Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 539
Filter
1.
J Nanobiotechnology ; 22(1): 290, 2024 May 27.
Article in English | MEDLINE | ID: mdl-38802884

ABSTRACT

Corneal neovascularization (CNV) is one of the common blinding factors worldwide, leading to reduced vision or even blindness. However, current treatments such as surgical intervention and anti-VEGF agent therapy still have some shortcomings or evoke some adverse effects. Recently, SU6668, an inhibitor targeting angiogenic tyrosine kinases, has demonstrated growth inhibition of neovascularization. But the hydrophobicity and low ocular bioavailability limit its application in cornea. Hereby, we proposed the preparation of SU6668 pure nanoparticles (NanoSU6668; size ~135 nm) using a super-stable pure-nanomedicine formulation technology (SPFT), which possessed uniform particle size and excellent aqueous dispersion at 1 mg/mL. Furthermore, mesenchymal stem cell membrane vesicle (MSCm) was coated on the surface of NanoSU6668, and then conjugated with TAT cell penetrating peptide, preparing multifunctional TAT-MSCm@NanoSU6668 (T-MNS). The T-MNS at a concentration of 200 µg/mL was treated for CNV via eye drops, and accumulated in blood vessels with a high targeting performance, resulting in elimination of blood vessels and recovery of cornea transparency after 4 days of treatment. Meanwhile, drug safety test confirmed that T-MNS did not cause any damage to cornea, retina and other eye tissues. In conclusion, the T-MNS eye drop had the potential to treat CNV effectively and safely in a low dosing frequency, which broke new ground for CNV theranostics.


Subject(s)
Cornea , Corneal Neovascularization , Nanoparticles , Ophthalmic Solutions , Corneal Neovascularization/drug therapy , Animals , Nanoparticles/chemistry , Ophthalmic Solutions/chemistry , Cornea/metabolism , Cornea/drug effects , Mice , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/therapeutic use , Angiogenesis Inhibitors/pharmacology , Particle Size , Humans , Male , Mice, Inbred C57BL , Rabbits
2.
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
3.
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
4.
J Nanobiotechnology ; 22(1): 134, 2024 Mar 28.
Article in English | MEDLINE | ID: mdl-38549081

ABSTRACT

BACKGROUND: Corneal neovascularization (CoNV) threatens vision by disrupting corneal avascularity, however, current treatments, including pharmacotherapy and surgery, are hindered by limitations in efficacy and adverse effects. Minocycline, known for its anti-inflammatory properties, could suppress CoNV but faces challenges in effective delivery due to the cornea's unique structure. Therefore, in this study a novel drug delivery system using minocycline-loaded nano-hydroxyapatite/poly (lactic-co-glycolic acid) (nHAP/PLGA) nanoparticles was developed to improve treatment outcomes for CoNV. RESULTS: Ultra-small nHAP was synthesized using high gravity technology, then encapsulated in PLGA by a double emulsion method to form nHAP/PLGA microspheres, attenuating the acidic by-products of PLGA degradation. The MINO@PLGA nanocomplex, featuring sustained release and permeation properties, demonstrated an efficient delivery system for minocycline that significantly inhibited the CoNV area in an alkali-burn model without exhibiting apparent cytotoxicity. On day 14, the in vivo microscope examination and ex vivo CD31 staining corroborated the inhibition of neovascularization, with the significantly smaller CoNV area (29.40% ± 6.55%) in the MINO@PLGA Tid group (three times daily) than that of the control group (86.81% ± 15.71%), the MINO group (72.42% ± 30.15%), and the PLGA group (86.87% ± 14.94%) (p < 0.05). Fluorescein sodium staining show MINO@PLGA treatments, administered once daily (Qd) and three times daily (Tid) demonstrated rapid corneal epithelial healing while the Alkali injury group and the DEX group showed longer healing times (p < 0.05). Additionally, compared to the control group, treatments with dexamethasone, MINO, and MINO@PLGA were associated with an increased expression of TGF-ß as evidenced by immunofluorescence, while the levels of pro-inflammatory cytokines IL-1ß and TNF-α demonstrated a significant decrease following alkali burn. Safety evaluations, including assessments of renal and hepatic biomarkers, along with H&E staining of major organs, revealed no significant cytotoxicity of the MINO@PLGA nanocomplex in vivo. CONCLUSIONS: The novel MINO@PLGA nanocomplex, comprising minocycline-loaded nHAP/PLGA microspheres, has shown a substantial capacity for preventing CoNV. This study confirms the complex's ability to downregulate inflammatory pathways, significantly reducing CoNV with minimal cytotoxicity and high biosafety in vivo. Given these findings, MINO@PLGA stands as a highly promising candidate for ocular conditions characterized by CoNV.


