Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 117
Filter
Add more filters

Publication year range
1.
Int J Mol Sci ; 24(8)2023 Apr 17.
Article in English | MEDLINE | ID: mdl-37108535

ABSTRACT

Post-surgical scarring is a known cause of trabeculectomy failure. The aim of this study was to investigate the effectiveness of ranibizumab as an adjuvant anti-scarring agent in experimental trabeculectomy. Forty New Zealand white rabbits were randomised into four eye treatment groups: groups A (control), B (ranibizumab 0.5 mg/mL), C (mitomycin C [MMC] 0.4 mg/mL), and D (ranibizumab 0.5 mg/mL and MMC 0.4 mg/mL). Modified trabeculectomy was performed. Clinical parameters were assessed on post-operative days 1, 2, 3, 7, 14, and 21. Twenty rabbits were euthanised on day 7, and the other twenty were euthanised on day 21. Eye tissue samples were obtained from the rabbits and stained with haematoxylin and eosin (H&E). All treatment groups showed a significant difference in IOP reduction compared with group A (p < 0.05). Groups C and D showed a significant difference in bleb status on days 7 (p = 0.001) and 21 (p = 0.002) relative to group A. H&E staining showed significantly low fibrotic activity (p < 0.001) in group C on both days and inflammatory cell grade in group B on day 7 (p < 0.001). The grade for new vessel formation was significantly low in groups B and D on day 7 (p < 0.001) and in group D on day 21 (p = 0.007). Ranibizumab plays a role in reducing scarring, and a single application of the ranibizumab-MMC combination showed a moderate wound-modulating effect in the early post-operative phase.


Subject(s)
Trabeculectomy , Animals , Rabbits , Cicatrix/drug therapy , Intraocular Pressure , Mitomycin/therapeutic use , Ranibizumab/pharmacology
2.
Arch Gynecol Obstet ; 305(1): 267-274, 2022 01.
Article in English | MEDLINE | ID: mdl-34081204

ABSTRACT

PURPOSE: To investigate the histological efficacy of ranibizumab and zoledronic acid in an experimentally induced endometriosis model as compared with danazol, buserelin acetate and dienogest. METHODS: Endometrial implants were introduced in 52 female Wistar albino rats, which were then randomly divided into six groups. The animals were, respectively, given dienogest, danazol, buserelin acetate, zoledronic acid, ranibizumab and 0.9% NaCl. After 4 weeks, the volumes and histopathological properties of the implants were evaluated and the implants were excised completely at the third laparotomy. A histopathological scoring system was used to evaluate the preservation of epithelia. Endometrial explants were evaluated immunohistochemically. RESULTS: Among the groups, the histological score was significantly lower in the zoledronic acid and ranibizumab groups compared with the controls (p < 0.001). There were no significant differences regarding ellipsoidal volume levels between groups (p > 0.05). However, there was a statistically significant difference regarding cell numbers according to the degree of Bcl-2, NF-κB, and CD31 staining (p < 0.001). There was no statistically significant difference in Bcl-2, CD31, or NF-κB staining in the binary comparisons between the other groups (p > 0.05). For Bcl-2 staining, the staining rate of the group treated with zoledronic acid was significantly lower compared with the dienogest and danazol groups (p < 0.05). The staining rates of CD31 and NF-κB were significantly lower in the zoledronic acid and ranibizumab groups compared with the controls (p < 0.05). CONCLUSION: According to these results, zoledronic acid and ranibizumab may be putative candidates for the treatment of endometriosis.


Subject(s)
Endometriosis , Animals , Danazol/pharmacology , Danazol/therapeutic use , Endometriosis/drug therapy , Endometriosis/pathology , Female , Ranibizumab/pharmacology , Ranibizumab/therapeutic use , Rats , Rats, Wistar , Zoledronic Acid
3.
Microvasc Res ; 138: 104207, 2021 11.
Article in English | MEDLINE | ID: mdl-34119535

ABSTRACT

INTRODUCTION: Choroidal neovascularization (CNV) is the main pathological change of wet age-related macular degeneration. Anti-VEGF drugs are the most commonly used treatment for CNV. The biggest drawback of anti-VEGF drugs is the recurrence of CNV, which requires repeated therapy several times. Autophagy activation may be involved in reducing the therapeutic effect of anti-VEGF drugs. So, this study aims to elucidate the effect and mechanism of anti-VEGF drugs on endothelial autophagy and neovascularization in vitro. METHODS: RF/6A cells were randomly divided into five groups: The control group, hypoxia group (1% O2, 5% CO2, 94% N2), anti-VEGF group (group1: Ranibizumab 100 µg/ml; group2: Aflibercept, 400 µg/ml; group3: Conbercept, 100 µg/ml). Autophagy-related proteins were examined by Western blot. RFP-GFP-LC3 was used to detect autophagy and autophagic flow. Subsequently, we used autophagy inhibitors (3-MA or CQ) to inhibit Conbercept induced autophagy and to observe its effect on angiogenesis in vitro. Proliferation, migration, and tube formation of endothelial cells can be used to study neovascularization in vitro. In this research, the CCK-8 assay was used to detect cell proliferation. Cell migration and tube formation were assessed by wound assay and matrix method, respectively. Flow cytometry and Tunel were used to detect cell apoptosis. Finally, the mechanism of Conbercept activated autophagy was studied. Western blot was used to detect the expression of p53 and DRAM (damage-regulated autophagy modulator), upstream activators of autophagy. RESULTS: The protein levels of Beclin-1 and LC3-2/1 in Ranibizumab and Conbercept groups were significantly higher than in the hypoxia group(P < 0.05). While the expression of P62 was decreased (P < 0.05). The autophagic flux was showed the same results. However, Aflibercept showed the opposite effect on autophagy. Compared with the Conbercept group, autophagy inhibitor 3-MA or CQ can further inhibit cell proliferation and promotes cell apoptosis (P < 0.05). Conbercept significantly inhibited cell migration compared with the hypoxia group (633.08 ± 72.52 vs. 546.33 ± 24.61), while the autophagy inhibitor group (3-MA or CQ) had a more obvious inhibition effect (309.75 ± 86.36 and 263.33 ± 68.67) (P < 0.05). For tube formation, the number of tube formation was decreased significantly in the Conbercept group (32.00 ± 2.00) compared to the hypoxia group (39.00 ± 1.53) and even further reduced in 3-MA or CQ group (24.00 ± 3.61, 20.00 ± 2.65). The length of master segments in the hypoxia group was 15,668.00 ± 894.11. It was decreased in Conbercept (13,885.34 ± 730.03). In 3-MA or CQ group, the length of master segments dropped further (11,997.00 ± 433.66, 10,617.67 ± 543.21). Compare with the hypoxia group, the expression P53 and DRAM were increased in the Conbercept group (P < 0.05). Autophagy-related proteins LC-3, Beclin-1, and DRAM were inhibited by P53 inhibitor Pifithrin-α (PFTα) (P < 0.05). CONCLUSION: Ranibizumab and Conbercept can trigger the autophagy of vascular endothelial cells while Aflibercept can inhibit it. The combination of Conbercept and autophagy inhibitor can significantly inhibit the formation of angiogenesis in vitro. The mechanism of autophagy activation is related to the activation of the p53/DRAM pathway.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Autophagy/drug effects , Choroidal Neovascularization/drug therapy , Endothelial Cells/drug effects , Neovascularization, Physiologic/drug effects , Ranibizumab/pharmacology , Recombinant Fusion Proteins/pharmacology , Vascular Endothelial Growth Factors/antagonists & inhibitors , Animals , Apoptosis/drug effects , Cell Line , Cell Movement/drug effects , Cell Proliferation/drug effects , Choroidal Neovascularization/metabolism , Choroidal Neovascularization/pathology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Macaca mulatta , Receptors, Vascular Endothelial Growth Factor , Signal Transduction , Tumor Suppressor Protein p53/metabolism , Vascular Endothelial Growth Factors/metabolism
4.
Int J Mol Sci ; 22(4)2021 Feb 22.
Article in English | MEDLINE | ID: mdl-33671690

