Through the analysis of existing literature, animal model assessments are further reported to investigate its pathological mechanism and exhibit remarkable therapeutic interventions. In conclusion, AS-OCTA holds broad prospects and extensive potential for clinical diagnostics and research applications in CoNV.


Subject(s)
Corneal Neovascularization , Fluorescein Angiography , Tomography, Optical Coherence , Corneal Neovascularization/diagnosis , Humans , Tomography, Optical Coherence/methods , Animals , Fluorescein Angiography/methods , Cornea/blood supply , Cornea/pathology , Cornea/diagnostic imaging , Microscopy, Confocal
2.
BMC Med Imaging ; 22(1): 17, 2022 02 03.
Article in English | MEDLINE | ID: mdl-35114961

ABSTRACT

BACKGROUND: Herpes simplex virus (HSV) keratitis remains a leading infectious cause of blindness worldwide. Although all forms of HSV keratitis are commonly recurrent, the risk is greatest in stromal keratitis, which is the most likely to result in corneal scarring, thinning, and neovascularization. Recent studies showed the ability of Optical Coherence Tomography Angiography (OCTA) to detect and study vascular abnormalities in the anterior segment, including abnormal corneal vessels. This study intends to investigate the potential of OCTA device to image and describe quantitatively the vascularization in eyes diagnosed with herpetic leucoma and to discuss and review the usefulness of this technique in this pathology. METHODS: A Cross-sectional study was made, including 17 eyes of 15 patients with leucoma secondary to herpetic keratitis. All eyes underwent anterior segment Slit-Lamp photography (SLP), and OCTA with en-face, b-scans and c-scans imaging. The vessel density (VD) was analyzed in the inferior, nasal and temporal corneal margin in all patients, and in the central area, in eyes with central corneal neovascularization (CoNV). The measurements were calculated after binarization with ImageJ software, using OCTA scans with 6 × 6 mm in a depth of 800 µm. RESULTS: Patients included had a mean age 53.267 ± 21.542 (years ± SD). The mean total vessel area was 50.907% ± 3.435%. VD was higher in the nasal quadrant (51.156% ± 4.276%) but there were no significant differences between the three analyzed areas (p = 0.940). OCTA was able to identify abnormal vessels when SLP apparently showed no abnormal vessels; OCTA was able to distinguish between larger and smaller vessels even in central cornea; OCTA scans allowed the investigation of several corneal planes and the relation of them with clinical findings. CONCLUSIONS: OCTA can be useful in both qualitative and quantitative follow-up of patients and may become a non-invasive alternative to objectively monitor treatment response in eyes with corneal vascularization due to herpetic infection.


Subject(s)
Corneal Opacity/diagnostic imaging , Corneal Opacity/virology , Keratitis, Herpetic/complications , Tomography, Optical Coherence/methods , Adolescent , Adult , Aged , Aged, 80 and over , Cornea/blood supply , Corneal Neovascularization/diagnostic imaging , Corneal Opacity/pathology , Cross-Sectional Studies , Female , Humans , Image Processing, Computer-Assisted/methods , Keratitis, Herpetic/diagnostic imaging , Keratitis, Herpetic/pathology , Male , Middle Aged , Young Adult
3.
Microvasc Res ; 138: 104233, 2021 11.
Article in English | MEDLINE | ID: mdl-34411571

ABSTRACT

OBJECTIVES: Vascular endothelial growth factor A (VEGFA) is one of the major factors initiating and regulating angiogenesis. LncRNA taurine up-regulated gene 1 (TUG1) has been implicated in the pathological neovascularization. The aim of this study is to explore the function of TUG1 in regulating VEGFA-mediated angiogenesis in endothelial cells. METHODS: A total of 12 corneal neovascularization (CRNV) samples were collected form patient undergoing corneal transplantation at Tongji Hospital, Wuhan, China. qRT-PCR and Western blotting were performed to examine gene expression and protein levels. Human umbilical vein endothelial cells (HUVECs) were used as an in vitro angiogenesis model. CCK-8 proliferation assay was used to determine cell proliferation capacity and wound healing was performed to analyze cell migration ability. Dual luciferase reporter assay was used for functional interaction validation between miR-505-3p and its targets. The in vitro angiogenic potential was evaluated by tube formation assay. RESULTS: TUG1 and VEGFA were upregulated in CRNV tissues and VEGFA-treated HUVECs. TUG1 knockdown inhibited proliferation, migration and tube formation capacity of HUVECs. TUG1 regulated the angiogenesis of HUVECs by modulating VEGFA expression through targeting miR-505-3p. CONCLUSIONS: Our results suggest that lncRNA TUG1 promotes the angiogenesis of HUVECs through modulating miR-505-3p/VEGFA axis.