Subject(s)
Corneal Neovascularization , Minocycline , Humans , Minocycline/pharmacology , Corneal Neovascularization/drug therapy , Corneal Neovascularization/prevention & control , Microspheres , Angiogenesis , Alkalies
5.
ACS Nano ; 18(11): 8209-8228, 2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38452114

ABSTRACT

Protein drugs have been widely used in treating various clinical diseases because of their high specificity, fewer side effects, and favorable therapeutic effect, but they greatly suffer from their weak permeability through tissue barriers, high sensitivity to microenvironments, degradation by proteases, and rapid clearance by the immune system. Herein, we disrupted the standard protocol where protein drugs must be delivered as the cargo via a delivery system and innovatively developed a free entrapping matrix strategy by simply mixing bevacizumab (Beva) with zinc ions to generate Beva-NPs (Beva-Zn2+), where Beva is coordinatively cross-linked by zinc ions with a loading efficiency as high as 99.2% ± 0.41%. This strategy was universal to generating various protein NPs, with different metal ions (Cu2+, Fe3+, Mg2+, Sr2+). The synthetic conditions of Beva-NPs were optimized, and the generated mechanism was investigated in detail. The entrapment, releasing profile, and the bioactivities of released Beva were thoroughly studied. By using in situ doping of the fourth-generation polyamindoamine dendrimer (G4), the Beva-G4-NPs exhibited extended ocular retention and penetration through biobarriers in the anterior segment through transcellular and paracellular pathways, effectively inhibiting corneal neovascularization (CNV) from 91.6 ± 2.03% to 13.5 ± 1.87% in a rat model of CNV. This study contributes to engineering of protein NPs by using a facile strategy for overcoming the weaknesses of protein drugs and protein NPs, such as weak tissue barrier permeability, low encapsulation efficiency, poor loading capacity, and susceptibility to inactivation.


Subject(s)
Corneal Neovascularization , Nanoparticles , Rats , Animals , Corneal Neovascularization/drug therapy , Nanoparticles/therapeutic use , Ions , Zinc
6.
Photodiagnosis Photodyn Ther ; 46: 104067, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38548042