ABSTRACT

Proliferative diabetic retinopathy (PDR), a major complication of diabetes mellitus, results from an inflammation-sustained interplay among endothelial cells, neurons, and glia. Even though anti-vascular endothelial growth factor (VEGF) interventions represent the therapeutic option for PDR, they are only partially efficacious. In PDR, Müller cells undergo reactive gliosis, produce inflammatory cytokines/chemokines, and contribute to scar formation and retinal neovascularization. However, the impact of anti-VEGF interventions on Müller cell activation has not been fully elucidated. Here, we show that treatment of MIO-M1 Müller cells with vitreous obtained from PDR patients stimulates cell proliferation and motility, and activates various intracellular signaling pathways. This leads to cytokine/chemokine upregulation, a response that was not mimicked by treatment with recombinant VEGF nor inhibited by the anti-VEGF drug ranibizumab. In contrast, fibroblast growth factor-2 (FGF2) induced a significant overexpression of various cytokines/chemokines in MIO-M1 cells. In addition, the FGF receptor tyrosine kinase inhibitor BGJ398, the pan-FGF trap NSC12, the heparin-binding protein antagonist N-tert-butyloxycarbonyl-Phe-Leu-Phe-Leu-Phe Boc2, and the anti-inflammatory hydrocortisone all inhibited Müller cell activation mediated by PDR vitreous. These findings point to a role for various modulators beside VEGF in Müller cell activation and pave the way to the search for novel therapeutic strategies in PDR.


Subject(s)
Diabetic Retinopathy/pathology , Ependymoglial Cells/pathology , Vascular Endothelial Growth Factor A/metabolism , Aged , Cell Proliferation , Cells, Cultured , Cholesterol/analogs & derivatives , Cholesterol/pharmacology , Diabetic Retinopathy/surgery , Ependymoglial Cells/drug effects , Ependymoglial Cells/physiology , Female , Fibroblast Growth Factor 2/pharmacology , Gene Expression Regulation , Humans , Hydrocortisone/pharmacology , Inflammation Mediators/metabolism , Male , Middle Aged , Phenylurea Compounds/pharmacology , Pyrimidines/pharmacology , Ranibizumab/pharmacology , Receptors, Vascular Endothelial Growth Factor/genetics , Receptors, Vascular Endothelial Growth Factor/metabolism , Vascular Endothelial Growth Factor A/pharmacology , Vitrectomy
5.
Int J Mol Sci ; 22(8)2021 Apr 09.
Article in English | MEDLINE | ID: mdl-33918777

ABSTRACT

It has been shown previously that a novel tetrapeptide, Arg-Leu-Tyr-Glu (RLYE), derived from human plasminogen inhibits vascular endothelial growth factor (VEGF)-induced angiogenesis, suppresses choroidal neovascularization in mice by an inhibition of VEGF receptor-2 (VEGFR-2) specific signaling pathway. In this study, we report that a modified tetrapeptide (Ac-RLYE) showed improved anti-choroidal neovascularization (CNV) efficacy in a number of animal models of neovascular age-related macular degeneration (AMD) which include rat, rabbit, and minipig. The preventive and therapeutic in vivo efficacy of Ac-RLYE via following intravitreal administration was determined to be either similar or superior to that of ranibizumab and aflibercept. Assessment of the intraocular pharmacokinetic and toxicokinetic properties of Ac-RLYE in rabbits demonstrated that it rapidly reached the retina with minimal systemic exposure after a single intravitreal dose, and it did not accumulate in plasma during repetitive dosing (bi-weekly for 14 weeks). Our results suggested that Ac-RLYE has a great potential for an alternative therapeutics for neovascular (wet) AMD. Since the amino acids in human VEGFR-2 targeted by Ac-RLYE are conserved among the animals employed in this study, the therapeutic efficacies of Ac-RLYE evaluated in those animals are predicted to be observed in human patients suffering from retinal degenerative diseases.