Subject(s)
Cornea/blood supply , Corneal Neovascularization/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , MicroRNAs/metabolism , Neovascularization, Physiologic , RNA, Long Noncoding/metabolism , Vascular Endothelial Growth Factor A/metabolism , Case-Control Studies , Cell Movement , Cell Proliferation , Cells, Cultured , Corneal Neovascularization/genetics , Corneal Neovascularization/pathology , Gene Expression Regulation , Human Umbilical Vein Endothelial Cells/pathology , Humans , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Signal Transduction , Vascular Endothelial Growth Factor A/genetics
4.
Int J Mol Sci ; 22(14)2021 Jul 08.
Article in English | MEDLINE | ID: mdl-34298979

ABSTRACT

Platelet extravasation during inflammation is under-appreciated. In wild-type (WT) mice, a central corneal epithelial abrasion initiates neutrophil (PMN) and platelet extravasation from peripheral limbal venules. The same injury in mice expressing low levels of the ß2-integrin, CD18 (CD18hypo mice) shows reduced platelet extravasation with PMN extravasation apparently unaffected. To better define the role of CD18 on platelet extravasation, we focused on two relevant cell types expressing CD18: PMNs and mast cells. Following corneal abrasion in WT mice, we observed not only extravasated PMNs and platelets but also extravasated erythrocytes (RBCs). Ultrastructural observations of engorged limbal venules showed platelets and RBCs passing through endothelial pores. In contrast, injured CD18hypo mice showed significantly less venule engorgement and markedly reduced platelet and RBC extravasation; mast cell degranulation was also reduced compared to WT mice. Corneal abrasion in mast cell-deficient (KitW-sh/W-sh) mice showed less venule engorgement, delayed PMN extravasation, reduced platelet and RBC extravasation and delayed wound healing compared to WT mice. Finally, antibody-induced depletion of circulating PMNs prior to corneal abrasion reduced mast cell degranulation, venule engorgement, and extravasation of PMNs, platelets, and RBCs. In summary, in the injured cornea, platelet and RBC extravasation depends on CD18, PMNs, and mast cell degranulation.


Subject(s)
Blood Platelets/physiology , CD18 Antigens/physiology , Cell Degranulation , Cornea/blood supply , Erythrocytes/physiology , Hyperemia/physiopathology , Mast Cells/physiology , Neutrophils/physiology , Transendothelial and Transepithelial Migration/physiology , Vasculitis/immunology , Venules/metabolism , Animals , CD18 Antigens/deficiency , Cell Movement , Chemotaxis, Leukocyte , Corneal Injuries/metabolism , Corneal Injuries/pathology , Epithelium, Corneal/physiology , Female , Hyperemia/blood , Macrophages/physiology , Male , Mice , Mice, Inbred C57BL , Microcirculation , Microscopy, Electron , Models, Animal , Phagocytosis , Regeneration/physiology , Vasculitis/blood , Venules/pathology , Wound Healing/physiology
5.
Int J Mol Sci ; 22(12)2021 Jun 08.
Article in English | MEDLINE | ID: mdl-34201218

ABSTRACT

Ocular graft-versus-host disease (oGVHD) is a fast progressing, autoimmunological disease following hematopoietic stem cell transplantation, leading to severe inflammation of the eye and destruction of the lacrimal functional unit with consecutive sight-threatening consequences. The therapeutic "window of opportunity" is narrow, and current treatment options are limited and often insufficient. To achieve new insights into the pathogenesis and to develop new therapeutic approaches, clinically relevant models of oGVHD are desirable. In this study, the ocular phenotype was described in a murine, chemotherapy-based, minor-mismatch GVHD model mimicking early-onset chronic oGVHD, with corneal epitheliopathy, inflammation of the lacrimal glands, and blepharitis. Additionally, corneal lymphangiogenesis was observed as part of oGVHD pathogenesis for the first time, thus opening up the investigation of lymphangiogenesis as a potential therapeutic and diagnostic tool.