ABSTRACT

BACKGROUND: To the best of our knowledge, no studies have been performed to determine the optimal parameters of photodynamic therapy (PDT) combined with subconjunctival injection of bevacizumab for corneal neovascularization. This study aimed to compare the effect of photodynamic therapy with two different sets of parameters combined with subconjunctival injection of bevacizumab for corneal neovascularization. METHODS: Patients with stable corneal neovascularization (CNV) unresponsive to conventional treatment (topical steroid) were included in this study. Patients were divided into two groups, receiving PDT with two different sets of parameters (group 1 receiving fluence of 50 J/cm2 at 15 min after intravenous injection of verteporfin with, group 2 receiving fluence of 150 J/cm2 at 60 min after intravenous injection of verteporfin with). Subconjunctival injection of bevacizumab was performed immediately after PDT. All patients were followed for 6 months. Best-corrected visual acuity and intraocular pressure were evaluated, and slit-lamp biomicroscopy as well as digital photography were performed. Average diameter and cumulative length of corneal neovascular were measured to evaluate the corneal neovascularization. RESULTS: Seventeen patients (20 eyes) were included in this study. At the last visit, the vision was improved in 12 eyes (60 %), steady in 4 eyes (20 %) and worsen in 4 eyes (20 %). The intraocular pressure (IOP) of all patients remained in normal range. A significant decrease in corneal neovascularization was showed in all the eyes after treatment. At 6 months after the combined treatment, the average diameter and cumulative length of vessels significantly decreased to 0.041 ± 0.023 mm (P < 0.05) and 18.78 ± 17.73 mm (P < 0.05), respectively, compared with the pretreatment data (0.062 ± 0.015 mm, 31.48 ± 18.21 mm). The reduction was more remarkable in group 2 compared to group 1.In group 1, the average diameter was 0.062 ± 0.013mm before and 0.056 ± 0.017mm after, the cumulative length of vessels was 38.66 ± 22.55mm before and 31.21 ± 17.30 after. In group 2, the date were 0.061 ± 0.016mm before and 0.029 ± 0.020mm after, 25.60 ± 8.95 mm before and 8.61 ± 8.26 mm. The reported complications included epithelial defect in four eyes, small white filaments in two eyes and corneal epithelial erosion in two eyes. CONCLUSION: The PDT combined with subconjunctival injection of bevacizumab was effective for the chronic corneal neovascularization. A more promising treatment outcome was observed when PDT was performed at 60 min after intravenous injection of verteporfin with fluence of 150 J/cm2. No serious complications or systemic events were observed throughout the follow-up period.


Subject(s)
Angiogenesis Inhibitors , Bevacizumab , Corneal Neovascularization , Photochemotherapy , Photosensitizing Agents , Verteporfin , Visual Acuity , Humans , Photochemotherapy/methods , Bevacizumab/administration & dosage , Bevacizumab/therapeutic use , Corneal Neovascularization/drug therapy , Female , Male , Photosensitizing Agents/administration & dosage , Photosensitizing Agents/therapeutic use , Verteporfin/therapeutic use , Angiogenesis Inhibitors/administration & dosage , Middle Aged , Visual Acuity/drug effects , Adult , Aged , Combined Modality Therapy , Injections, Intraocular , Intraocular Pressure/drug effects , Porphyrins/administration & dosage , Conjunctiva/blood supply
7.
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
8.
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
9.
Small ; 20(2): e2302765, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37679056

ABSTRACT

Corneal neovascularization (CoNV) is a major cause of visual impairment worldwide. Currently, available treatment options have limited efficacy and are associated with adverse effects due to biological barriers and clearance mechanisms. To address this challenge, a novel topical delivery system is developed-Gel 2_1&Eylea-an aflibercept-loaded eye-drop hydrogel mediated with cell-penetrating peptide 1. Gel 2_1&Eylea demonstrates superior membrane permeability, increased stability, and prolonged drug retention time on the ocular surface, and thus may improve drug efficacy. In a rabbit CoNV model, Gel 2_1&Eylea significantly reduces the density of neovascularization with no adverse effects on normal corneoscleral limbal vessels, demonstrating high efficacy and biocompatibility. This work identifies a promising treatment for CoNV which has the potential to benefit other ocular neovascular diseases.


Subject(s)
Cell-Penetrating Peptides , Corneal Neovascularization , Receptors, Vascular Endothelial Growth Factor , Recombinant Fusion Proteins , Animals , Rabbits , Corneal Neovascularization/drug therapy , Hydrogels , Ophthalmic Solutions/therapeutic use
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.
Biomed J ; 47(1): 100592, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37004870