Subject(s)
Macular Degeneration/etiology , Macular Degeneration/metabolism , Oligopeptides/pharmacology , Acetylation , Animals , Choroidal Neovascularization/drug therapy , Choroidal Neovascularization/metabolism , Choroidal Neovascularization/pathology , Disease Models, Animal , Disease Susceptibility , Fluorescein Angiography , Humans , Macular Degeneration/diagnosis , Macular Degeneration/drug therapy , Male , Mice , Oligopeptides/chemistry , Promoter Regions, Genetic , Rabbits , Ranibizumab/pharmacology , Receptors, Vascular Endothelial Growth Factor , Recombinant Fusion Proteins/pharmacology , Retina/metabolism , Retina/pathology , Retinal Neovascularization/drug therapy , Retinal Neovascularization/metabolism , Retinal Neovascularization/pathology , Swine , Treatment Outcome , Vascular Endothelial Growth Factor A/chemistry , Vascular Endothelial Growth Factor A/metabolism
6.
Cell Physiol Biochem ; 54(2): 161-179, 2020 Feb 12.
Article in English | MEDLINE | ID: mdl-32045141

ABSTRACT

BACKGROUND/AIMS: We performed co-culture experiments between human RPE cells (ARPE-19) and human umbilical vascular endothelial cells (HUVEC) in order to evaluate how anti-VEGF drugs could affect NO release, mitochondrial function, the oxidative status, proliferation and migration of RPE cells through modulation of their cross talk with vascular endothelial cells. METHODS: The co-culture HUVEC/RPE, was exposed to Ranibizumab/Aflibercept in the absence/presence of the NO synthase (NOS) inhibitor, the phosphatidylinositol 3'-kinase (PI3K), the extracellular-signal-regulated kinases 1/2 (ERK1/2) and the p38 mitogen-activated protein kinase (p38 MAPK) blockers. Specific kits were used for cell viability, mitochondrial membrane potential, NO, ROS and GSH production. Western blot was performed for apoptosis markers, NOS isoforms, and others kinases detection. Cell migration was analyzed by scratch assay, whereas cell proliferation and cell cycle through xCELLigence and flow cytometry. RESULTS: In RPE cells co-cultured with HUVEC in physiological conditions, Aflibercept/Ranibizumab increased NO release in a dose and time-dependent way. Opposite results were obtained in peroxidative conditions. Both anti-VEGF agents were able to prevent the fall of cell viability and mitochondrial membrane potential, an effect which was reduced by various inhibitors, and increased cell migration. Aflibercept/Ranibizumab counteracted the changes of apoptosis markers, NOS expression/activation, PI3K and ERK1/2 activation caused by peroxidation. These results were confirmed by cell cycle analysis. CONCLUSION: This study has shown new mechanisms at the basis of protective effects elicited by Aflibercept/Ranibizumab in RPE cells. HUVEC stimulated with Aflibercept/Ranibizumab, could release some paracrine factors that can modulate the RPE cells response in both physiologic and peroxidative conditions.


Subject(s)
Cell Communication/drug effects , Ranibizumab/pharmacology , Recombinant Fusion Proteins/pharmacology , Cell Cycle Checkpoints/drug effects , Cell Movement/drug effects , Cell Survival/drug effects , Coculture Techniques , Glutathione/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Membrane Potential, Mitochondrial/drug effects , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide Synthase Type III/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , Receptors, Vascular Endothelial Growth Factor , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/metabolism
7.
FASEB J ; 33(3): 3451-3464, 2019 03.
Article in English | MEDLINE | ID: mdl-30475644

ABSTRACT

Retinopathy of prematurity is a vision-threatening condition, and therapies based on antagonizing VEGF may elicit serious side effects in premature infants. Mechanisms of retinal angiogenesis, particularly the signaling pathways independent of VEGF, remain elusive. The goals of our study were to explore TLR4-mediated signaling pathways in human retinal microvascular endothelial cells (HRMECs) and to examine the effects of TLR4 antagonists in models of oxygen-induced retinopathy (OIR). Our results show that intravitreal injection of the TLR4 antagonist TAK-242 reduced areas of nonperfusion, inhibited aberrant angiogenesis, and improved vascular density in the retina of OIR mice. The effects were further potentiated by the anti-VEGF antibody ranibizumab. In cultured HRMECs, the TLR4 agonist LPS up-regulated TLR4/MAPKK kinase kinase 4 (MAP4K4) signaling, and promoted cell proliferation and migration, and reduced barrier functions of the cells. Down-regulation of MAP4K4 in HRMECs abolished the proangiogenic effects by LPS. Our data suggest that the TLR4-MAP4K4 pathway can regulate retinal neovascularization via mechanisms independent of VEGF.-Chen, W., Zhang, J., Zhang, P., Hu, F., Jiang, T., Gu, J., Chang, Q. Role of TLR4-MAP4K4 signaling pathway in models of oxygen-induced retinopathy.


Subject(s)
Oxygen/metabolism , Protein Serine-Threonine Kinases/metabolism , Retinal Diseases/metabolism , Signal Transduction/physiology , Toll-Like Receptor 4/metabolism , Animals , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Disease Models, Animal , Down-Regulation/drug effects , Down-Regulation/physiology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Female , Humans , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Pregnancy , Ranibizumab/pharmacology , Retina/drug effects , Retina/metabolism , Retinal Diseases/drug therapy , Retinal Neovascularization/metabolism , Retinal Vessels/drug effects , Retinal Vessels/metabolism , Signal Transduction/drug effects , Up-Regulation/drug effects , Up-Regulation/physiology , Vascular Endothelial Growth Factor A/metabolism , NF-kappaB-Inducing Kinase
8.
J Cell Physiol ; 234(10): 18297-18307, 2019 08.
Article in English | MEDLINE | ID: mdl-30891770

ABSTRACT

This study sought to evaluate the prospective role exerted by vascular endothelial growth factor (VEGF) in the modulation of nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF) signalling pathways in the rabbit retina. To reach this aim, the anti-VEGF agents aflibercept and ranibizumab were used as a pharmacological approach to evaluate the putative consequences elicited by VEGF inhibition on neurotrophin signalling. VEGF inhibition determined a marked imbalance in proneurotrophin expression, a significant reduction in TrkA and TrkB phosphorylation states and a decrease in the pan-neurotrophin receptor p75. Importantly, VEGF blockade also caused a strong increase in cleaved caspase-3, beclin-1 and lipidated LC3. The effects were more pronounced in the aflibercept group when compared with ranibizumab-treated rabbits, particularly 1 week after injection. This study demonstrates that VEGF exerts pivotal physiological roles in regulating NGF and BDNF pathways in the retina, as its inhibition by anti-VEGF agents deeply impacts neurotrophin homeostasis. These events are accompanied by a sustained induction of apoptotic and autophagic markers, suggesting that anti-VEGF-dependent impairments in neurotrophin signalling could be responsible for the activation of retinal cell death pathways.


