Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 431
Filter
1.
Int J Mol Sci ; 25(10)2024 May 17.
Article in English | MEDLINE | ID: mdl-38791518

ABSTRACT

Corneal neovascularization can impair vision and result in a poor quality of life. The pathogenesis involves a complex interplay of angiogenic factors, notably vascular endothelial growth factor (VEGF). This review provides a comprehensive overview of potential therapies for corneal neovascularization, covering tissue inhibitors of metalloproteinases (TIMPs), transforming growth factor beta (TGF-ß) inhibitors, interleukin-1L receptor antagonist (IL-1 Ra), nitric oxide synthase (NOS) isoforms, galectin-3 inhibitors, retinal pigment epithelium-derived factor (PEDF), platelet-derived growth factor (PDGF) receptor inhibitors, and surgical treatments. Conventional treatments include anti-VEGF therapy and laser interventions, while emerging therapies such as immunosuppressive drugs (cyclosporine and rapamycin) have been explored. Losartan and decorin are potential antifibrotic agents that mitigate TGF-ß-induced fibrosis. Ocular nanosystems are innovative drug-delivery platforms that facilitate the targeted release of therapeutic agents. Gene therapies, such as small interfering RNA and antisense oligonucleotides, are promising approaches for selectively inhibiting angiogenesis-related gene expression. Aganirsen is efficacious in reducing the corneal neovascularization area without significant adverse effects. These multifaceted approaches underscore the corneal neovascularization management complexity and highlight ideas for enhancing therapeutic outcomes. Furthermore, the importance of combination therapies and the need for further research to develop specific inhibitors while considering their therapeutic efficacy and potential adverse effects are discussed.


Subject(s)
Corneal Neovascularization , Humans , Corneal Neovascularization/drug therapy , Corneal Neovascularization/therapy , Corneal Neovascularization/metabolism , Animals , Genetic Therapy/methods , Angiogenesis Inhibitors/therapeutic use , Transforming Growth Factor beta/metabolism
2.
Eur J Pharmacol ; 977: 176673, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38815785

ABSTRACT

Corneal neovascularization (CoNV) is predominantly initiated by inflammatory processes, resulting in aberrant vascular proliferation and consequent visual impairment. Existing therapeutic interventions for CoNV demonstrate limited efficacy and potential for adverse reactions. Protein arginine methyltransferase 1 (PRMT1) is associated with the regulation of inflammation and M2 macrophage polarization. Nevertheless, the precise mechanism by which PRMT1 operates in CoNV remains uncertain. This study explored the impact of PRMT1 inhibition in a murine model of CoNV induced by alkali burn. Our findings indicated a direct relationship between PRMT1 levels and corneal damage. Moreover, our observations indicated an increase in fibroblast growth factor 2 (FGF2) expression in CoNV, which was reduced after treatment with a PRMT1 inhibitor. The inhibition of PRMT1 alleviated both corneal injury and CoNV, as evidenced by decreased corneal opacity and neovascularization. Immunofluorescence analysis and evaluation of inflammatory factor expression demonstrated that PRMT1 inhibition attenuated M2 macrophage polarization, a phenomenon that was reversed by the administration of recombinant FGF2 protein. These results were confirmed through experimentation on Human Umbilical Vein Endothelial Cells (HUVECs) and Mouse leukemia cells of monocyte macrophage cells (RAW264.7). Furthermore, it was established that FGF2 played a role in PI3K/Akt signal transduction, a critical regulatory pathway for M2 macrophage polarization. Importantly, the activity of this pathway was found to be suppressed by PRMT1 inhibitors. Mechanistically, PRMT1 was shown to promote M2 macrophage polarization, thereby contributing to CoNV, through the FGF2/PI3K/Akt pathway. Therefore, targeting PRMT1 may offer a promising therapeutic approach.


Subject(s)
Corneal Neovascularization , Fibroblast Growth Factor 2 , Human Umbilical Vein Endothelial Cells , Macrophages , Phosphatidylinositol 3-Kinases , Protein-Arginine N-Methyltransferases , Proto-Oncogene Proteins c-akt , Signal Transduction , Protein-Arginine N-Methyltransferases/metabolism , Protein-Arginine N-Methyltransferases/antagonists & inhibitors , Protein-Arginine N-Methyltransferases/genetics , Animals , Fibroblast Growth Factor 2/metabolism , Mice , Macrophages/drug effects , Macrophages/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Humans , Corneal Neovascularization/pathology , Corneal Neovascularization/metabolism , Corneal Neovascularization/prevention & control , RAW 264.7 Cells , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Male , Mice, Inbred C57BL , Disease Models, Animal , Repressor Proteins
3.
Adv Sci (Weinh) ; 11(25): e2401710, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38582513

ABSTRACT

Corneal neovascularization (CNV) is a common clinical finding seen in a range of eye diseases. Current therapeutic approaches to treat corneal angiogenesis, in which vascular endothelial growth factor (VEGF) A plays a central role, can cause a variety of adverse side effects. The technology of Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/Cas9 can edit VEGFA gene to suppress its expression. CRISPR offers a novel opportunity to treat CNV. This study shows that depletion of VEGFA with a novel CRISPR/Cas9 system inhibits proliferation, migration, and tube formation of human umbilical vein endothelial cells (HUVECs) in vitro. Importantly, subconjunctival injection of this dual AAV-SpCas9/sgRNA-VEGFA system is demonstrated which blocks suture-induced expression of VEGFA, CD31, and α-smooth muscle actin as well as corneal neovascularization in mice. This study has established a strong foundation for the treatment of corneal neovascularization via a gene editing approach for the first time.