ABSTRACT

BACKGROUND: To overcome the drawbacks of traditional therapy for corneal neovascularization (CNV), we evaluated the efficacy of polyethylene glycol (PEG)-conjugated Ala-Pro-Arg-Pro-Gly (APRPG) peptide modified dexamethasone (Dex), a novel nano-prodrug (Dex-PEG-APRPG, DPA). METHODS: Characterization of DPA nano-prodrug were measured with transmission electron microscopy (TEM) and dynamic light scattering (DLS) analyses. Cytotoxicity and effects on cell migration and tube formation of DPA were evaluated in vitro. A murine CNV model was established by cornea alkali burn. The injured corneas were given eye drops of DPA (0.2 mM), Dex solution (0.2 mM), Dexp (2 mM), or normal saline three times a day. After two weeks, eyes were obtained for the analysis of histopathology, immunostaining, and mRNA expression. RESULTS: DPA with an average diameter of 30 nm, presented little cytotoxicity and had good ocular biocompatibility. More importantly, DPA showed specific targeting to vascular endothelial cells with efficient inhibition on cell migration and tube formation. In a mouse CNV model, clinical, histological, and immunohistochemical examination results revealed DPA had a much stronger angiogenesis suppression than Dex, resembling a clinical drug with an order of magnitude higher concentration. This was ascribed to the significant downregulations in the expression of pro-angiogenic and pro-inflammatory factors in the corneas. In vivo imaging results also demonstrated that APRPG could prolong ocular retention time. CONCLUSIONS: This study suggests that DPA nano-prodrug occupies advantages of specific targeting ability and improved bioavailability over conventional therapy, and holds great potential for safe and efficient CNV therapy.


Subject(s)
Corneal Neovascularization , Prodrugs , Mice , Animals , Corneal Neovascularization/drug therapy , Prodrugs/therapeutic use , Endothelial Cells , Polyethylene Glycols/pharmacology , Polyethylene Glycols/chemistry , Polyethylene Glycols/therapeutic use , Dexamethasone/pharmacology , Dexamethasone/therapeutic use , Neovascularization, Pathologic/drug therapy
12.
Exp Eye Res ; 238: 109747, 2024 01.
Article in English | MEDLINE | ID: mdl-38072353

ABSTRACT

Corneal neovascularization (CNV) is a vision-threatening disease that is becoming a growing public health concern. While Yes-associated protein (YAP) plays a critical role in neovascular disease and allow for the sprouting angiogenesis. Verteporfin (VP) is a classical inhibitor of the YAP-TEAD complex, which is used for clinical treatment of neovascular macular degeneration through photodynamic therapy. The purpose of this study is to explore the effect of verteporfin (VP) on the inhibition of CNV and its potential mechanism. Rat CNV model were established by suturing in the central cornea and randomly divided into three groups (control, CNV and VP group). Neovascularization was observed by slit lamp to extend along the corneal limbus to the suture line. RNA-sequencing was used to reveal the related pathways on the CNV and the results revealed the vasculature development process and genes related with angiogenesis in CNV. In CNV group, we detected the nuclear translocation of YAP and the expression of CD31 in corneal neovascular endothelial cells through immunofluorescence. After the application of VP, the proliferation, migration and the tube formation of HUVECs were significantly inhibited. Furthermore, VP showed the CNV inhibition by tail vein injection without photoactivation. Then we found that the expression of phosphorylated YAP significantly decreased, and its downstream target protein connective tissue growth factor (CTGF) increased in the CNV group, while the expression was just opposite in other groups. Besides, both the expression of vascular endothelial growth factor receptor 2 (VEGFR2) and cofilin significantly increased in CNV group, and decreased after VP treatment. Therefore, we conclude that Verteporfin could significantly inhibited the CNV without photoactivation by regulating the activation of YAP.


Subject(s)
Choroidal Neovascularization , Corneal Neovascularization , Verteporfin , Animals , Rats , Choroidal Neovascularization/drug therapy , Choroidal Neovascularization/metabolism , Corneal Neovascularization/drug therapy , Endothelial Cells/metabolism , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Vascular Endothelial Growth Factor A/metabolism , Verteporfin/pharmacology , Verteporfin/therapeutic use
13.
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
14.
Adv Drug Deliv Rev ; 201: 115084, 2023 10.
Article in English | MEDLINE | ID: mdl-37689278