Subject(s)
Autophagy/physiology , Caspase 3/metabolism , Nerve Growth Factors/metabolism , Signal Transduction/physiology , Vascular Endothelial Growth Factor A/metabolism , Angiogenesis Inhibitors/pharmacology , Animals , Autophagy/drug effects , Beclin-1/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Male , Nerve Growth Factor/metabolism , Neurons/drug effects , Neurons/metabolism , Prospective Studies , Rabbits , Ranibizumab/pharmacology , Receptor, Nerve Growth Factor/metabolism , Receptors, Vascular Endothelial Growth Factor , Recombinant Fusion Proteins/pharmacology , Retina/drug effects , Retina/metabolism , Signal Transduction/drug effects
9.
J Cell Biochem ; 120(10): 17912-17925, 2019 10.
Article in English | MEDLINE | ID: mdl-31135072

ABSTRACT

OBJECTIVE: To investigate the regulation of special protein 1 (SP1) and hypoxia-inducible factor-1α (HIF1α) on human microvascular endothelial cells (HMEC-1) under hypoxic conditions. METHODS: The expression of SP1 and HIF1α under normoxia and hypoxic conditions were assessed by Western blot. SP1 and HIF1α were knocked down by small interfering RNA (siRNA) under hypoxic conditions. The proliferation, migration, and invasion of HMEC-1 were measured by cell counting kit 8, 5-ethynyl-2'-deoxyuridine and Transwell coculture system. Western blot analysis and Immunofluorescence were carried out to study the mechanisms of simultaneously inhibiting the adenosine triphosphatase (CD39), 5'-nucleotidase (CD73), adenosine, and vascular endothelial growth factor (VEGF). We compared the inhibitory effects between groups concurrently interfering SP1, HIF-1α, and ranibizumab under hypoxic conditions. RESULTS: Under hypoxic conditions, the protein expression of SP1 and HIF1α was increased in HMEC-1, contrarily, SP1 siRNA and HIF1α siRNA downregulated the expression. Simultaneous inhibition of SP1 and HIF1α demonstrated a much greater restraint of proliferation, migration, and invasion characteristics on HMEC-1 than respectively knocking down SP1 or HIF1α and anti-VEGF drugs (0.5 mg/mL ranibizumab) (siRNA and the VEGF inhibitor were applied separately in different groups). Meanwhile, simultaneous inhibition of SP1 and HIF1α effectively reduced the expression of CD39, CD73, adenosine, and VEGF on HMEC-1 under hypoxic conditions. CONCLUSIONS: Our study demonstrated that both SP1 and HIF1α played important roles in HMEC-1 under hypoxia condition. Simultaneous inhibition of SP1 and HIF1α effectively decreased the activity of HMEC-1 under hypoxic conditions through the CD39-CD73-adenosine and VEGF angiogenesis pathways. Our study may provide a new approach to the treatment of retinal neovascular diseases.


Subject(s)
Cell Movement , Endothelial Cells/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Microvessels/pathology , Sp1 Transcription Factor/metabolism , Adenosine/pharmacology , Antigens, CD/metabolism , Cell Hypoxia/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Gene Silencing/drug effects , Humans , RNA, Small Interfering/metabolism , Ranibizumab/pharmacology , Up-Regulation/drug effects
10.
Exp Eye Res ; 181: 98-104, 2019 04.
Article in English | MEDLINE | ID: mdl-30615884

ABSTRACT

BACKGROUND: Human retinal microvascular endothelial cells (HRMVECs) are involved in the pathogenesis of retinopathy of prematurity. In this study, the microRNA (miRNA) expression profiles of HRMVECs were investigated under resting conditions, angiogenic stimulation (VEGF treatment) and anti-VEGF treatment. MATERIALS AND METHODS: The miRNA profiles of HRMVECs under resting and angiogenic conditions (VEGF treatment), as well as after addition of aflibercept, bevacizumab or ranibizumab were evaluated by analyzing the transcriptome of small non-coding RNAs. Differentially expressed miRNAs were validated using qPCR and classified using Gene Ontology enrichment analysis. RESULTS: Ten miRNAs were found to be significantly changed more than 2-fold. Seven of these miRNAs were changed between resting conditions and angiogenic stimulation. Four of these miRNAs (miR-139-5p/-3p and miR-335-5p/-3p) were validated by qPCR in independent experiments and were found to be associated with angiogenesis and cell migration in Gene Ontology analysis. In addition, analysis of the most abundant miRNAs in the HRMVEC miRNome (representing at least 1% of the miRNome) was conducted and identified miR-21-5p, miR-29a-3p, miR-100-5p and miR-126-5p/-3p to be differently expressed by at least 15% between resting conditions and angiogenic conditions. These miRNAs were found to be associated with apoptotic signaling, regulation of kinase activity, intracellular signal transduction, cell surface receptor signaling and positive regulation of cell differentiation in Gene Ontology analysis. No differentially regulated miRNAs between angiogenic stimulation and angiogenic stimulation plus anti-VEGF treatment were identified. CONCLUSION: In this study we characterized the miRNA profile of HRMVECs under resting, angiogenic and anti-angiogenic conditions and identified several miRNAs of potential pathophysiologic importance for angioproliferative retinal diseases. Our results have implications for possible miRNA-targeted angiomodulatory approaches in diseases like diabetic retinopathy or retinopathy of prematurity.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Endothelial Cells/drug effects , MicroRNAs/drug effects , Retina/cytology , Vascular Endothelial Growth Factor A/pharmacology , Bevacizumab/pharmacology , Cell Differentiation/drug effects , Endothelial Cells/metabolism , Humans , MicroRNAs/metabolism , Ranibizumab/pharmacology , Receptors, Vascular Endothelial Growth Factor , Recombinant Fusion Proteins/pharmacology , Retinopathy of Prematurity , Vascular Endothelial Growth Factor A/antagonists & inhibitors
11.
Ophthalmologica ; 241(3): 137-142, 2019.
Article in English | MEDLINE | ID: mdl-30001546