Subject(s)
CRISPR-Cas Systems , Corneal Neovascularization , Disease Models, Animal , Gene Editing , Human Umbilical Vein Endothelial Cells , Vascular Endothelial Growth Factor A , Corneal Neovascularization/genetics , Corneal Neovascularization/therapy , Corneal Neovascularization/metabolism , Animals , Gene Editing/methods , Mice , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Humans , Human Umbilical Vein Endothelial Cells/metabolism , CRISPR-Cas Systems/genetics , Mice, Inbred C57BL , Cell Proliferation/genetics
4.
J Tradit Chin Med ; 44(2): 268-276, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38504533

ABSTRACT

OBJECTIVE: To investigate the effects of emodin on alkali burn-induced corneal inflammation and neovascularization. METHODS: The ability of emodin to target vascular endothelial growth factor receptor 2 (VEGFR2) was predicted by molecular docking. The effects of emodin on the invasion, migration, and proliferation of human umbilical vein endothelial cells (HUVEC) were determined by cell counting kit-8, Transwell, and tube formation assays. Analysis of apoptosis was performed by flow cytometry. CD31 levels were examined by immunofluorescence. The abundance and phosphorylation state of VEGFR2, protein kinase B (Akt), signal transducer and activator of transcription 3 (STAT3), and P38 were examined by immunoblot analysis. Corneal alkali burn was performed on 40 mice. Animals were divided randomly into two groups, and the alkali-burned eyes were then treated with drops of either 10 µM emodin or phosphate buffered saline (PBS) four times a day. Slit-lamp microscopy was used to evaluate inflammation and corneal neovascularization (CNV) in all eyes on Days 0, 7, 10, and 14. The mice were killed humanely 14 d after the alkali burn, and their corneas were removed and preserved at -80 ℃ until histological study or protein extraction. RESULTS: Molecular docking confirmed that emodin was able to target VEGFR2. The findings revealed that emodin decreased the invasion, migration, angiogenesis, and proliferation of HUVEC in a dose-dependent manner. In mice, emodin suppressed corneal inflammatory cell infiltration and inhibited the development of corneal neovascularization induced by alkali burn. Compared to those of the PBS-treated group, lower VEGFR2 expression and CD31 levels were found in the emodin-treated group. Emodin dramatically decreased the expression of VEGFR2, p-VEGFR2, p-Akt, p-STAT3, and p-P38 in VEGF-treated HUVEC. CONCLUSION: This study provides a new avenue for evaluating the molecular mechanisms underlying corneal inflammation and neovascularization. Emodin might be a promising new therapeutic option for corneal alkali burns.


Subject(s)
Burns, Chemical , Corneal Neovascularization , Emodin , Humans , Mice , Animals , Corneal Neovascularization/drug therapy , Corneal Neovascularization/genetics , Corneal Neovascularization/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Burns, Chemical/drug therapy , Burns, Chemical/metabolism , Burns, Chemical/pathology , Proto-Oncogene Proteins c-akt/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism , Molecular Docking Simulation , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/genetics , Signal Transduction , Human Umbilical Vein Endothelial Cells , Inflammation/drug therapy , Disease Models, Animal
5.
Invest Ophthalmol Vis Sci ; 65(3): 30, 2024 Mar 05.
Article in English | MEDLINE | ID: mdl-38517430

ABSTRACT

Purpose: Intraflagellar transport 46 (IFT46) is an integral subunit of the IFT-B complex, playing a key role in the assembly and maintenance of primary cilia responsible for transducing signaling pathways. Despite its predominant expression in the basal body of cilia, the precise role of Ift46 in ocular development remains undetermined. This study aimed to elucidate the impact of neural crest (NC)-specific deletion of Ift46 on ocular development. Methods: NC-specific conditional knockout mice for Ift46 (NC-Ift46F/F) were generated by crossing Ift46F mice with Wnt1-Cre2 mice, enabling the specific deletion of Ift46 in NC-derived cells (NCCs). Sonic Hedgehog (Shh) and Notch signaling activities in NC-Ift46F/F mice were evaluated using Gli1lacZ and CBF:H2B-Venus reporter mice, respectively. Cell fate mapping was conducted using ROSAmTmG reporter mice. Results: The deletion of Ift46 in NCCs resulted in a spectrum of ocular abnormalities, including thickened corneal stroma, hypoplasia of the anterior chamber, irregular iris morphology, and corneal neovascularization. Notably, this deletion led to reduced Shh signal activity in the periocular mesenchyme, sustained expression of key transcription factors Foxc1, Foxc2 and Pitx2, along with persistent cell proliferation. Additionally, it induced increased Notch signaling activity and the development of ectopic neovascularization within the corneal stroma. Conclusions: The absence of primary cilia due to Ift46 deficiency in NCCs is associated with anterior segment dysgenesis (ASD) and corneal neovascularization, suggesting a potential link to Axenfeld-Rieger syndrome, a disorder characterized by ASD. This underscores the pivotal role of primary cilia in ensuring proper anterior segment development and maintaining an avascular cornea.