ABSTRACT

Ocular surface neovascularization and its resulting pathological changes significantly alter corneal refraction and obstruct the light path to the retina, and hence is a major cause of vision loss. Various factors such as infection, irritation, trauma, dry eye, and ocular surface surgery trigger neovascularization via angiogenesis and lymphangiogenesis dependent on VEGF-related and alternative mechanisms. Recent advances in antiangiogenic drugs, nanotechnology, gene therapy, surgical equipment and techniques, animal models, and drug delivery strategies have provided a range of novel therapeutic options for the treatment of ocular surface neovascularization. In this review article, we comprehensively discuss the etiology and mechanisms of corneal neovascularization and other types of ocular surface neovascularization, as well as emerging animal models and drug delivery strategies that facilitate its management.


Subject(s)
Corneal Neovascularization , Molecular Medicine , Animals , Neovascularization, Pathologic/drug therapy , Corneal Neovascularization/drug therapy , Corneal Neovascularization/pathology , Retina/pathology , Angiogenesis Inhibitors/therapeutic use
15.
Nanomedicine (Lond) ; 18(17): 1095-1108, 2023 07.
Article in English | MEDLINE | ID: mdl-37610088

ABSTRACT

Background: Corneal neovascularization is a sight-threatening disease. It can be treated using antiangiogenic and anti-inflammatory compounds. Therefore, atorvastatin (ATV) constitutes a suitable candidate to be administered topically. To attain suitable efficacy, ATV can be encapsulated into custom-developed nanocarriers such as peptide amphiphiles. Methods: Three peptide amphiphiles bearing one, two or four C16-alkyl groups (mC16-Tat47-57, dC16-Tat47-57 and qC16-Tat47-57) were synthesized, characterized and loaded with ATV. Drug release and ocular tolerance were assessed as well as anti-inflammatory and antiangiogenic properties. Results: ATV-qC16-Tat47-57 showed higher encapsulation efficiency than mC16-Tat47-57 and dC16-Tat47-57 and more defined nanostructures. ATV-qC16-Tat47-57 showed ATV prolonged release with suitable ocular tolerance. Moreover, ATV-qC16-Tat47-57 was antiangiogenic and prevented ocular inflammation. Conclusion: ATV-qC16-Tat47-57 constitutes a promising topical medication against corneal neovascularization.


Corneal neovascularization is an eye disease that affects over 1 million people every year and can lead to blindness. It is caused by inflammation and the unwanted formation of blood vessels in the eye. Current treatments for this disease are not fully effective. Atorvastatin (ATV) is one drug that has been partially successful at treating corneal neovascularization, but it does not stay in the eye long enough and does not mix well with the water-based environment of the eye. To overcome this, ATV was combined with three specially designed nanocarriers. These nanocarriers were peptides, short stretches of protein. They were designed to be amphiphilic, meaning that one section is hydrophilic (literally meaning 'water loving') and one section is hydrophobic ('water hating'). These peptide nanocarriers allowed ATV to stay in the water-based environment of the eye longer. The peptide with the most hydrophobic chains (qC16-Tat47-57) was able to carry more ATV than the other peptides and produced particles of a desired shape. ATV-qC16-Tat47-57 nanocarriers were found to release slowly. These nanocarriers were also found to prevent the development of new blood vessels on a membrane in a hen's egg used to mimic the eye. There was also no sign of irritation on this membrane or in the eyes of New Zealand rabbits. These results show ATV-qC16-Tat47-57 has a prolonged therapeutic effect, prevents the formation of new blood vessels and is tolerated in the eye. ATV-qC16-Tat47-57 is therefore potentially a more effective alternative to ATV treatment alone.


Subject(s)
Corneal Neovascularization , Humans , Corneal Neovascularization/drug therapy , Atorvastatin/pharmacology , Atorvastatin/therapeutic use , Eye , Drug Liberation , Peptides
16.
J Control Release ; 360: 818-830, 2023 08.
Article in English | MEDLINE | ID: mdl-37481212

ABSTRACT

Corneal neovascularization (CNV) badly damages the corneal transparency, resulting in visual disturbance and blindness. The frequent administration of glucocorticoid eye drops in clinical increases the possibility of side effects and reduces patient compliance. Considering CNV is often accompanied by an increase in ROS production, a ROS-responsive monomer 2-(methylthio)ethyl methacrylate was introduced into the matrix as a "gating switch". The prepared dexamethasone contact lenses (MCLs@Dex) showed a significant H2O2-responsive release for 168 h. To avoid corneal hypoxia and neovascularization caused by long-term wearing, high­oxygen-permeability fluorosiloxane materials were incorporated. The oxygen permeability of MCLs@Dex was 4 times that of commercially available hydrogel contact lenses and had ultra-low protein adsorption, which meets the requirements of long-term wearing. In vivo pharmacokinetic studies showed that MCLs@Dex increased the mean residence time by 19.7 times and bioavailability by 2.29 times compared with eye drops, validating the ROS response and sustained release properties. More importantly, MCLs@Dex had satisfactory effects on reducing inflammation and decreasing the related cytokines and oxidative stress levels, and demonstrated significant inhibition of neovascularization, with a suppression rate of 76.53% on the 14th day. This responsive drug delivery system provides a promising new method for the safe and effective treatment of ocular surface diseases.


Subject(s)
Contact Lenses , Corneal Neovascularization , Humans , Adult , Corneal Neovascularization/drug therapy , Hydrogen Peroxide , Reactive Oxygen Species , Inflammation/drug therapy , Oxygen , Ophthalmic Solutions
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.
Digit J Ophthalmol ; 29(1): 9-13, 2023.
Article in English | MEDLINE | ID: mdl-37101562

ABSTRACT

Corneal neovascularization is a determinant of corneal graft survival and preservation of immune privilege after keratoplasty. We report the outcomes in 2 patients with failed corneal grafts who underwent mitomycin C (MMC) intravascular chemoembolization (MICE) in the affected eye. A 30-year-old woman with failed penetrating keratoplasty (PK) in the right eye was started on prednisolone acetate eyedrops. Graft sutures were removed, and bevacizumab was injected subconjunctivally. The eye remained intermittently painful, and MICE was performed on the main feeding vessel, with regression of the vessels apparent within the first day following the procedure. The second case was a 40-year-old man who had a history of repaired penetrating injury in the left eye followed by failed PK. Prednisolone acetate eyedrops were initiated, and corneal sutures were removed. The patient failed to improve with three subconjunctival injections of bevacizumab. MICE was performed, but in this case neovascularization did not regress until 20 weeks post-procedure. MMC is thought to inhibit vascular endothelial cell proliferation, but its use in corneal injection is debated. In these cases, MICE was not associated with any concerning adverse events.


Subject(s)
Corneal Neovascularization , Corneal Transplantation , Humans , Bevacizumab , Corneal Neovascularization/drug therapy , Corneal Neovascularization/surgery , Mitomycin/therapeutic use , Angiogenesis Inhibitors/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Corneal Transplantation/methods , Cornea/surgery , Keratoplasty, Penetrating
19.
Int J Nanomedicine ; 18: 1413-1431, 2023.
Article in English | MEDLINE | ID: mdl-36992821

ABSTRACT

Background: Corneal neovascularization (NV) is a process of abnormal vessel growth into the transparent cornea from the limbus and can disturb the light passing through the cornea, resulting in vision loss or even blindness. The use of nanomedicine as an effective therapeutic formulation in ophthalmology has led to higher drug bioavailability and a slow drug release rate. In this research, we designed and explored the feasibility of a new nanomedicine, gp91 ds-tat (gp91) peptide-encapsulated gelatin nanoparticles (GNP-gp91), for inhibiting corneal angiogenesis. Methods: GNP-gp91 were prepared by a two-step desolvation method. The characterization and cytocompatibility of GNP-gp91 were analyzed. The inhibition effect of GNP-gp91 on HUVEC cell migration and tube formation was observed by an inverted microscope. The drug retention test in mouse cornea was observed by in vivo imaging system, fluorescence microscope, and DAPI/TAMRA staining. Finally, the therapeutic efficacy and evaluation of neovascularization-related factors were conducted through the in vivo corneal NV mice model via topical delivery. Results: The prepared GNP-gp91 had a nano-scale diameter (550.6 nm) with positive charge (21.7 mV) slow-release behavior (25%, 240hr). In vitro test revealed that GNP-gp91 enhanced the inhibition of cell migration and tube formation capacity via higher internalization of HUVEC. Topical administration (eyedrops) of the GNP-gp91 significantly prolongs the retention time (46%, 20 min) in the mouse cornea. In chemically burned corneal neovascularization models, corneal vessel area with a significant reduction in GNP-gp91 group (7.89%) was revealed when compared with PBS (33.99%) and gp91 (19.67%) treated groups via every two days dosing. Moreover, GNP-gp91 significantly reduced the concentration of Nox2, VEGF and MMP9 in NV's cornea. Conclusion: The nanomedicine, GNP-gp91, was successfully synthesized for ophthalmological application. These data suggest that GNP-gp91 contained eyedrops that not only have a longer retention time on the cornea but also can treat mice corneal NV effectively delivered in a low dosing frequency, GNP-gp91 eyedrops provides an alternative strategy for clinical ocular disease treatment in the culture.


Subject(s)
Corneal Neovascularization , Nanoparticles , Mice , Animals , Corneal Neovascularization/drug therapy , Gelatin/pharmacology , Ophthalmic Solutions/pharmacology , Cornea , Peptides/pharmacology , Nanoparticles/chemistry
20.
Int Ophthalmol ; 43(8): 2989-2997, 2023 Aug.
Article in English | MEDLINE | ID: mdl-36971928

ABSTRACT

PURPOSE: This study aimed to compare the efficacy of topical bevacizumab and motesanib in an experimental corneal neovascularization model, and find the most effective motesanib dose. MATERIALS AND METHODS: In experiments, 42 Wistar Albino rats were randomly divided into six groups (n = 7). Corneal cauterization was applied to all groups except the group 1. Group 1 did not receive any treatment. Topical dimethylsulfoxide was applied to sham group three times a day(tid). Topical bevacizumab drops (5 mg/ml) were applied to Group 3 tid. Topical motesanib drops with a dose of 2.5, 5, and 7.5 mg/ml were respectively applied in Groups 4, 5, and 6 tid. On the 8th day, corneal photographs of all rats were taken under general anesthesia, and the percentage of corneal neovascular area was calculated. VEGF-A mRNA, VEGFR-2 mRNA, miRNA-21, miRNA-27a, miRNA-31, miRNA-126, miRNA-184, and miRNA-204 were evaluated by the qRT-PCR method in corneas taken after decapitation. RESULTS: The percentage of corneal neovascularization areas and VEGF-A mRNA expression levels were decreased in all treatment groups compared to group 2 (p < 0.05). VEGFR-2 mRNA levels were found to be statistically significantly decreased in groups 4 and 6 compared to group 2 (p < 0.05). Statistically significant changes were detected in the expression levels of only miRNA-126 among all miRNAs. CONCLUSION: Motesanib with a dose of 7.5 mg/ml statistically significantly suppressed the VEGFR-2 mRNA level compared with other treatment doses and may be more effective than bevacizumab. Further, miRNA-126 can be used as a proangiogenic marker.


Subject(s)
Corneal Neovascularization , MicroRNAs , Rats , Animals , Bevacizumab/therapeutic use , Corneal Neovascularization/drug therapy , Angiogenesis Inhibitors , Vascular Endothelial Growth Factor Receptor-2/therapeutic use , Vascular Endothelial Growth Factor A , Antibodies, Monoclonal, Humanized , Rats, Wistar , Disease Models, Animal , Administration, Topical
SELECTION OF CITATIONS
SEARCH DETAIL
...