ABSTRACT

AIM: Treatment of exudative age-related macular degeneration by using vascular endothelial growth factor (VEGF) antagonists is the gold standard today. So far, several bioactive molecules have been approved for therapeutic use. In this study, we investigate the effects of ranibizumab (Lucentis®), bevacizumab (Avastin®), and aflibercept (Eylea®) on primary human retinal pigment epithelial (hRPE) cells in vitro. METHODS: hRPE cells were prepared from donor eyes and cultured under standard culture conditions. Scleral fibroblasts also prepared from donor tissue served as physiological controls. The impact of the anti-VEGF molecules on cell viability was investigated with the trypan blue exclusion assay, whereas proliferation was measured using the MTT assay. Biological activity of the molecules was quantified in a VEGF-enzyme-linked immunosorbent assay (ELISA). RESULTS: All tested substances were biologically active in vitro. They displayed no cytotoxicity on RPE cells or scleral fibroblasts. However, proliferation of RPE cells was significantly decreased after treatment with ranibizumab or bevacizumab but not with aflibercept. CONCLUSIONS: The humanized antibodies (fragments) interfered specifically with the RPE cells. The thereby measured inhibition of cell proliferation may indicate possible side effects on the physiology of RPE cells.


Subject(s)
Bevacizumab/pharmacology , Macular Degeneration/drug therapy , Ranibizumab/pharmacology , Recombinant Fusion Proteins/pharmacology , Retinal Pigment Epithelium/pathology , Angiogenesis Inhibitors , Cell Proliferation , Cell Survival , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Humans , Macular Degeneration/pathology , Receptors, Vascular Endothelial Growth Factor , Retinal Pigment Epithelium/drug effects , Vascular Endothelial Growth Factor A/antagonists & inhibitors
12.
Clin Exp Ophthalmol ; 47(8): 1074-1081, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31265210

ABSTRACT

BACKGROUND: Retinal pigment epithelium (RPE) is known to secrete factors important for retinal homeostasis. How this secretome changes in diabetic eyes treated with anti-vascular endothelial growth factor (VEGF) inhibitors is unclear. METHODS: Diabetic conditions were simulated in vitro using ARPE-19 cell-line culture, with high glucose (25 mM) culture media, and hypoxia was chemically induced using cobalt chloride. Stress was assessed using cell viability assays as well as Western blots and enzyme-linked immunosorbent assay (ELISA) for production of HIF-1a and VEGF-A. Production of neurotrophic factors was quantified once conditions were established using ELISA under stress with and without the addition of VEGF inhibitors. Changes were analysed with one-way ANOVA. RESULTS: Hypoxia downregulated pigment epithelium-derived factor (PEDF) expression. The addition of bevacizumab, ranibizumab and aflibercept in normoxic conditions all led to a significant downregulation of PEDF. Glucose concentration had no effect on secretion of PEDF. Brain-derived neurotrophic factor (BDNF) secretion was downregulated in high glucose and was upregulated in hypoxia. Placental growth factor (PlGF) secretion by ARPE-19 was undetectable by ELISA. CONCLUSIONS: We found that hypoxia, high glucose or VEGF inhibitors affected secretion of neurotrophic factors. This variation under different conditions may influence neuron and photoreceptor survival in the diabetic state and VEGF inhibitor treated eyes.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Glucose/pharmacology , Hyperglycemia/pathology , Hypoxia/pathology , Nerve Growth Factors/metabolism , Retinal Pigment Epithelium/drug effects , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Bevacizumab/pharmacology , Blotting, Western , Brain-Derived Neurotrophic Factor/metabolism , Cell Line , Cell Survival , Enzyme-Linked Immunosorbent Assay , Eye Proteins/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Membrane Proteins/metabolism , Ranibizumab/pharmacology , Receptors, Vascular Endothelial Growth Factor , Recombinant Fusion Proteins/pharmacology , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Serpins/metabolism , Vascular Endothelial Growth Factor A/metabolism
13.
Biochem Biophys Res Commun ; 503(4): 2742-2748, 2018 09 18.
Article in English | MEDLINE | ID: mdl-30103950

ABSTRACT

Choroidal neovascularization (CNV) is a type of wet age-related macular degeneration (AMD) which is a major cause of blindness in elder patients. Tumor necrosis factor receptor-associated factor 6 (TRAF6) promotes tumor angiogenesis via upregulating the expression of hypoxia-inducible factor 1α (HIF-1α) and vascular endothelial growth factor (VEGF). Additionally, TRAF6 facilitates the inflammatory response in macrophages and microglia. Here, using mouse laser-induced CNV model and TRAF6 siRNA, the study shows that TRAF6 is a critical player in CNV. The expression of TRAF6, HIF-1α, and VEGF increased in the model. TFAF6 siRNA intravitreal (IVT) injection inhibited CNV formation, as well as expression of HIF-1α and VEGF, activation of macrophages and microglia. Together, our data suggest that TFAF6 inhibition reduces CNV formation via down-regulating expression of HIF-1α and VEGF and activation of macrophages and microglia, demonstrating the unique advantages of TRAF6 inhibition in the alleviation of AMD.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Choroidal Neovascularization/prevention & control , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Ranibizumab/pharmacology , TNF Receptor-Associated Factor 6/genetics , Vascular Endothelial Growth Factor A/genetics , Animals , Choroidal Neovascularization/genetics , Choroidal Neovascularization/metabolism , Choroidal Neovascularization/pathology , Disease Models, Animal , Gene Expression Regulation , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lasers , Light Coagulation , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , TNF Receptor-Associated Factor 6/antagonists & inhibitors , TNF Receptor-Associated Factor 6/metabolism , Vascular Endothelial Growth Factor A/metabolism
14.
Mol Vis ; 24: 83-93, 2018.
Article in English | MEDLINE | ID: mdl-29422766