Subject(s)
Cilia , Corneal Neovascularization , Eye Abnormalities , Mice , Animals , Cilia/metabolism , Neural Crest/metabolism , Corneal Neovascularization/metabolism , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Cornea , Mice, Knockout , Cytoskeletal Proteins/metabolism
6.
Int J Biol Macromol ; 261(Pt 2): 129933, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38309411

ABSTRACT

Corneal neovascularization (CNV) is a common multifactorial sequela of anterior corneal segment inflammation, which could lead to visual impairment and even blindness. The main treatments available are surgical sutures and invasive drug injections, which could cause serious ocular complications. To solve this problem, a thermo-sensitive drug-loaded hydrogel with high transparency was prepared in this study, which could achieve the sustained-release of drugs without affecting normal vision. In briefly, the thermo-sensitive hydrogel (PFNOCMC) was prepared from oxidized carboxymethyl cellulose (OCMC) and aminated poloxamer 407 (PF127-NH2). The results proved the PFNOCMC hydrogels possess high transparency, suitable gel temperature and time. In the CNV model, the PFNOCMC hydrogel loading bone morphogenetic protein 4 (BMP4) showed significant inhibition of CNV, this is due to the hydrogel allowed the drug to stay longer in the target area. The animal experiments on the ocular surface were carried out, which proved the hydrogel had excellent biocompatibility, and could realize the sustained-release of loaded drugs, and had a significant inhibitory effect on the neovascularization after ocular surface surgery. In conclusion, PFNOCMC hydrogels have great potential as sustained-release drug carriers in the biomedical field and provide a new minimally invasive option for the treatment of neovascular ocular diseases.


Subject(s)
Corneal Neovascularization , Hydrogels , Animals , Hydrogels/pharmacology , Corneal Neovascularization/drug therapy , Corneal Neovascularization/metabolism , Carboxymethylcellulose Sodium/therapeutic use , Delayed-Action Preparations/therapeutic use , Poloxamer/therapeutic use
7.
Ocul Surf ; 32: 13-25, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38191093

ABSTRACT

PURPOSE: Corneal fibrosis and neovascularization (CNV) after ocular trauma impairs vision. This study tested therapeutic potential of tissue-targeted adeno-associated virus5 (AAV5) mediated decorin (DCN) and pigment epithelium-derived factor (PEDF) combination genes in vivo. METHODS: Corneal fibrosis and CNV were induced in New Zealand White rabbits via chemical trauma. Gene therapy in stroma was delivered 30-min after chemical-trauma via topical AAV5-DCN and AAV5-PEDF application using a cloning cylinder. Clinical eye examinations and multimodal imaging in live rabbits were performed periodically and corneal tissues were collected 9-day and 15-day post euthanasia. Histological, cellular, and molecular and apoptosis assays were used for efficacy, tolerability, and mechanistic studies. RESULTS: The AAV5-DCN and AAV5-PEDF combination gene therapy significantly reduced corneal fibrosis (p < 0.01 or p < 0.001) and CNV (p < 0.001) in therapy-given (chemical-trauma and AAV5-DCN + AAV5-PEDF) rabbit eyes compared to the no-therapy given eyes (chemical-trauma and AAV5-naked vector). Histopathological analyses demonstrated significantly reduced fibrotic α-smooth muscle actin and endothelial lectin expression in therapy-given corneas compared to no-therapy corneas on day-9 (p < 0.001) and day-15 (p < 0.001). Further, therapy-given corneas showed significantly increased Fas-ligand mRNA levels (p < 0.001) and apoptotic cell death in neovessels (p < 0.001) compared to no-therapy corneas. AAV5 delivered 2.69 × 107 copies of DCN and 2.31 × 107 copies of PEDF genes per µg of DNA. AAV5 vector and delivered DCN and PEDF genes found tolerable to the rabbit eyes and caused no significant toxicity to the cornea. CONCLUSION: The combination AAV5-DCN and AAV5-PEDF topical gene therapy effectively reduces corneal fibrosis and CNV with high tolerability in vivo in rabbits. Additional studies are warranted.


Subject(s)
Corneal Neovascularization , Fibrosis , Genetic Therapy , Nerve Growth Factors , Serpins , Animals , Rabbits , Cornea/pathology , Cornea/metabolism , Corneal Neovascularization/therapy , Corneal Neovascularization/genetics , Corneal Neovascularization/pathology , Corneal Neovascularization/metabolism , Decorin/genetics , Decorin/metabolism , Dependovirus/genetics , Disease Models, Animal , Eye Proteins/genetics , Eye Proteins/metabolism , Fibrosis/therapy , Genetic Therapy/methods , Genetic Vectors , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Serpins/genetics , Serpins/metabolism
8.
Invest Ophthalmol Vis Sci ; 65(1): 37, 2024 Jan 02.
Article in English | MEDLINE | ID: mdl-38252525