Subject(s)
Antineoplastic Agents/toxicity , Blepharitis/pathology , Cornea/blood supply , Graft vs Host Disease/pathology , Hematopoietic Stem Cell Transplantation/adverse effects , Inflammation/pathology , Lacrimal Apparatus/pathology , Animals , Blepharitis/etiology , Blepharitis/metabolism , Disease Models, Animal , Female , Graft vs Host Disease/etiology , Graft vs Host Disease/metabolism , Inflammation/etiology , Inflammation/metabolism , Lacrimal Apparatus/metabolism , Lymphangiogenesis , Mice , Mice, Inbred C57BL
6.
Rom J Ophthalmol ; 65(1): 64-69, 2021.
Article in English | MEDLINE | ID: mdl-33817436

ABSTRACT

Aim: The purpose of this experimental study was to evaluate the existence of adrenergic receptors in ketamine-induced corneal blood vessels in rat pups. Methods: The study of corneal neovascularization motricity was performed on 45-day-old Wistar rats in which, starting from the 15th day of life, corneal blood vessels were obtained by injecting intraperitoneal ketamine at a dose of 150 mg/ kg body weight, a total of 5 successive doses. The examination of the neovascularization was done with the help of a Nikon stereomicroscope connected to a video camera and a computer, the total magnification being 400X. The reactivity of the new corneal blood vessels to the administration in conjunctival instillations of a 1.5 mmol/L adrenaline solution was tested. The parameters followed were represented by variations in the caliber of corneal blood vessels. The data were analyzed using Microsoft Office Excel. Results: Administration of distilled water did not produce statistically significant changes in corneal blood vessels, while adrenaline produced a statistically significant constriction of vascular diameter (p=0.01 at T9, p=0.004 at T10, p=0.019 at time T11 of examinations). Conclusions: The results showed that adrenaline produces vasoconstriction in the new corneal blood vessels, which allows us to assume that they contain α-adrenergic receptors. However, we cannot say that corneal pathological vessels do not contain ß2-type adrenergic receptors, because the effect of adrenaline may be an algebraic sum between vasoconstriction produced by stimulating α-adrenergic receptors and vasodilation produced by stimulating ß2-adrenergic receptors, but in which the vasodilating effect may be masked by the vasoconstrictor effect given by a higher density of α-adrenergic receptors. Abbreviations: A= adrenaline, DNM = non-measurable diameter, NA= noradrenaline, Std.Er.= Standard error.


Subject(s)
Cornea/blood supply , Epinephrine/pharmacology , Receptors, Adrenergic/metabolism , Vasoconstriction/drug effects , Animals , Cornea/drug effects , Cornea/metabolism , Models, Animal , Rats , Rats, Wistar , Vasoconstrictor Agents/pharmacology
7.
Front Immunol ; 12: 667830, 2021.
Article in English | MEDLINE | ID: mdl-33897716

ABSTRACT

Macrophages are critical mediators of tissue vascularization both in health and disease. In multiple tissues, macrophages have been identified as important regulators of both blood and lymphatic vessel growth, specifically following tissue injury and in pathological inflammatory responses. In development, macrophages have also been implicated in limiting vascular growth. Hence, macrophages provide an important therapeutic target to modulate tissue vascularization in the clinic. However, the molecular mechanisms how macrophages mediate tissue vascularization are still not entirely resolved. Furthermore, mechanisms might also vary among different tissues. Here we review the role of macrophages in tissue vascularization with a focus on their role in blood and lymphatic vessel formation in the barrier tissues cornea and skin. Comparing mechanisms of macrophage-mediated hem- and lymphangiogenesis in the angiogenically privileged cornea and the physiologically vascularized skin provides an opportunity to highlight similarities but also tissue-specific differences, and to understand how macrophage-mediated hem- and lymphangiogenesis can be exploited for the treatment of disease, including corneal wound healing after injury, graft rejection after corneal transplantation or pathological vascularization of the skin.


Subject(s)
Blood Vessels/metabolism , Cornea/blood supply , Corneal Neovascularization , Lymphangiogenesis , Lymphatic Vessels/metabolism , Macrophages/metabolism , Neovascularization, Physiologic , Skin/blood supply , Animals , Blood Vessels/immunology , Blood Vessels/pathology , Humans , Lymphatic Vessels/immunology , Lymphatic Vessels/pathology , Macrophages/immunology , Macrophages/pathology , Phenotype , Signal Transduction , Wound Healing
9.
Animal Model Exp Med ; 4(4): 300-310, 2021 12.
Article in English | MEDLINE | ID: mdl-34977481

ABSTRACT

The cornea is an avascular, transparent tissue that is essential for visual function. Any disturbance to the corneal transparency will result in a severe vision loss. Due to the avascular nature, the cornea acquires most of the oxygen supply directly or indirectly from the atmosphere. Corneal tissue hypoxia has been noticed to influence the structure and function of the cornea for decades. The etiology of hypoxia of the cornea is distinct from the rest of the body, mainly due to the separation of cornea from the atmosphere, such as prolonged contact lens wearing or closed eyes. Corneal hypoxia can also be found in corneal inflammation and injury when a higher oxygen requirement exceeds the oxygen supply. Systemic hypoxic state during lung diseases or high altitude also leads to corneal hypoxia when a second oxygen consumption route from aqueous humor gets blocked. Hypoxia affects the cornea in multiple aspects, including disturbance of the epithelium barrier function, corneal edema due to endothelial dysfunction and metabolism changes in the stroma, and thinning of corneal stroma. Cornea has also evolved mechanisms to adapt to the hypoxic state initiated by the activation of hypoxia inducible factor (HIF). The aim of this review is to introduce the pathology of cornea under hypoxia and the mechanism of hypoxia adaptation, to discuss the current animal models used in this field, and future research directions.


Subject(s)
Corneal Edema , Lung Diseases , Animals , Cornea/blood supply , Corneal Edema/complications , Hypoxia/etiology , Lung Diseases/complications , Models, Animal
10.
Clin Transl Sci ; 14(3): 829-836, 2021 05.
Article in English | MEDLINE | ID: mdl-33202098

ABSTRACT

The purpose of this study was to investigate the influence of smartphone reading on the ocular surface and to compare the various effects of different screens and light conditions on the ocular surface. One hundred nineteen volunteers were randomly divided into: light + organic light-emitting diode (OLED), light + electronic ink (eINK), dark + OLED, and dark + eINK. Ocular surface examinations, including noninvasive break-up time (NIBUT), noninvasive keratograph tear meniscus height (NIKTMH), ocular redness, fluorescein break-up time (FBUT), corneal fluorescein staining, meibomian gland assessment, Schirmer I Test, and blinking frequency, were performed before and after a reading task. Symptoms were evaluated using the Ocular Surface Disease Index (OSDI) and Computer Vision Syndrome Questionnaire (CVS-Q). NIBUT and FBUT were decreased statistically significantly after participants read on an OLED screen for 2 hours compared with the baseline in light and dark environments, whereas no statistically significant decrease was observed on an eINK screen. NIKTMH was statistically significantly decreased after reading on an OLED screen in light and dark settings, and the eINK screen had a lesser effect on NIKTMH. An obvious increase in the ocular redness, OSDI and CVS-Q scores was observed after reading on an OLED screen, whereas the eINK screen had a lesser effect on these indicators. Blink rate increased gradually in OLED subgroups during the reading task, whereas no statistically significant difference was observed in the eINK subgroups. Our research suggested that reading on an OLED screen can cause ocular surface disorder and obvious subjective discomfort, whereas reading on an eINK screen can minimize ocular surface disorder in both dark and light environments.


Subject(s)
Cornea/radiation effects , Dry Eye Syndromes/etiology , Light/adverse effects , Reading , Smartphone , Adult , Blinking/radiation effects , Cornea/blood supply , Cornea/diagnostic imaging , Dry Eye Syndromes/prevention & control , Female , Healthy Volunteers , Humans , Male , Prospective Studies , Semiconductors/adverse effects , Young Adult
11.
Sci Rep ; 10(1): 22249, 2020 12 17.
Article in English | MEDLINE | ID: mdl-33335224

ABSTRACT

The aim was to assess the relationships between cardiovascular activity, corneal pulse characteristics, and corneal biomechanics in rabbits. Seventeen rabbits were randomly assigned to one of two anesthetic regimens to induce differences in arterial blood pressure and heart rate. Experimental protocol included measuring blood flow parameters in the ophthalmic artery by color Doppler imaging, corneal biomechanical parameters using a non-contact tonometer Corvis ST, and the corneal pulse (CP) signal using a non-contact ultrasonic technique. Statistically significantly lower mean values of normalized amplitudes of higher CP harmonics and changes in eight of the twelve corneal biomechanical parameters were observed in the rabbit group with lower arterial blood pressure and higher heart rate, intraocular pressure, and resistive index. The results of partial correlations showed that the CP signal energy and amplitude of its first harmonic correlate with the resistive index, diastolic and mean arterial pressures, whereas no statistically significant correlation was found between any of the CP parameters and intraocular pressure. Our pilot study indicates, for the first time, that non-contact and continuous measuring of corneal pulse allows indirectly assessing changes in cardiovascular activity when the confounding effect of intraocular pressure is eliminated.


Subject(s)
Biomechanical Phenomena , Cardiovascular Physiological Phenomena , Cardiovascular System/physiopathology , Cornea/physiology , Animals , Blood Circulation , Blood Pressure , Cornea/blood supply , Cornea/diagnostic imaging , Intraocular Pressure , Models, Biological , Rabbits , Tonometry, Ocular , Ultrasonography, Doppler, Color
12.
Oxid Med Cell Longev ; 2020: 7435260, 2020.
Article in English | MEDLINE | ID: mdl-32655773

ABSTRACT

Corneal alkali burns are potentially blinding injuries. Alkali induces oxidative stress in corneas followed by excessive corneal inflammation, neovascularization, and untransparent scar formation. Molecular hydrogen (H2), a potent reactive oxygen species (ROS) scavenger, suppresses oxidative stress and enables corneal healing when applied on the corneal surface. The purpose of this study was to examine whether the H2 pretreatment of healthy corneas evokes a protective effect against corneal alkali-induced oxidative stress. Rabbit eyes were pretreated with a H2 solution or buffer solution, by drops onto the ocular surface, and the corneas were then burned with 0.25 M NaOH. The results obtained with immunohistochemistry and pachymetry showed that in the corneas of H2-pretreated eyes, slight oxidative stress appeared followed by an increased expression of antioxidant enzymes. When these corneas were postburned with alkali, the alkali-induced oxidative stress was suppressed. This was in contrast to postburned buffer-pretreated corneas, where the oxidative stress was strong. These corneas healed with scar formation and neovascularization, whereas corneas of H2-pretreated eyes healed with restoration of transparency in the majority of cases. Corneal neovascularization was strongly suppressed. Our results suggest that the corneal alkali-induced oxidative stress was reduced via the increased antioxidant capacity of corneal cells against reactive oxygen species (ROS). It is further suggested that the ability of H2 to induce the increase in antioxidant cell capacity is important for eye protection against various diseases or external influences associated with ROS production.


Subject(s)
Alkalies/toxicity , Antioxidants/metabolism , Burns, Chemical/drug therapy , Cornea/metabolism , Eye Burns/drug therapy , Hydrogen/therapeutic use , Oxidative Stress/drug effects , Animals , Burns, Chemical/metabolism , Burns, Chemical/pathology , Cornea/blood supply , Cornea/drug effects , Cornea/pathology , Corneal Neovascularization/prevention & control , Disease Models, Animal , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Eye Burns/chemically induced , Eye Burns/metabolism , Eye Burns/pathology , Female , Hydrogen/pharmacology , Rabbits , Reactive Oxygen Species/metabolism , Wound Healing/drug effects
13.
Biomed Pharmacother ; 130: 110391, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32674015

ABSTRACT

PURPOSE: To evaluate the safety and potential healing efficacy of the topical ocular administration of a gelatin membrane containing usnic acid/liposomes (UALs) for corneal cicatrization. UALs have shown healing activity in animal models of dermal burn lesions. We evaluated the safety of topical ocular administration of UAL and its potential healing efficacy as an ophthalmic treatment on chemical lesions in rabbit eyes. METHOD: The Draize test was used to check for ocular toxicity and the score was zero at each observation, indicating the ocular safety of a gelatin membrane containing usnic acid/liposome. Its potential healing efficacy as an ophthalmic treatment on chemical lesions in rabbit eyes was also assessed. RESULTS: After epithelial removal and treatment with UAL, there was a 49.4 % reduction in injury under in vivo conditions compared with a 36.6 % reduction in the control, a gelatin membrane containing liposome without usnic acid. Histological analysis of ocular surface chemical injury-tissue sections after treatment with UAL supported these observations. The corneal expression of VEGF and TGF-ß1increased by 70 % and 50 % respectively following treatment with UAL gelatin membrane. CONCLUSION: These results indicate the potential therapeutic application of UAL gelatin membranes as an ophthalmic treatment that may be used for corneal cicatrization.


Subject(s)
Benzofurans/administration & dosage , Cicatrix/drug therapy , Cornea/drug effects , Drug Delivery Systems , Wound Healing/drug effects , Administration, Ophthalmic , Animals , Benzofurans/chemistry , Chickens , Cornea/blood supply , Female , Gelatin/administration & dosage , Gelatin/chemistry , Liposomes/administration & dosage , Liposomes/chemistry , Neovascularization, Physiologic/drug effects , Ophthalmic Solutions/chemistry
14.
Biomed Pharmacother ; 128: 110248, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32454287

ABSTRACT

Selective inhibition of vascular endothelial growth factor receptor (VEGFR), particularly VEGFR-2, is an efficient method for the treatment of ocular neovascularization. SU1498 is a specific inhibitor of VEGFR-2. In this study, we investigated the role of SU1498 in ocular neovascularization. Administration of SU1498 did not show any cytotoxicity and tissue toxicity at the tested concentrations. Administration of SU1498 reduced the size and thickness of choroidal neovascularization and decreased the mean length and mean number of corneal neovascular vessels induced by alkali burn. Pretreatment of SU1498 significantly reduced the proliferation, migration, and tube formation ability of HUVECs. SU1498 played the anti-angiogenic role through the regulation of p38-MAPK signaling. Taken together, inhibition of VEGFR-2 by SU1498 provides a novel therapeutic approach for ocular neovascularization.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Choroid/blood supply , Choroidal Neovascularization/prevention & control , Cinnamates/pharmacology , Cornea/blood supply , Corneal Neovascularization/prevention & control , Endothelial Cells/drug effects , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Animals , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Choroidal Neovascularization/metabolism , Choroidal Neovascularization/pathology , Corneal Neovascularization/metabolism , Corneal Neovascularization/pathology , Disease Models, Animal , Endothelial Cells/metabolism , Endothelial Cells/pathology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Male , Mice, Inbred C57BL , Neovascularization, Physiologic/drug effects , Signal Transduction , Vascular Endothelial Growth Factor Receptor-2/metabolism
15.
Angiogenesis ; 23(3): 459-477, 2020 08.
Article in English | MEDLINE | ID: mdl-32372335

ABSTRACT

Blood vessels and nerve tissues are critical to the development and functionality of many vital organs. However, little is currently known about their interdependency during development and after injury. In this study, dual fluorescence transgenic reporter mice were utilized to observe blood vessels and nervous tissues in organs postnatally. Thy1-YFP and Flt1-DsRed (TYFD) mice were interbred to achieve dual fluorescence in the offspring, with Thy1-YFP yellow fluorescence expressed primarily in nerves, and Flt1-DsRed fluorescence expressed selectively in blood vessels. Using this dual fluorescent mouse strain, we were able to visualize the networks of nervous and vascular tissue simultaneously in various organ systems both in the physiological state and after injury. Using ex vivo high-resolution imaging in this dual fluorescent strain, we characterized the organizational patterns of both nervous and vascular systems in a diverse set of organs and tissues. In the cornea, we also observed the dynamic patterns of nerve and blood vessel networks following epithelial debridement injury. These findings highlight the versatility of this dual fluorescent strain for characterizing the relationship between nerve and blood vessel growth and organization.


Subject(s)
Blood Vessels , Cornea , Isoantibodies , Luminescent Proteins , Optical Imaging , Peripheral Nerves , Vascular Endothelial Growth Factor Receptor-1 , Animals , Blood Vessels/diagnostic imaging , Blood Vessels/growth & development , Cornea/blood supply , Cornea/diagnostic imaging , Cornea/innervation , Female , Isoantibodies/biosynthesis , Isoantibodies/genetics , Luminescent Proteins/biosynthesis , Luminescent Proteins/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Transgenic , Peripheral Nerves/diagnostic imaging , Peripheral Nerves/growth & development , Vascular Endothelial Growth Factor Receptor-1/biosynthesis , Vascular Endothelial Growth Factor Receptor-1/genetics
16.
Cutan Ocul Toxicol ; 39(3): 223-228, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32338080

ABSTRACT

PURPOSE: The purpose of this study was to compare the neovascularization inhibiting the effect of topical bevacizumab and sorafenib and to determine the effective dose of sorafenib. MATERIAL AND METHODS: Forty-two healthy Wistar albino rats were randomly divided into six groups. The right corneas of all rats except group 1 were cauterised with silver nitrate. Group 2 received DMSO, group 3 received topical bevacizumab (5 mg/dL, 3 times a day) and group 4, 5 and 6 received topical sorafenib (2.5 mg/dl, 5 mg/dL, 7.5 mg/dL, 2 times a day respectively), between days 1 and 7. Corneal photographs were taken on day 8 and the corneal neovascular area percentage was calculated. Following decapitation, the corneas were removed to determine the levels of VEGF ELISA and corneal immune staining. The Mann-Whitney U-test was used for statistical analysis. RESULTS: The neovascular corneal area percentage was statistically significantly lower in the treatment groups than group 2 (p < 0.05). The intensity of VEGF immune staining was also lower in groups 3, 5 and 6 from the group 2. Group 3, 5 and 6 were no significant differences compared to group 1. The VEGF ELISA levels were statistically significantly lower in group 3, 5 and 6 compared to group 2 (p < 0.05). There was no statistically difference between VEGF ELISA levels of group 2 and 4 (p > 0.05). CONCLUSIONS: Sorafenib was as effective as bevacizumab in the regression of corneal neovascularization. The effect of sorafenib seems to be dose-dependent. The low doses and twice a day administration are important advantages of sorafenib.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Bevacizumab/therapeutic use , Corneal Neovascularization/drug therapy , Protein Kinase Inhibitors/therapeutic use , Sorafenib/therapeutic use , Angiogenesis Inhibitors/pharmacology , Animals , Bevacizumab/pharmacology , Cornea/blood supply , Cornea/drug effects , Cornea/metabolism , Corneal Neovascularization/metabolism , Disease Models, Animal , Male , Protein Kinase Inhibitors/pharmacology , Rats, Wistar , Sorafenib/pharmacology , Vascular Endothelial Growth Factor A/metabolism
17.
Sci Rep ; 10(1): 6592, 2020 04 20.
Article in English | MEDLINE | ID: mdl-32313133

ABSTRACT

The aim of the current study is to identify possible new Ocular Response Analyzer (ORA) waveform parameters related to changes of retinal structure/deformation, as measured by the peripapillary retinal arteries angle (PRAA), using a generative deep learning method of variational autoencoder (VAE). Fifty-four eyes of 52 subjects were enrolled. The PRAA was calculated from fundus photographs and was used to train a VAE model. By analyzing the ORA waveform reconstructed (noise filtered) using VAE, a novel ORA waveform parameter (Monot1-2), was introduced, representing the change in monotonicity between the first and second applanation peak of the waveform. The variables mostly related to the PRAA were identified from a set of 41 variables including age, axial length (AL), keratometry, ORA corneal hysteresis, ORA corneal resistant factor, 35 well established ORA waveform parameters, and Monot1-2, using a model selection method based on the second-order bias-corrected Akaike information criterion. The optimal model for PRAA was the AL and six ORA waveform parameters, including Monot1-2. This optimal model was significantly better than the model without Monot1-2 (p = 0.0031, ANOVA). The current study suggested the value of a generative deep learning approach in discovering new useful parameters that may have clinical relevance.


Subject(s)
Cornea/diagnostic imaging , Glaucoma, Open-Angle/diagnosis , Retina/diagnostic imaging , Retinal Artery/diagnostic imaging , Tonometry, Ocular/methods , Adult , Aged , Aged, 80 and over , Cornea/blood supply , Cornea/physiopathology , Diagnostic Techniques, Ophthalmological/trends , Female , Glaucoma, Open-Angle/diagnostic imaging , Glaucoma, Open-Angle/physiopathology , Humans , Intraocular Pressure/physiology , Male , Middle Aged , Retina/physiopathology , Retinal Artery/physiopathology
18.
J Ocul Pharmacol Ther ; 36(4): 238-246, 2020 05.
Article in English | MEDLINE | ID: mdl-32077779

ABSTRACT

Purpose: The objective of this study was to describe the short-term results of allogenic transplantation of limbal stem cells expanded on amniotic membrane for the ocular surface reconstruction. Methods: Prospective nonrandomized, nonmasked study in a single ophthalmological center. Ten patients with bilateral total limbal stem cell deficiency (LSCD) were included. Expression and presence of ABCB5 and Δp63α in amniotic membrane-cultured limbal epithelial stem cells were analyzed, in relationship with clinical changes after allogenic transplantation. An objective evaluation was performed to determine corneal transparency and superficial vascularization. Results: In a median follow-up time of 11.6 months, 7 patients (70%) were considered as failure compared with the preoperative status. ABCB5 and Δp63α are expressed in similar amount in the limbal epithelial cells expanded in vitro and transplanted in patients with bilateral LSCD. Conclusions: Transplantation of allogenic epithelial limbal cells expanded in amniotic membrane could be considered in patients with LSCD due to burns or congenital etiologies such as aniridia, but its benefit is limited for patients with immunologic diseases.


Subject(s)
Amnion/transplantation , Corneal Diseases/etiology , Epithelium, Corneal/transplantation , Limbus Corneae/pathology , Stem Cells/cytology , Transplantation, Homologous/methods , ATP Binding Cassette Transporter, Subfamily B/metabolism , Adolescent , Adult , Amnion/cytology , Amnion/metabolism , Aniridia/complications , Case-Control Studies , Cornea/blood supply , Cornea/metabolism , Corneal Diseases/diagnosis , Corneal Diseases/metabolism , Corneal Diseases/surgery , Corneal Injuries/complications , Epithelium, Corneal/abnormalities , Epithelium, Corneal/cytology , Epithelium, Corneal/metabolism , Female , Follow-Up Studies , Humans , Limbus Corneae/cytology , Limbus Corneae/metabolism , Male , Mexico/epidemiology , Middle Aged , Non-Randomized Controlled Trials as Topic/methods , Prospective Studies , Stem Cell Transplantation/adverse effects , Stem Cells/metabolism , Stem Cells/pathology , Transcription Factors/metabolism , Treatment Outcome , Tumor Suppressor Proteins/metabolism , Young Adult
19.
Biomolecules ; 10(2)2020 01 31.
Article in English | MEDLINE | ID: mdl-32023953

ABSTRACT

Inflammation-induced angiogenesis is closely related to many diseases and has been regarded as a therapeutic target. Caspase-8 has attracted increasing attention for its immune properties and therapeutic potential in inflammatory disorders. The aim of our study is to investigate the clinical application of pharmacological inhibition of caspase-8 and the underlying molecular mechanisms in inflammation-induced angiogenesis in the cornea. A model of alkali burn (AB)-induced corneal neovascularization (CNV) in C57BL/6 wild-type (WT) mice and toll-like receptor 4 knockout (Tlr4-/-) mice was used. We found that AB increased caspase-8 activity and the pharmacological inhibition of caspase-8 exerted substantial inhibitory effects on CNV, with consistent decreases in caspase-8 activity, inflammatory cell infiltration, macrophage recruitment and activation, VEGF-A, TNF-α, IL-1ß, MIP-1, and MCP-1 expression in the cornea. In vitro, caspase-8 mediated TLR4-dependent chemokines and VEGF-A production by macrophages. The TLR4 knockout significantly alleviated CNV, suppressed caspase-8 activity and downregulated expression of inflammatory cytokines and chemokines after AB. Taken together, these findings provide the first demonstration that the pharmacological inhibition of caspase-8 suppresses inflammation-induced angiogenesis and support the use of a pharmacological caspase-8 inhibitor as a novel clinical treatment for CNV and other angiogenic disorders.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Caspase 8/metabolism , Caspase Inhibitors/pharmacology , Cornea/blood supply , Inflammation/drug therapy , Neovascularization, Pathologic/drug therapy , Animals , Caspase 1/metabolism , Caspase 8/genetics , Cornea/pathology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , RAW 264.7 Cells , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Vascular Endothelial Growth Factor A/metabolism
20.
J AAPOS ; 24(1): 51-53, 2020 02.
Article in English | MEDLINE | ID: mdl-31935453

ABSTRACT

An African American girl born at 37 weeks via spontaneous vaginal delivery to a 33-year-old woman was noted on delivery to have a unilateral absent red reflex in the right eye, which was enlarged. Intraocular pressure was elevated, and the cornea had a straw-colored opacity. B-scan ultrasonography of the right eye showed diffuse hyperechoic vitreous opacities and a retrolental mass, with a hyperechoic band stretching from the optic disk to the posterior lens. Neuroimaging showed a unilateral enlarged globe, intraocular hemorrhage, and persistent fetal vasculature, with no other intracranial pathology. An anterior chamber washout revealed liquified blood; the presence of corneal blood staining was confirmed. A spontaneous intraocular hemorrhage associated with persistent fetal vasculature was suspected, leading to secondary glaucoma and corneal blood staining.


Subject(s)
Cornea/blood supply , Corneal Diseases/etiology , Eye Hemorrhage/etiology , Intraocular Pressure/physiology , Persistent Hyperplastic Primary Vitreous/complications , Cornea/diagnostic imaging , Corneal Diseases/diagnosis , Eye Hemorrhage/diagnosis , Female , Humans , Infant, Newborn , Magnetic Resonance Imaging , Persistent Hyperplastic Primary Vitreous/diagnosis , Ultrasonography
SELECTION OF CITATIONS
SEARCH DETAIL
...