ABSTRACT

Purpose: Age-related macular degeneration (AMD) is the leading cause of central visual loss among patients over the age of 55 years worldwide. Neovascular-type AMD (nAMD) accounts for approximately 10% of patients with AMD and is characterized by choroidal neovascularization (CNV). The proliferation of choroidal endothelial cells (CECs) is one important step in the formation of new vessels. Transcriptional coactivator Yes-associated protein (YAP) can promote the proliferation of multiple cancer cells, corneal endothelial cells, and vascular smooth muscle cells, which participate in angiogenesis. This study intends to reveal the expression and functions of YAP during the CNV process. Methods: In the study, a mouse CNV model was generated by laser photocoagulation. YAP expression was detected with western blotting and immunohistochemistry. YAP siRNA and ranibizumab, a VEGF monoclonal antibody, were injected intravitreally in CNV mice. The YAP and VEGF expression levels after injection were detected with western blotting. The incidence and leakage area of CNV were measured with fundus fluorescein angiography, choroidal flat mounting, and hematoxylin and eosin (HE) staining. Immunofluorescent double staining was used to detect YAP cellular localization with CD31 (an endothelial cell marker) antibody. Proliferating cell nuclear antigen (PCNA) expression in CNV mice without or with YAP siRNA intravitreal injection and the colocalization of PCNA and CD31 were measured with western blotting and immunofluorescent double staining, respectively. Results: YAP expression increased following laser exposure, in accordance with vascular endothelial growth factor (VEGF) expression. YAP siRNA and ranibizumab decreased VEGF expression and the incidence and leakage area of CNV. YAP was localized in the vascular endothelium within the CNV site. Additionally, after laser exposure, YAP siRNA inhibited the increased expression of PCNA, which was colocalized with endothelial cells. Conclusions: This study showed that YAP upregulation promoted CNV formation by upregulating the proliferation of endothelial cells, providing evidence for the molecular mechanisms of CNV and suggesting a novel molecular target for nAMD treatment.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Choroid/metabolism , Choroidal Neovascularization/genetics , Endothelial Cells/metabolism , Macular Degeneration/genetics , Phosphoproteins/genetics , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Cycle Proteins , Cell Proliferation , Choroid/pathology , Choroidal Neovascularization/etiology , Choroidal Neovascularization/metabolism , Choroidal Neovascularization/pathology , Disease Models, Animal , Endothelial Cells/pathology , Gene Expression Regulation , Humans , Intravitreal Injections , Light Coagulation/adverse effects , Macular Degeneration/metabolism , Macular Degeneration/pathology , Mice , Mice, Inbred C57BL , Phosphoproteins/antagonists & inhibitors , Phosphoproteins/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/genetics , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Ranibizumab/pharmacology , Signal Transduction , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , YAP-Signaling Proteins
15.
Toxicol Appl Pharmacol ; 356: 65-75, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30025850

ABSTRACT

Although the precise etiology of Rheumatoid arthritis (RA) remains obscure, heightened immune response is thought to play a vital role in provoking joint inflammation and bone erosion. This study aims at comparatively evaluating the effects of two monoclonal antibodies Ranibizumab (RANI) as anti-VEGF antibody and Tocilizumab (TCZ) as interleukin-6 receptor (IL-6R) antagonist, against adjuvant induced arthritis in rats. CFA-induced arthritic rats were treated for three consecutive weeks with Methotrexate (MTX), TCZ and RANI monotherapy. Clinical assessment of RA, bone erosion, inflammatory, angiogenic and apoptotic markers were determined to assess the anti-arthritic effect. Liver enzymes and histopathological examination of liver and spleen were assessed to evaluate the toxicity profile of the tested therapeutic agents. MTX, TCZ and RANI monotherapy significantly enhanced the anti-arthritic parameters in comparison with the Complete Freund's Adjuvant (CFA)-induced arthritic rats through significant reduction of ankle and paw swelling. Also, they significantly reduced inflammatory, angiogenic and apoptotic markers. Importantly, Ranibizumab showed better effect than the standard anti-rheumatic drugs Methotrexate (MTX) or Tocilizumab (TCZ) in bone protection and cartilage health; hence proves to be a promising new therapeutic agent for RA.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Antirheumatic Agents/therapeutic use , Arthritis, Experimental/drug therapy , Ranibizumab/therapeutic use , Receptors, Interleukin-6/antagonists & inhibitors , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Angiogenesis Inhibitors/pharmacology , Animals , Antibodies, Monoclonal, Humanized/pharmacology , Antirheumatic Agents/pharmacology , Apoptosis/drug effects , Arthritis, Experimental/pathology , Bone and Bones/pathology , Cartilage/pathology , Inflammation/pathology , Inflammation/prevention & control , Liver/enzymology , Liver/pathology , Male , Methotrexate/pharmacology , Ranibizumab/pharmacology , Rats , Spleen/pathology
16.
Exp Eye Res ; 177: 112-116, 2018 12.
Article in English | MEDLINE | ID: mdl-30071215

ABSTRACT

PURPOSE: A critical biological function of retina pigment epithelium (RPE) cells is phagocytosis of photoreceptor outer segment (POS) disc membranes. Mitochondrial damage and dysfunction are associated with RPE cells of age-related macular degeneration (AMD) retinas. In this study, we use a transmitochondrial cybrid model to compare the phagocytic properties of RPE cells that contain AMD mitochondria versus age-matched normal mitochondria and their response to treatment with anti-vascular endothelial growth factor (VEGF) drugs: bevacizumab, ranibizumab, and aflibercept. METHODS: Cybrids, which are cell lines with identical nuclei but mitochondria (mt) from different subjects, are created by fusing mtDNA depleted ARPE-19 cells with platelets from AMD or age-matched normal patients. AMD (n = 5) and normal (n = 5) cybrids were treated with 1 µm fluorescent latex beads (1.52 × 107 beads/mL) and either 2.09 µM of bevacizumab, 2.59 µM of ranibizumab, or 5.16 µM of aflibercept. These doses of anti-VEGF drugs are equivalent to intravitreal injections given to AMD patients with choroidal neovascularization. Flow cytometry was performed using the ImageStreamX Mark II to assess phagocytic bead-uptake. The average fold values for bead-uptake and SEM were calculated using GraphPad Prism software. RESULTS: Normal cybrids showed decreased bead-uptake with a fold value of 0.65 ±â€¯0.103 (p = 0.01) after treatment with bevacizumab, 0.80 ±â€¯0.034 (p = 0.0003) with ranibizumab, and 0.81 ±â€¯0.053 (p = 0.007) with aflibercept compared to the untreated normal cybrids (baseline fold of 1). The bevacizumab-treated, ranibizumab-treated, and aflibercept-treated AMD cybrids had decreased bead-uptake with a fold value of 0.71 ±â€¯0.061 (p = 0.001), 0.70 ±â€¯0.101 (p = 0.02), and 0.74 ±â€¯0.125 (p = 0.07), respectively, compared to the untreated AMD cybrids (baseline fold of 1). CONCLUSIONS: Our initial findings showed that when treated with bevacizumab and ranibizumab, both AMD cybrids and age-matched normal cybrids had a significant decrease in bead-uptake. A similar decrease in bead-uptake was found in normal cybrids treated with aflibercept and while the AMD values trended lower, they were not significant. This data suggests that anti-VEGF drugs can cause loss of phagocytic function.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Bevacizumab/pharmacology , Epithelial Cells/drug effects , Macular Degeneration/drug therapy , Mitochondria/physiology , Phagocytosis/drug effects , Ranibizumab/pharmacology , Recombinant Fusion Proteins/pharmacology , Retinal Pigment Epithelium/cytology , Cells, Cultured , Humans , Receptors, Vascular Endothelial Growth Factor
17.
Exp Eye Res ; 171: 1-11, 2018 06.
Article in English | MEDLINE | ID: mdl-29522724