ABSTRACT

Purpose: Previously we demonstrated that the secreted Ly-6/uPAR related protein 1 (SLURP1), abundantly expressed in the corneal epithelium (CE) and secreted into the tear fluid, serves as an antiangiogenic molecule. Here we describe the Slurp1-null (Slurp1X-/-) mouse corneal response to silver nitrate (AgNO3) cautery. Methods: Five days after AgNO3 cautery, we compared the wild-type (WT) and Slurp1X-/- mouse (1) corneal neovascularization (CNV) and immune cell influx by whole-mount immunofluorescent staining for CD31 and CD45, (2) macrophage and neutrophil infiltration by flow cytometry, and (3) gene expression by quantitative RT-PCR. Quantitative RT-PCR, immunofluorescent staining, and immunoblots were employed to evaluate the expression, phosphorylation status, and subcellular localization of NF-κB pathway components. Results: Unlike the WT, the Slurp1X-/- corneas displayed denser CNV in response to AgNO3 cautery, with more infiltrating macrophages and neutrophils and greater upregulation of the transcripts encoding VEGFA, MMP2, IL-1b, and vimentin. At 2, 7, and 10 days after AgNO3 cautery, Slurp1 expression was significantly downregulated in the WT corneas. Compared with the WT, naive Slurp1X-/- CE displayed increased phosphorylation of IKK(a/b), elevated phosphorylation of IκB with decreased amounts of total IκB, and higher phosphorylation of NF-κB, suggesting that NF-κB signaling is constitutively active in naive Slurp1X-/- corneas. Conclusions: Enhanced angiogenic inflammation in AgNO3 cauterized Slurp1X-/- corneas and constitutively active status of NF-κB signaling in the absence of Slurp1 suggest that Slurp1 modulates corneal angiogenic inflammation via NF-κB signaling.


Subject(s)
Corneal Neovascularization , Keratitis , Signal Transduction , Animals , Mice , Cornea , Corneal Neovascularization/metabolism , Corneal Neovascularization/pathology , Inflammation , Keratitis/metabolism , NF-kappa B
9.
Acta Pharmacol Sin ; 45(1): 166-179, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37605050

ABSTRACT

Dry eye disease (DED) is a prevalent ocular disorder with a multifactorial etiology. The pre-angiogenic and pre-inflammatory milieu of the ocular surface plays a critical role in its pathogenesis. DZ2002 is a reversible type III S-adenosyl-L-homocysteine hydrolase (SAHH) inhibitor, which has shown excellent anti-inflammatory and immunosuppressive activities in vivo and in vitro. In this study, we evaluated the therapeutic potential of DZ2002 in rodent models of DED. SCOP-induced dry eye models were established in female rats and mice, while BAC-induced dry eye model was established in female rats. DZ2002 was administered as eye drops (0.25%, 1%) four times daily (20 µL per eye) for 7 or 14 consecutive days. We showed that topical application of DZ2002 concentration-dependently reduced corneal neovascularization and corneal opacity, as well as alleviated conjunctival irritation in both DED models. Furthermore, we observed that DZ2002 treatment decreased the expression of genes associated with angiogenesis and the levels of inflammation in the cornea and conjunctiva. Moreover, DZ2002 treatment in the BAC-induced DED model abolished the activation of the STAT3-PI3K-Akt-NF-κB pathways in corneal tissues. We also found that DZ2002 significantly inhibited the proliferation, migration, and tube formation of human umbilical endothelial cells (HUVECs) while downregulating the activation of the STAT3-PI3K-Akt-NF-κB pathway. These results suggest that DZ2002 exerts a therapeutic effect on corneal angiogenesis in DED, potentially by preventing the upregulation of the STAT3-PI3K-Akt-NF-κB pathways. Collectively, DZ2002 is a promising candidate for ophthalmic therapy, particularly in treating DED.


Subject(s)
Corneal Neovascularization , Dry Eye Syndromes , Rats , Humans , Mice , Animals , Female , Corneal Neovascularization/drug therapy , Corneal Neovascularization/metabolism , Corneal Neovascularization/pathology , NF-kappa B/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Rodentia/metabolism , Endothelial Cells/metabolism , Angiogenesis , Inflammation/drug therapy , Dry Eye Syndromes/drug therapy , Dry Eye Syndromes/chemically induced , STAT3 Transcription Factor/metabolism
10.
Wien Klin Wochenschr ; 136(5-6): 154-162, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37261487

ABSTRACT

BACKGROUND: Corneal neovascularization (CNV) is a vision-threatening disease and an increasing public health concern. It was found that administering an Akt inhibitor in the second phase of retinopathy significantly decreased retinal neovascularization. METHODS: This study investigated the effect of an Akt inhibitor on the angiogenesis of human umbilical vein endothelial cells (HUVECs) and its impacts on the degree of CNV and corneal opacity in a rat keratoplasty model. Cell Counting Kit-8 (CCK-8) and 5-ethynyl-2'-deoxyuridine (EdU) assays, tube formation assays, cell scratch experiments, and a fully allogeneic corneal transplant model were performed. RESULTS: It was found that an Akt inhibitor inhibited the proliferation, angiogenesis, and migration of HUVECs induced by vascular endothelial growth factor (VEGF). The results showed that both CNV and corneal opacity were decreased in rats after Akt inhibitor administration. CONCLUSION: The research illustrates the vital role of Akt inhibitors in mediating CNV. The analysis shows that the Akt inhibitor may provide a novel and feasible therapeutic approach to prevent CNV, but its mechanism needs further investigation.


Subject(s)
Corneal Neovascularization , Corneal Opacity , Humans , Rats , Animals , Corneal Neovascularization/drug therapy , Corneal Neovascularization/metabolism , Corneal Neovascularization/prevention & control , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-akt/pharmacology , Proto-Oncogene Proteins c-akt/therapeutic use , Human Umbilical Vein Endothelial Cells/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor A/therapeutic use , Angiogenesis , Corneal Opacity/drug therapy , Corneal Opacity/metabolism , Cell Proliferation , Angiogenesis Inhibitors
11.
Adv Healthc Mater ; 13(5): e2302192, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38018632

ABSTRACT

Corneal neovascularization (CNV) is one of the leading causes of blindness in the world. In clinical practice; however, it remains a challenge to achieve a noninvasive and safe treatment. Herein, a biocompatible shell with excellent antioxidant and antivascularity is prepared by co-assembly of epigallocatechin gallate/gallic acid and Cu (II). After loading glucose oxidase (GOx) inside, the shell is modified with dimeric DPA-Zn for codelivering vascular endothelial growth factor (VEGF) small interfering RNA (VEGF-siRNA). Meanwhile, the Arg-Gly-Asp peptide (RGD) peptide-engineered cell membranes coating improves angiogenesis-targeting and is biocompatible for the multifunctional nanomedicine (CEGs/RGD). After eye drops administration, CEGs/RGD targets enrichment in neovascularization and CEGs NPs enter cells. Then, the inner GOx consumes glucose with a decrease in local pH, which in turn leads to the release of EGCE and VEGF-siRNA. As a result, the nanomedicines significantly reduce angiogenesis and inhibit CNV formation through synergistic effect of antioxidant and antivascular via down-regulation of cluster of differentiation 31 and VEGF. The nanomedicine represents a safe and efficient CNV treatment through the combined effect of antioxidant/gene, which provides important theoretical and clinical significance.


Subject(s)
Corneal Neovascularization , Humans , Corneal Neovascularization/drug therapy , Corneal Neovascularization/metabolism , Vascular Endothelial Growth Factor A/metabolism , Antioxidants/pharmacology , Antioxidants/therapeutic use , RNA, Small Interfering/pharmacology , Oligopeptides/pharmacology
12.
Exp Eye Res ; 238: 109739, 2024 01.
Article in English | MEDLINE | ID: mdl-38042515

ABSTRACT

Corneal alkali burns often occur in industrial production and daily life, combined with infection, and may cause severe eye disease. Oxidative stress and neovascularization (NV) are important factors leading to a poor prognosis. URP20 is an antimicrobial peptide that has been proven to treat bacterial keratitis in rats through antibacterial and anti-NV effects. Therefore, in this study, the protective effect and influence mechanism of URP20 were explored in a rat model of alkali burn together with pathogenic bacteria (Staphylococcus aureus and Escherichia coli) infection. In addition, human umbilical vein endothelial cells (HUVECs) and human corneal epithelial cells (HCECs) were selected to verify the effects of URP20 on vascularization and oxidative stress. The results showed that URP20 treatment could protect corneal tissue, reduce corneal turbidity, and reduce the NV pathological score. Furthermore, URP20 significantly inhibited the expression of the vascularization marker proteins VEGFR2 and CD31. URP20 also reduced the migration ability of HUVECs. In terms of oxidative stress, URP20 significantly upregulated SOD and GSH contents in corneal tissue and HCECs (treated with 200 µM H2O2) and promoted the expression of the antioxidant protein Nrf2/HO-1. At the same time, MDA and ROS levels were also inhibited. In conclusion, URP20 could improve corneal injury combined with bacterial infection in rats caused by alkali burns through antibacterial, anti-NV, and antioxidant activities.


Subject(s)
Bacterial Infections , Burns, Chemical , Corneal Injuries , Corneal Neovascularization , Eye Burns , Rats , Humans , Animals , Burns, Chemical/complications , Burns, Chemical/drug therapy , Burns, Chemical/metabolism , Corneal Neovascularization/metabolism , Antioxidants/pharmacology , Antioxidants/therapeutic use , Hydrogen Peroxide/pharmacology , Neovascularization, Pathologic/metabolism , Corneal Injuries/drug therapy , Human Umbilical Vein Endothelial Cells , Anti-Bacterial Agents/therapeutic use , Anti-Bacterial Agents/pharmacology , Eye Burns/chemically induced , Eye Burns/drug therapy , Eye Burns/pathology , Disease Models, Animal , Alkalies/toxicity
13.
Exp Eye Res ; 237: 109680, 2023 12.
Article in English | MEDLINE | ID: mdl-37858608

ABSTRACT

Corneal neovascularization (CNV) can lead to impaired corneal transparency, resulting in vision loss or blindness. The primary pathological mechanism underlying CNV is an imbalance between pro-angiogenic and anti-angiogenic factors, with inflammation playing a crucial role. Notably, a vascular endothelial growth factor(VEGF)-A gradient triggers the selection of single endothelial cells(ECs) into primary tip cells that guide sprouting, while a dynamic balance between tip and stalk cells maintains a specific ratio to promote CNV. Despite the central importance of tip-stalk cell selection and shuffling, the underlying mechanisms remain poorly understood. In this study, we examined the effects of bone morphogenetic protein 4 (BMP4) on VEGF-A-induced lumen formation in human umbilical vein endothelial cells (HUVECs) and CD34-stained tip cell formation. In vivo, BMP4 inhibited CNV caused by corneal sutures. This process was achieved by BMP4 decreasing the protein expression of VEGF-A and VEGFR2 in corneal tissue after corneal suture injury. By observing the ultrastructure of the cornea, BMP4 inhibited the sprouting of tip cells and brought forward the appearance of intussusception. Meanwhile, BMP4 attenuated the inflammatory response by inhibiting neutrophil extracellular traps (NETs)formation through the NADPH oxidase-2(NOX-2)pathway. Our results indicate that BMP4 inhibits the formation of tip cells by reducing the generation of NETs, disrupting the dynamic balance of tip and stalk cells and thereby inhibiting CNV, suggesting that BMP4 may be a potential therapeutic target for CNV.


Subject(s)
Corneal Injuries , Corneal Neovascularization , Humans , Corneal Neovascularization/metabolism , Bone Morphogenetic Protein 4/metabolism , Vascular Endothelial Growth Factor A/metabolism , Cornea/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Corneal Injuries/metabolism , Neovascularization, Physiologic
14.
Exp Eye Res ; 233: 109539, 2023 08.
Article in English | MEDLINE | ID: mdl-37315833

ABSTRACT

Alkali burn-induced corneal injury often causes inflammation and neovascularization and leads to compromised vision. We previously reported that rapamycin ameliorated corneal injury after alkali burns by methylation modification. In this study, we aimed to investigate the rapamycin-medicated mechanism against corneal inflammation and neovascularization. Our data showed that alkali burn could induce a range of different inflammatory response, including a stark upregulation of pro-inflammatory factor expression and an increase in the infiltration of myeloperoxidase- and F4/80-positive cells from the corneal limbus to the central stroma. Rapamycin effectively downregulated the mRNA expression levels of tumor necrosis factor-alpha (TNF-α), interleukin-1beta (IL-1ß), toll-like receptor 4 (TLR4), nucleotide binding oligomerization domain-like receptors (NLR) family pyrin domain-containing 3 (NLRP3), and Caspase-1, and suppressed the infiltration of neutrophils and macrophages. Inflammation-related angiogenesis mediated by matrix metalloproteinase-2 (MMP-2) and rapamycin restrained this process by inhibiting the TNF-α upregulation in burned corneas of mice. Rapamycin also restrained corneal alkali burn-induced inflammation by regulating HIF-1α/VEGF-mediated angiogenesis and the serum cytokines TNF-α, IL-6, Interferon-gamma (IFN-γ) and granulocyte-macrophage colony-stimulating factor (GM-CSF). The findings of this study indicated rapamycin may reduce inflammation-associated infiltration of inflammatory cells, shape the expression of cytokines, and balance the regulation of MMP-2 and HIF-1α-mediated inflammation and angiogenesis by suppressing mTOR activation in corneal wound healing induced by an alkali injury. It offered novel insights relevant for a potent drug for treating corneal alkali burn.


Subject(s)
Burns, Chemical , Corneal Injuries , Corneal Neovascularization , Eye Burns , Mice , Animals , Matrix Metalloproteinase 2/metabolism , Burns, Chemical/metabolism , Corneal Neovascularization/metabolism , Tumor Necrosis Factor-alpha/metabolism , Sirolimus/pharmacology , Sirolimus/therapeutic use , Alkalies/toxicity , Cornea/metabolism , Neovascularization, Pathologic/metabolism , Corneal Injuries/metabolism , Inflammation/metabolism , Cytokines/metabolism , Eye Burns/chemically induced , Eye Burns/drug therapy , Eye Burns/pathology , Disease Models, Animal
15.
Invest Ophthalmol Vis Sci ; 64(4): 10, 2023 04 03.
Article in English | MEDLINE | ID: mdl-37040097

ABSTRACT

Purpose: Chemokine receptor 4 (CXCR4) plays an essential role in the early stage of corneal neovascularization (CNV), but the underlying key molecular mechanism has yet to be addressed. This study aimed to explore the new molecular mechanism of CXCR4 in CNV and the related pathological events. Methods: CXCR4 was assayed by immunofluorescence or Western blotting. The function of the supernatant from hypoxia-treated human corneal epithelial cells (HCE-T) cells was examined by culturing with human umbilical vein endothelial cells. MicroRNA sequencing was used to detect the downstream microRNAs upon CXCR4 knockdown and analyzed by preliminary bioinformatics. The proangiogenic functions and downstream target genes of microRNA were investigated by gene interference and luciferase assay. An alkali-burned murine model was introduced to examine the function and mechanism of miR-1910-5p in vivo. Results: High CXCR4 expression was confirmed in corneal tissues of patients with CNV and hypoxic HCE-T cells. The supernatant from hypoxia-treated HCE-T cells is involved in the CXCR4-mediated angiogenesis of human umbilical vein endothelial cells. Notably, miR-1910-5p was demonstrated to be at a high level in wild-type HCE-T cells and its supernatant, and in CNV patient tears. The proangiogenic functions of miR-1910-5p were demonstrated with the assays of cell migration, tube formation, and aortic ring. Moreover, miR-1910-5p significantly inhibited multimerin-2 expression by targeting its 3' untranslated region and caused significant extracellular junctional defects in human umbilical vein endothelial cells. MiR-1910-5p antagomir could significantly increase multimerin-2 level and decrease vascular leakage, and ultimately inhibit CNV in a murine model. Conclusions: Our results revealed a novel CXCR4-mediated mechanism and proved that targeting the miR-1910-5p/multimerin-2 pathway could be a promising therapeutic target for CNV.


Subject(s)
Corneal Neovascularization , MicroRNAs , Animals , Humans , Mice , Capillary Permeability , Corneal Neovascularization/metabolism , Disease Models, Animal , Human Umbilical Vein Endothelial Cells/metabolism , Hypoxia/metabolism , MicroRNAs/genetics , Receptors, CXCR4/metabolism
16.
Biochim Biophys Acta Mol Basis Dis ; 1869(6): 166708, 2023 08.
Article in English | MEDLINE | ID: mdl-37019244

ABSTRACT

BACKGROUND: Corneal neovascularization (CNV) can be caused by chemical burns. Macrophages are involved in angiogenesis and lymphangiogenesis during CNV. The aim of this study was to investigate whether Wilms' tumor 1-associated protein (WTAP) is involved in macrophage recruitment and VEGF secretion via N6-methyladenosine (m6A) modification. METHODS: A CNV mouse model was established by corneal alkali burn. Tumor necrosis factor alpha (TNF-α) was used to stimulate vascular endothelial cells. m6A immunoprecipitation qPCR was used to determine the enrichment of m6A levels in mRNAs. The H3K9me3 enrichment in the promoter region of CC motif chemokine ligand 2 (CCL2) was detected by chromatin immunoprecipitation assay. The WTAP inhibition in vivo was performed using the adeno-associated virus. RESULTS: In the alkali burn corneal tissues, angiogenesis and lymphangiogenesis were promoted as CD31 and LYVE-1 expressions were elevated, and the number of macrophages as well as WTAP expression were increased. Under the TNF-α stimulation, WTAP promoted the recruitment of endothelial cells to macrophages by promoting CCL2 secretion. Mechanistically, WTAP affected the enrichment of H3K9me3 at the CCL2 promoter by regulating the m6A level of SUV39H1 mRNA. The in vivo experiment showed that VEGFA/C/D secretion of macrophages was reduced after WTAP interference. Mechanistically, WTAP regulated the translational efficiency of HIF-1α via m6A modification. CONCLUSION: WTAP affected macrophage recruitment to endothelial cells via regulation of H3K9me3-mediated CCL2 transcription. WTAP also affected macrophage secretion of VEGFA/C/D via m6A-mediated translation regulation of HIF-1α. Both pathways were involved in the WTAP regulation of angiogenesis and lymphangiogenesis during CNV.


Subject(s)
Burns, Chemical , Corneal Neovascularization , Mice , Animals , Corneal Neovascularization/genetics , Corneal Neovascularization/metabolism , Corneal Neovascularization/pathology , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Endothelial Cells/metabolism , Burns, Chemical/metabolism , Burns, Chemical/pathology , Macrophages/metabolism
17.
Semin Ophthalmol ; 38(7): 670-678, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37058000

ABSTRACT

OBJECTIVE: To study the efficacy of Conbercept for the treatment of corneal neovascularization (NV) in a rabbit model. METHODS: NV was induced by placing sutures. Eight rabbits were used as a control. The other 136 rabbits were randomly divided into two equal groups, and 68 rabbits in each group were divided into four subgroups and given different treatments. Time-course photographs, histological examination, and enzyme-linked immunoassay ELISA analysis for vascular endothelial growth factor were performed at weeks 1, 2, and 3 after injection placement. RESULTS: At weeks 1, 2, and 3 after injection placement, there was less expression of corneal NV and VEGF in the conbercept-treated groups than in the saline-treated control groups and less corneal NV and VEGF were expressed in the early treatment group than in the late treatment group. At weeks 2 and 3 after injection, there were fewer corneal NV (length and area) in the early intrastromal injection group with conbercept than in the early subconjunctival injection group with conbercept and a smaller diameter of corneal NV than in the late intrastromal injection group treated with conbercept. Histological examination showed a smaller diameter of corneal NV in all eyes in conbercept-treated groups 1 w after injection than before injection. Treatment with subconjunctival injection with conbercept led to a larger diameter at weeks 2 and 3 than at week 1. CONCLUSIONS: Subconjunctival and intrastromal administrations of conbercept effectively inhibit corneal NV in rabbits, and the latter has the better effect. The effect is the best in the group with cornea intrastromal injection of conbercept 1 w after suture. Early administration of conbercept may successfully inhibit corneal NV in an animal model.


Subject(s)
Angiogenesis Inhibitors , Corneal Neovascularization , Animals , Humans , Rabbits , Bevacizumab/therapeutic use , Corneal Neovascularization/drug therapy , Corneal Neovascularization/metabolism , Corneal Neovascularization/pathology , Vascular Endothelial Growth Factor A , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Disease Models, Animal
18.
Invest Ophthalmol Vis Sci ; 64(3): 20, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36897151

ABSTRACT

Purpose: To investigate the role of the sympathetic nervous system in corneal neovascularization (CNV) and to identify the downstream pathway involved in this regulation. Methods: Three types of CNV models were constructed with C57BL/6J mice, including the alkali burn model, suture model, and basic fibroblast growth factor (bFGF) corneal micropocket model. Subconjunctival injection of the sympathetic neurotransmitter norepinephrine (NE) was administered in these three models. Control mice received injections of water of the same volume. The corneal CNV was detected using slit-lamp microscopy and immunostaining with CD31, and the results were quantified by ImageJ. The expression of ß2-adrenergic receptor (ß2-AR) was stained with mouse corneas and human umbilical vein endothelial cells (HUVECs). Furthermore, the anti-CNV effects of ß2-AR antagonist ICI-118,551 (ICI) were examined with HUVEC tube formation assay and with a bFGF micropocket model. Additionally, partial ß2-AR knockdown mice (Adrb2+/-) were used to establish the bFGF micropocket model, and the corneal CNV size was quantified based on the slit-lamp images and vessel staining. Results: Sympathetic nerves invaded the cornea in the suture CNV model. The NE receptor ß2-AR was highly expressed in corneal epithelium and blood vessels. The addition of NE significantly promoted corneal angiogenesis, whereas ICI effectively inhibited CNV invasion and HUVEC tube formation. Adrb2 knockdown significantly reduced the cornea area occupied by CNV. Conclusions: Our study found that sympathetic nerves grow into the cornea in conjunction with newly formed vessels. The addition of the sympathetic neurotransmitter NE and activation of its downstream receptor ß2-AR promoted CNV. Targeting ß2-AR could potentially be used as an anti-CNV strategy.


Subject(s)
Corneal Neovascularization , Mice , Humans , Animals , Corneal Neovascularization/metabolism , Norepinephrine/pharmacology , Mice, Inbred C57BL , Cornea/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Disease Models, Animal
19.
Int Immunopharmacol ; 116: 109680, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36739832

ABSTRACT

Alkali burn-induced corneal inflammation and subsequent corneal neovascularization (CNV) are major causes of corneal opacity and vision loss. M1 macrophages play a central role in inflammation and CNV. Therefore, modulation of M1 macrophage polarization is a promising strategy for corneal alkali burns. Here, we illustrate the effect and underlying mechanisms of upadacitinib on corneal inflammation and CNV induced by alkali burns in mice. The corneas of BALB/c mice were administered with 1 M NaOH for 30 s and randomly assigned to the vehicle group and the upadacitinib-treated group. Corneal opacity and corneal epithelial defects were assessed clinically. Quantitative real-time PCR (qRT-PCR), immunohistochemistry, and western blot analysis were performed to detect M1 macrophage polarization and CD31+ corneal blood vessels. The results showed that upadacitinib notably decreased corneal opacity, and promoted corneal wound healing. On day 7 and 14 after alkali burns, upadacitinib significantly suppressed CNV. Corneal alkali injury caused M1 macrophage recruitment in the cornea. In contrast to the vehicle, upadacitinib suppressed M1 macrophage infiltration and decreased the mRNA expression levels of inducible nitric oxide synthase (iNOS), monocyte chemotactic protein-1 (MCP-1), tumor necrosis factor-alpha (TNF-α), interleukin (IL)-6, IL-1ß, and vascular endothelial growth factor A (VEGF-A) in alkali-injured corneas. Moreover, upadacitinib dose-dependently inhibited M1 macrophage polarization by suppressing interferon (IFN)-γ-/lipopolysaccharide-stimulated STAT1 activation in vitro. Our findings reveal that upadacitinib can efficiently alleviate alkali-induced corneal inflammation and neovascularization by inhibiting M1 macrophage infiltration. These data demonstrate that upadacitinib is an effective drug for the treatment of corneal alkali burns.


Subject(s)
Burns, Chemical , Corneal Injuries , Corneal Neovascularization , Corneal Opacity , Eye Burns , Keratitis , Mice , Animals , Burns, Chemical/drug therapy , Burns, Chemical/pathology , Vascular Endothelial Growth Factor A/metabolism , Alkalies/adverse effects , Alkalies/metabolism , Cornea , Corneal Neovascularization/chemically induced , Corneal Neovascularization/drug therapy , Corneal Neovascularization/metabolism , Corneal Injuries/metabolism , Macrophages/metabolism , Keratitis/chemically induced , Keratitis/drug therapy , Inflammation/metabolism , Corneal Opacity/complications , Corneal Opacity/metabolism , Corneal Opacity/pathology , Eye Burns/chemically induced , Eye Burns/drug therapy , Eye Burns/pathology , Disease Models, Animal
20.
Cells ; 12(2)2023 01 14.
Article in English | MEDLINE | ID: mdl-36672254

ABSTRACT

Corneal lymphangiogenesis is one component of the neovascularization observed in several inflammatory pathologies of the cornea including dry eye disease and corneal graft rejection. Following injury, corneal (lymph)angiogenic privilege is impaired, allowing ingrowth of blood and lymphatic vessels into the previously avascular cornea. While the mechanisms underlying pathological corneal hemangiogenesis have been well described, knowledge of the lymphangiogenesis guidance mechanisms in the cornea is relatively scarce. Various signaling pathways are involved in lymphangiogenesis guidance in general, each influencing one or multiple stages of lymphatic vessel development. Most endogenous factors that guide corneal lymphatic vessel growth or regression act via the vascular endothelial growth factor C signaling pathway, a central regulator of lymphangiogenesis. Several exogenous factors have recently been repurposed and shown to regulate corneal lymphangiogenesis, uncovering unique signaling pathways not previously known to influence lymphatic vessel guidance. A strong understanding of the relevant lymphangiogenesis guidance mechanisms can facilitate the development of targeted anti-lymphangiogenic therapeutics for corneal pathologies. In this review, we examine the current knowledge of lymphatic guidance cues, their regulation of inflammatory states in the cornea, and recently discovered anti-lymphangiogenic therapeutic modalities.


Subject(s)
Corneal Neovascularization , Lymphatic Vessels , Humans , Lymphangiogenesis , Corneal Neovascularization/drug therapy , Corneal Neovascularization/metabolism , Corneal Neovascularization/pathology , Vascular Endothelial Growth Factor C/metabolism , Cornea/metabolism , Lymphatic Vessels/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...