ABSTRACT

Anti-Vascular Endothelial Growth Factor (VEGF) therapy is given repeatedly for an extended period of time to patients when treated for age-related macular degeneration. While short-term effects of anti-VEGF agents on retinal pigment epithelial (RPE) cells have been investigated, the effects of long-term and repeated treatment on these cells are scarce. In this study, we have investigated the effects of anti-VEGF treatment after long-term, repeated treatment on cell aging and morphology. The experiments were conducted in primary porcine RPE cells passage one and two. Cells were treated with 125 µg/ml bevacizumab, ranibizumab, aflibercept or rituximab once a week for 1 day, 4 days, 7 days, 4 weeks and 12 weeks. Cell survival was evaluated with 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide tetrazolium (MTT) and trypan blue exclusion assay. Activity of ß-galactosidase was assessed in a commercially available assay. Influence of these compounds was investigated on the expression of cathepsin D and amyloid ß, and the expression and phosphorylation of mechanistic target of rapamycin (mTOR), all proteins involved in senescence and aging, in Western blot. The secretion of Pigment Epithelium Derived Factor (PEDF) and Transforming Growth Factor (TGF)-ß was investigated in Enzyme-linked Immunosorbent Assay (ELISA). The cellular morphology was investigated with electron microscopy, investigating the number and area of mitochondria and autophagosomes. Statistical analysis was conducted using a mixed linear model. Weekly treatment up to 12 weeks displayed no toxic effects on RPE cells in any of the substances tested. Ranibizumab showed a significant increase in ß-galactosidase signal on day 4 (p < 0.05) and 7 (p < 0.05) after treatment. In long-term, however, ranibizumab displayed no significant difference to untreated cells. Bevacizumab displayed a significant reduction of the ß-galactosidase signal after 12 weeks (p < 0.05). Aflibercept significantly decreased ß-galactosidase after 1 day (p < 0.01) and 12 weeks (p < 0.05). Rituximab and bevacizumab also decreased ß-galactosidase signal after 12 weeks (p < 0.05). The expression of mTOR, phospho-mTOR, amyloid ß and cathepsin D was not significantly altered by any of the compounds tested. RPE cells secreted considerate amounts of TGF-ß. Bevacizumab treated cells showed significantly lower TGF-ß secretion than ranibizumab and rituximab (p < 0.05). In contrast, only small amounts of PEDF were secreted which were not altered by any substance tested. Ultrastructural analysis showed no alterations in mitochondria after long-term treatment with either substance. Autophagosomes were not reduced by long-term anti-VEGF treatment compared to control. However, the area of autophagosomes in bevacizumab and aflibercept treated cells was significantly less compared to both ranibizumab and rituximab treated cells (all p < 0.05). Taken together, weekly treatment with VEGF-antagonists up to 3 months does not induce premature aging in primary RPE cells in any tested compound. A significant difference can be found between bevacizumab and aflibercept on the one hand, and ranibizumab (and rituximab) on the other hand, with more autophagosomal area in ranibizumab and (rituximab). Taken together, our data provide indications for long-term safety of anti-VEGF compounds. Further research is warranted.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Cellular Senescence/drug effects , Retinal Pigment Epithelium/drug effects , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Angiogenesis Inhibitors/toxicity , Animals , Bevacizumab/pharmacology , Blotting, Western , Cathepsin D/metabolism , Cell Survival , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Eye Proteins/metabolism , Follow-Up Studies , Microscopy, Electron , Nerve Growth Factors/metabolism , Phosphorylation , Ranibizumab/pharmacology , Receptors, Vascular Endothelial Growth Factor , Recombinant Fusion Proteins/pharmacology , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/ultrastructure , Rituximab/pharmacology , Serpins/metabolism , Swine , TOR Serine-Threonine Kinases/metabolism , Transforming Growth Factor beta/metabolism , beta-Galactosidase/metabolism
18.
Mol Pharm ; 15(7): 2770-2784, 2018 07 02.
Article in English | MEDLINE | ID: mdl-29734810

ABSTRACT

Neovascular age-related macular degeneration (wet AMD) results from the pathological angiogenesis of choroidal capillaries, which leak fluid within or below the macular region of the retina. The current standard of care for treating wet AMD utilizes intravitreal injections of anti-VEGF antibodies or antibody fragments to suppress ocular vascular endothelial growth factor (VEGF) levels. While VEGF suppression has been demonstrated in wet AMD patients by serial measurements of free-VEGF concentrations in aqueous humor samples, it is presumed that anti-VEGF molecules also permeate across the inner limiting membrane (ILM) of the retina as well as the retinal pigmented epithelium (RPE) and suppress VEGF levels in the retina and/or choroidal regions. The latter effects are inferred from serial optical coherence tomography (OCT) measurements of fluid in the retinal and sub-retinal spaces. In order to gain theoretical insights to the dynamics of retinal levels of free-VEGF following intravitreal injection of anti-VEGF molecules, we have extended our previous two-compartment pharmacokinetic/pharmacodynamic (PK/PD) model of ranibizumab-VEGF suppression in vitreous and aqueous humors to a three-compartment model that includes the retinal compartment. In the new model, reference values for the macromolecular permeability coefficients between retina and vitreous ( pILM) and between retina and choroid ( pRPE) were estimated from PK data obtained in rabbit. With these values, the three-compartment model was used to re-analyze the aqueous humor levels of free-VEGF obtained in wet AMD patients treated with ranibizumab and to compare them to the simulated retinal levels of free-VEGF, including the observed variability in PK and PD. We have also used the model to explore the impact of varying pILM and pRPE to assess the case in which an anti-VEGF molecule is impermeable to the ILM and to assess the potential effects of AMD pathology on the RPE barrier. Our simulations show that, for the reference values of pILM and pRPE, the simulated duration of VEGF suppression in the retina is approximately 50% shorter than the observed duration of VEGF suppression in the aqueous humor, a finding that may explain the short duration of suppressed disease activity in the "high anti-VEGF demand" patients reported by Fauser and Muether ( Br. J. Ophthalmol. 2016, 100, 1494-1498 ). At 10-fold lower values of pRPE, the durations of VEGF suppression in the retina and aqueous humor are comparable. Lastly we have used the model to explore the impact of dose and binding parameters on the duration and depth of VEGF suppression in the aqueous and retinal compartments. Our simulations with the three-compartment PK/PD model provide new insights into inter-patient variability in response to anti-VEGF therapy and offer a mechanistic framework for developing treatment regimens and molecules that may prolong the duration of retinal VEGF suppression.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Ranibizumab/pharmacology , Retina/pathology , Vascular Endothelial Growth Factor A/metabolism , Wet Macular Degeneration/drug therapy , Angiogenesis Inhibitors/therapeutic use , Aqueous Humor/drug effects , Aqueous Humor/metabolism , Humans , Intravitreal Injections , Models, Biological , Ranibizumab/therapeutic use , Retina/drug effects , Retinal Vessels/drug effects , Retinal Vessels/pathology , Vitreous Body/drug effects , Vitreous Body/metabolism , Wet Macular Degeneration/pathology
19.
Retina ; 38(6): 1120-1126, 2018 Jun.
Article in English | MEDLINE | ID: mdl-28520638

ABSTRACT

PURPOSE: The goal of this study was to assess the effects of a single injection of intravitreal ranibizumab (RAN) or bevacizumab (BEV) on the retinal vessel size in eyes with diabetic macular edema. MATERIALS AND METHODS: In total, 32 patients were enrolled in the RAN group, and 30 patients were included in BEV group. Each of these groups was also subdivided into two others groups: a study group and a control group. The study groups were composed of the injected eyes, whereas the noninjected fellow eyes served as the control groups. The patients underwent complete ophthalmic examinations, including optical coherence tomography and fundus fluorescein angiography, and the primary outcome measures included the central retinal artery equivalent, central retinal vein equivalent, and artery-to-vein ratio. RESULTS: In the RAN study group (n = 32), the preinjection mean central retinal artery equivalent (175.42 µm) decreased to 169.01 µm after 1 week, and to 167.47 µm after 1 month (P < 0.001), whereas the baseline central retinal vein equivalent (235.29 µm) decreased initially to 219.90 µm after 1 week, and to 218.36 µm after 1 month (P < 0.001). In the BEV study group (n = 30), the preinjection central retinal artery equivalent (150.21 µm) decreased to 146.25 µm after 1 week, and to 145.89 µm after 1 month (P < 0.001); whereas the baseline central retinal vein equivalent (211.87 µm) decreased initially to 204.59 µm after 1 week and was 205.24 µm after 1 month (P < 0.001). The preinjection artery-to-vein ratio values changed significantly (P = 0.001) after 1 week and after 1 month in the RAN group, but no significant alteration in the artery-to-vein ratio was observed in the BEV group (P = 0.433). In both the RAN (n = 32) and BEV (n = 30) control groups, none of the 3 parameters changed throughout the study period, when compared with the baseline. CONCLUSION: The results of this study showed that both RAN and BEV injections significantly constricted the retinal blood vessel diameters.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Bevacizumab/therapeutic use , Diabetic Retinopathy/drug therapy , Macular Edema/drug therapy , Ranibizumab/therapeutic use , Retinal Artery/pathology , Retinal Vein/pathology , Aged , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/pharmacology , Bevacizumab/administration & dosage , Bevacizumab/pharmacology , Diabetic Retinopathy/pathology , Female , Fluorescein Angiography , Humans , Intravitreal Injections , Macular Edema/pathology , Male , Middle Aged , Prospective Studies , Ranibizumab/administration & dosage , Ranibizumab/pharmacology , Retinal Artery/drug effects , Retinal Vein/drug effects
20.
Int J Mol Sci ; 19(6)2018 May 31.
Article in English | MEDLINE | ID: mdl-29857531

ABSTRACT

Retinal ischemia is an important factor in several eye disorders. To investigate the impact of VEGF inhibitors, as a therapeutic option, we studied these in a retinal ischemia animal model. Therefore, animals received bevacizumab or ranibizumab intravitreally one day after ischemia induction. Via electroretinography, a significant decrease in a- and b-wave amplitudes was detected fourteen days after ischemia, but they were reduced to a lesser extent in the ranibizumab group. Ischemic and bevacizumab retinae displayed fewer retinal ganglion cells (RGCs), while no significant cell loss was noted in the ranibizumab group. Apoptosis was reduced after therapy. More autophagocytotic cells were observed in ischemic and bevacizumab eyes, but not in ranibizumab eyes. Additionally, more microglia, as well as active ones, were revealed in all ischemic groups, but the increase was less prominent under ranibizumab treatment. Fewer cone bipolar cells were detected in ischemic eyes, in contrast to bevacizumab and ranibizumab-treated ones. Our results demonstrate a reduced apoptosis and autophagocytosis rate after ranibizumab treatment. Furthermore, a certain protection was seen regarding functionality, RGC, and bipolar cell availability, as well as microglia activation by ranibizumab treatment after ischemic damage. Thus, ranibizumab could be an option for treatment of retinal ischemic injury.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Ischemia/complications , Ischemia/metabolism , Ranibizumab/pharmacology , Retinal Diseases/etiology , Retinal Diseases/physiopathology , Animals , Biomarkers , Cell Death , Disease Models, Animal , Electroretinography , Immunohistochemistry , Male , Rats , Retinal Diseases/diagnosis , Retinal Diseases/drug therapy , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/metabolism , Retinal Vessels/pathology , Retinal Vessels/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL