Next Article in Journal
Study of the Bcl-2 Interactome by BiFC Reveals Differences in the Activation Mechanism of Bax and Bak
Previous Article in Journal
Transcriptional Response to Hypoxia: The Role of HIF-1-Associated Co-Regulators
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Melatonin Inhibits VEGF-Induced Endothelial Progenitor Cell Angiogenesis in Neovascular Age-Related Macular Degeneration

1
Graduate Institute of Biomedical Sciences, China Medical University, Taichung 403433, Taiwan
2
Department of Medicine, MacKay Medical College, New Taipei City 25245, Taiwan
3
Institute of Biomedical Sciences, Mackay Medical College, New Taipei City 25245, Taiwan
4
School of Pharmacy, College of Pharmacy, Kaohsiung 807378, Taiwan
5
Drug Development Center, China Medical University, Taichung 403433, Taiwan
6
Department of Medical Laboratory Science and Biotechnology, College of Medical and Health Science, Asia University, Taichung 40354, Taiwan
7
Research Center for Cancer Biology and Center for Molecular Medicine, China Medical University, Taichung 403433, Taiwan
8
Center for Molecular Medicine, China Medical University Hospital, Taichung 40402, Taiwan
9
Department of Orthopedic Surgery, China Medical University Hospital, Taichung 40402, Taiwan
10
Department of Sports Medicine, College of Health Care, China Medical University, Taichung 403433, Taiwan
11
Department of Orthopedic Surgery, China Medical University Beigang Hospital, Yun-Lin County 65152, Taiwan
12
Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
13
Department of Medical Research, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
14
Department of Pharmacology, School of Medicine, China Medical University, Taichung 403433, Taiwan
15
Chinese Medicine Research Center, China Medical University, Taichung 403433, Taiwan
16
Department of Medical Research, China Medical University Hsinchu Hospital, Hsinchu 40402, Taiwan
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Cells 2023, 12(5), 799; https://doi.org/10.3390/cells12050799
Submission received: 20 January 2023 / Revised: 15 February 2023 / Accepted: 28 February 2023 / Published: 3 March 2023

Abstract

:
Neovascular age-related macular degeneration (AMD) is described as abnormal angiogenesis in the retina and the leaking of fluid and blood that generates a huge, dark, blind spot in the center of the visual field, causing severe vision loss in over 90% of patients. Bone marrow-derived endothelial progenitor cells (EPCs) contribute to pathologic angiogenesis. Gene expression profiles downloaded from the eyeIntegration v1.0 database for healthy retinas and retinas from patients with neovascular AMD identified significantly higher levels of EPC-specific markers (CD34, CD133) and blood vessel markers (CD31, VEGF) in the neovascular AMD retinas compared with healthy retinas. Melatonin is a hormone that is mainly secreted by the pineal gland, and is also produced in the retina. Whether melatonin affects vascular endothelial growth factor (VEGF)-induced EPC angiogenesis in neovascular AMD is unknown. Our study revealed that melatonin inhibits VEGF-induced stimulation of EPC migration and tube formation. By directly binding with the VEGFR2 extracellular domain, melatonin significantly and dose-dependently inhibited VEGF-induced PDGF-BB expression and angiogenesis in EPCs via c-Src and FAK, NF-κB and AP-1 signaling. The corneal alkali burn model demonstrated that melatonin markedly inhibited EPC angiogenesis and neovascular AMD. Melatonin appears promising for reducing EPC angiogenesis in neovascular AMD.

1. Introduction

Age-related macular degeneration (AMD) is an age-related ocular disease that leads to visual impairment, drusen, retinal pigmentary changes, and blood vessel angiogenesis in the retina [1]. Increasing life expectancies and aging populations in most countries worldwide are contributing to a steady increase in the global prevalence of AMD [2]. AMD can be broadly classified as the non-neovascular (dry) or neovascular (wet) type. Neovascular AMD is described as abnormal angiogenesis in the retina and the leaking of fluid and blood that creates a large blind spot in the center of the visual field, causing severe vision loss in more than 90% of patients [3].
Angiogenesis is a complex process that regulates many physiological functions, including wound healing and tissue development, as well as reproduction [4], while it also contributes to pathological processes, such as atherosclerosis [5] and inflammatory diseases [6], as well as neovascular AMD [7]. Angiogenic factors such as vascular endothelial growth factor (VEGF) and platelet-derived growth factor-BB (PDGF-BB) contribute to neovascular AMD [8]. Inhibiting angiogenesis is therefore a critical strategy in neovascular AMD. Currently, anti-VEGF therapy is the only available treatment for neovascular AMD; this therapy targets the vascular endothelial growth factor receptor (VEGFR) [9]. Anti-VEGF/VEGFR2 therapy can effectively inhibit choroidal neovascularization (CNV) and downregulate levels of VEGF mRNA expression in mice with laser-induced CNV [10]. However, although anti-VEGF drugs have provided positive results in clinical trials, these outcomes have not translated into real-world outcomes [3,11]. Thus, other treatment targets besides VEGF and VEGFR are necessary for neovascular AMD.
Endothelial progenitor cells (EPCs) are derived from bone marrow-derived endothelial stem cells, and are involved in physiological and pathological angiogenesis [12]. EPCs are recruited in response to angiogenesis, and regulate several cellular functions such as proliferation and migration [13]. Characterized by their surface markers CD34 and CD133, as well as by VEGFR2, EPCs play an important role in the progression of new blood vessel formation [14]. Importantly, VEGF stimulates EPC angiogenesis, including the survival, motility, and tube formation of EPCs [15,16], via the VEGFR2/c-Src/FAK signaling pathway [17] as well as the transcription factors NF-κB [18] and AP-1 [19]; this mobilization of EPCs enables the development of neovascular AMD [20]. EPC-dependent angiogenesis appears to be a valuable treatment target for neovascular AMD, as the anti-VEGF drug ranibizumab significantly reduces the high levels of circulating EPCs that are involved in AMD angiogenesis [21].
The synthesis and release of the neurohormone melatonin enables organisms to respond to circadian and seasonal rhythms [22]. Melatonin is mainly secreted by the pineal gland under the influence of light stimulation of the retina; to a lesser extent, melatonin is also synthesized within the eye, where melatonin uses ocular structures to mediate a variety of diurnal rhythms and physiological processes within the eye [23]. Increasing the concentration of melatonin at night promotes sleep, while decreasing the concentration of melatonin during the day promotes alertness [23]. Melatonin concentrations tend to decrease with age [24]. The effects of melatonin are beneficial for numerous physiological functions, including the promotion of ocular surface wound healing [25], reductions in inflammation and oxidative stress [24], and angiogenesis [24]. The expression and secretion of VEGF is important in retinal physiology [26]. The release of VEGF from retinal pigment epithelial cells helps to protect neuronal cells and the choroid, maintaining a healthy retina [26]. Notably, melatonin reduces retinal levels of VEGF and protects against ocular angiogenesis diseases [26]. However, whether melatonin affects VEGF-induced EPC angiogenesis in neovascular AMD is unclear. This study therefore aimed to determine whether melatonin treatment inhibits VEGF-induced EPC angiogenesis during the development of neovascular AMD. The study also sought to define any other underlying mechanisms, such as signaling pathways, which may mediate this process.

2. Materials and Methods

2.1. Materials

Recombinant human VEGF (100-20) was bought from PeproTech (Rocky Hill, NJ, USA). VEGFR2 (07-158), phospho-VEGFR2 (orb106137), CD133 (orb13002), and β-actin (a5441) antibodies, and melatonin (M5250) were purchased from Sigma-Aldrich (St. Louis, MO, USA). Phospho-FAK (3283S) antibodies was purchased from Cell Signaling (Danvers, MA, USA). FAK (sc-1688), phospho-c-Src (sc-12928-R), c-Src (2105S), phospho-c-Jun (sc-822), c-Jun (sc-74543), phospho-p65 (sc-101752), p65 (sc-8008), CD31 (sc-18916), and CD34 (sc-74499) antibodies, as well as the angiotensin II (FAK activator; sc-363643) and c-Src activator (sc-3052), were purchased from Santa Cruz Biotechnology (Dallas, TX, USA). The NF-κB activator (prostratin; ab120880) was purchased from Abcam (Cambridge, MA, USA). PDGF-BB antibody (MBS9404630) was purchased from MyBioSource (San Diego, CA, USA). The VEGFR2 short hairpin RNA (shRNA) plasmid was purchased from the National RNAi Core Facility Platform (Taipei, Taiwan).

2.2. Cell Culture

Human primary EPCs were prepared as described in our previous protocols [6]. EPCs were cultured in MV2 complete medium (PromoCell, Heidelberg, Germany) with 20% fetal bovine serum (FBS; HyClone, Logan, UT, USA), and maintained in a humidified incubator at 37 °C, 5% CO2 [27].

2.3. Analysis of the Eyeintegration v1.0 Database

Gene expression profile records were downloaded from the eyeIntegration v1.0 database (https://eyeIntegration.nei.nih.gov, accessed on 13 January 2022) for 57 normal retina tissue samples and 448 retina tissue samples from AMD patients, for analysis of CD31, CD34, CD133, VEGF, and PDGF-BB expression [28].

2.4. Migration Assay

Transwell chambers (8.0 µm pore sizes; Corning, NY, USA) were used for the migration assay. EPCs (8 × 103 in each well) were applied to the upper chambers of 24-well plates in 200 μL of serum-free MV2 complete medium containing 100 ng/mL VEGF. The lower chambers were filled with 300 μL of MV2 complete medium combined with 5% FBS and different concentrations of melatonin (0.1, 0.3, or 1 mM), then incubated for 18 h at 37 °C and 5% CO2. The cells in the lower chambers were fixed with 3.7% formaldehyde and stained with 0.5% crystal violet solution. The numbers of migrated cells were counted by microscope (Nikon, Tokyo, Japan) and analyzed by MacBiophotonics ImageJ software [29].

2.5. Tube Formation Assay

The 48-well plates were coated with 150 μL of Matrigel (BD Biosciences, MA, USA) and incubated for 30 min at 37 °C. EPCs (2 × 104 cells/well) were seeded onto the gel layer in a 50% MV2 complete medium containing VEGF with melatonin (0.1, 0.3, or 1 mM), then incubated for 6 h at 37 °C. EPC tube formation was evaluated under an inverted phase-contrast microscope (Nikon, Tokyo, Japan). The number of tube branches was quantified by MacBioPhotonics ImageJ software [30].

2.6. Proliferation Assay

EPCs (5 × 103) were seeded in a 96-well plate and incubated with melatonin for 24 h or 48 h, before adding 10 μL of CCK-8 reagent (96992, Sigma-Aldrich, St. Louis, MO, USA) and incubating the EPCs at 37 °C for 2–4 h. The samples were quantified by a microplate reader (Bio-Tek, Winooski, VT, USA) at OD 570 nm.

2.7. Angiogenesis Protein Array

The cell lysate was examined with the Human Angiogenesis Protein Array (ARY007, R&D Systems, Minneapolis, MN, USA), following the manufacturer’s protocol [31]. The results of the protein array were quantified with the ImageQuant™ LAS 4000 biomolecular imager (GE Healthcare Life Sciences, Chicago, IL, USA).

2.8. Chromatin Immunoprecipitation (ChIP) Assay

The chromatin immunoprecipitation analysis followed the previously described methodology [32,33]. DNA was immunoprecipitated with an anti-p65 or anti-c-Jun antibody and purified by phenol-chloroform extraction. Immune complexes were collected with protein G-Sepharose beads and eluted from the beads using an elution buffer. The purified DNA pellet was subjected to PCR, and then the PCR products were analyzed on 1.5% agarose gel electrophoresis and visualized by UV light. The primers 5′-CCAAGAGGCTAGATTCACAGTCAC and 3′-TTCAGCTGTTCCGGCCTTT were used to amplify across the PDGF-BB promoter region (−197/−7).

2.9. Chick Chorioallantoic Membrane (CAM) Assay

The CAM assay was performed according to our previously published method [34]. VEGF and melatonin (0.1, 0.3, or 1 mM) were mixed with Matrigel and dropped onto the developing chicken egg, then incubated at 38 °C in an 80% humidified atmosphere. After 14 days, CAMs were observed by microscopy and photographic documentation. The number of CAM blood vessels was quantified by MacBioPhotonics ImageJ software.

2.10. Matrigel Plug Assay

The Matrigel plug angiogenesis assay was performed according to our previous research [34,35]. Male nude mice (4 weeks old) were subcutaneously injected with 300 μL Matrigel containing VEGF with the indicated concentrations of melatonin. The plugs were collected after 7 days and processed by immunofluorescence staining for co-staining with CD31, CD34 and CD133 antibodies. All samples were then stained with 40,6-diamidino-2-phenylindole (DAPI) and captured by the TissueFAXS-S-plus imaging system (TissueGnostics, Vienna, Austria). Hemoglobin levels were examined using Drabkin’s reagent and detected at 450 nm with a microplate reader (Bio-Tek, Winooski, VT, USA).

2.11. Molecular Docking

The structures of melatonin (PDB code: 5mxb) and VEGFR2 (PDB code: 3V2A) were downloaded from the Protein Data Bank (PDB, https://www.rcsb.org/, accessed on 11 May 2022). The molecular docking analysis between melatonin and the VEGFR2 extracellular domain (ECD) was performed using Discovery Studio software [36].

2.12. Corneal Alkali Burn Model

Male C57BL/6J mice (6–8 weeks old) were anesthetized with an intramuscular injection of Zoletil (0.2 mL/kg). Alkali burn injury was induced by placing 2 mm diameter filter paper (soaked in 1 mol/L NaOH) on the center of the right cornea for 30 s, before gently washing the ocular surface with 30 mL of 1X PBS solution. The mice were then immediately treated with melatonin by intraperitoneal (IP) injection (20 mg/kg or 60 mg/kg), or 1X PBS solution by IP injection, or the VEGF inhibitor bevacizumab (5 mg/μL) by eye drop, continuing with the same treatment regimen every 2 days. The eyeballs were observed by a microscope (Nikon, Tokyo, Japan) on days 0, 3, 5, and 7. On day 7, all mice were sacrificed, and their eyeballs were collected and fixed in Davidson’s fluid for histological analysis [37,38]. All animal procedures were performed according to an approved protocol issued by the Institutional Animal Care and Use Committee of China Medical University (Taichung, Taiwan).

2.13. Statistical Analysis

All statistical analyses were performed using GraphPad Prism 7.0 software, and all values are expressed as the mean  ±  standard deviation (SD). Differences between selected pairs from the study groups were analyzed for statistical significance using the Student’s t-test. One-way analysis of variance (ANOVA) followed by post hoc testing was used for statistical analyses of multiple group comparisons. The difference was considered to be significant if the p value was <0.05.

3. Results

3.1. Higher Levels of EPC Markers Correlate with AMD Progression

Angiogenesis plays an important role in AMD [39]. To confirm the involvement of EPCs in neovascular AMD, we analyzed gene expression profiles from the eyeIntegration v1.0 database. EPC-specific markers CD34 and CD133 and blood vessel markers CD31 and VEGF were all highly expressed in AMD retinas and graded with Minnesota Grading System (MGS) scores of 1 to 4, whereas their expression was low in retinas from healthy individuals (Figure 1), indicating that EPCs contribute to the progression of neovascular AMD.

3.2. Melatonin Inhibits VEGF-Promoted Proliferation, Migration, and the Tube Formation of EPCs without Cytotoxicity

In previous research, melatonin treatment has shown strong antiangiogenic effects by suppressing the proliferation, migration and invasion of endothelial cells [40], and melatonin effectively disrupted the tube formation of human umbilical vein endothelial cells (HUVECs) and dose-dependently reduced the viability of HUVECs [41], although the effects of melatonin are unclear in EPCs. When we examined the apoptotic effects of melatonin in EPCs, MTT assay results showed that incubating EPCs with different concentrations of melatonin (0.1–1 mM) for 24 h or 48 h did not affect cell viability (Figure 2A), so this concentration range was used for further experiments. The incubation of EPCs with VEGF (100 ng/mL) and melatonin (0.1–1 mM) for 24 h showed that VEGF alone promoted the proliferation of EPCs; VEGF plus melatonin significantly reduced this proliferation in a concentration-dependent manner (Figure 2B). Next, the migration assay results showed that melatonin significantly inhibited VEGF-induced cell migration in a dose-dependent manner (Figure 2C), while the tube formation assay showed that VEGF alone stimulated the reorganization and the formation of capillary-like structures in EPCs, whereas the addition of melatonin to VEGF significantly suppressed these effects (Figure 2D).

3.3. In Vivo Results Show that Melatonin Inhibits VEGF-Induced EPC Recruitment and Angiogenesis

To examine whether melatonin inhibits VEGF-induced angiogenesis in vivo, Matrigel was mixed with VEGF (100 ng/mL) and different concentrations of melatonin as indicated in the CAM assay (Figure 3A). The results showed that melatonin reduced VEGF-induced neoangiogenesis (Figure 3A). Using the Matrigel plug assay to examine the in vivo angiogenesis activity of melatonin, we observed that VEGF promoted microvessel formation and hemoglobin levels over 7 days in the Matrigel plugs, whereas melatonin significantly and dose-dependently inhibited this process (Figure 3B). In the co-immunofluorescent staining results for the blood vessel marker CD31 and EPC-specific markers CD34 and CD133, all markers were significantly and dose-dependently reduced by melatonin (Figure 3C,D). Our findings show that melatonin effectively suppresses VEGF-induced EPC recruitment and angiogenesis in vivo.

3.4. Melatonin Inhibits VEGF-Induced Increases in PDGF-BB Expression and EPC Angiogenesis

The results of the angiogenesis protein array show that VEGF treatment increased the amount of angiogenic factors in EPCs and that the addition of melatonin decreased these factors (Figure 4A); in particular, the expression of PDGF-BB was significantly reduced after melatonin treatment (Figure 4B). Furthermore, we found that melatonin decreased VEGF-induced PDGF-BB expression in EPCs in a dose-dependent manner (Figure 4C). Whereas melatonin significantly inhibited VEGF-induced EPC migration and tube formation, these effects were significantly reversed when the EPCs were transfected with PDGF-BB cDNA for 24 h (Figure 4D,E; the tube formation images are shown in Supplementary Figure S1). Moreover, we found that the levels of PDGF-BB gene expression were significantly higher in retinas from patients with neovascular AMD than in retinas from healthy individuals (Figure 4F). These results suggest that melatonin suppresses angiogenesis by inhibiting VEGF-induced PDGF-BB production in EPCs.

3.5. Melatonin Reduces VEGF-Induced EPC Angiogenic Functions by Inhibiting the VEGFR2/c-Src/FAK Signaling Pathway

We examined whether melatonin is capable of blocking the VEGF/VEGFR2 interaction. Using molecular docking software to predict melatonin-VEGFR2 binding affinity, we found that melatonin interacts with the VEGFR2 ECD, with an energy value of −27.121 kCal/mol for the docked complex (Figure 5A), indicating that melatonin inhibits VEGF-induced angiogenesis by directly binding with VEGFR2. Next, we found that melatonin inhibits the VEGF-induced phosphorylation of VEGFR2, c-Src, and FAK in EPCs (Figure 5B–D). We then transfected EPCs with VEGFR2 cDNA or treated the EPCs with FAK or c-Src activators to verify whether VEGF/VEGFR2 signaling regulates EPC angiogenesis. Whereas melatonin treatment significantly inhibited VEGF-induced EPC migration and tube formation, these effects were significantly reversed by transfection with VEGFR2 cDNA, and also by treatment with the FAK and c-Src activators (Figure 5E,F; the tube formation images are shown in Supplementary Figure S2), suggesting that melatonin inhibits the VEGF-induced phosphorylation of VEGFR2, c-Src, and FAK in EPCs via VEGFR2/c-Src/FAK signaling.

3.6. NF-κB and AP-1 Contribute to Melatonin-Induced Inhibition of EPC Angiogenesis

VEGF-induced phosphorylation of the NF-κB subunit (p65) and AP-1 subunit (c-Jun) was significantly and dose-dependently reduced by melatonin (Figure 6A,B). Next, we tested whether NF-κB or AP-1 are involved in the melatonin-induced inhibition of EPC angiogenesis. Treatment with NF-κB and AP-1 activators significantly promoted EPC angiogenesis, which was significantly reduced by melatonin treatment (Figure 6C,D). We then observed that melatonin significantly reduced NF-κB or AP-1 luciferase activity and their translocation into the nucleus (Figure 6E,F; the tube formation images are shown in Supplementary Figure S3). We also explored whether PDGF-BB is involved in the melatonin-induced inhibition of NF-κB or AP-1 transcriptional activation, using the ChIP assay to assess the in vitro recruitment of NF-κB or AP-1 to the PDGF-BB promoter. As shown in Figure 6G,H, VEGF induced the binding of NF-κB or AP-1 to the PDGF-BB element, and melatonin treatment reduced this binding. These results indicate that melatonin inhibits VEGF-induced PDGF-BB production and angiogenesis in EPCs via the suppression of NF-κB and AP-1 activity.

3.7. Melatonin Treatment Inhibits EPC Angiogenesis in the Corneal Alkali Burn Model

As shown in Figure 7A, corneal neovascularization was increased in the PBS-treated eyes, and decreased after melatonin treatment in a dose-dependent manner. Notably, the effects of melatonin in the highest dose group (60 mg/kg) were similar to those of the bevacizumab treatment group. After the animals were sacrificed on Day 7, the eyeballs were removed to examine corneal epithelial defects. H&E staining showed that corneal swelling was dose-dependently decreased by melatonin (Figure 7B,C). Co-immunofluorescence staining for EPCs and vessel markers showed that melatonin significantly and dose-dependently inhibited EPC angiogenesis and the recruitment of CD31-, CD34- and CD133-positive colonies (Figure 7D). The effects of melatonin 60 mg/kg were similar to those of bevacizumab treatment. Levels of PDGF-BB expression were increased in the control group and significantly reduced after melatonin or bevacizumab treatment (Figure 7E). Thus, melatonin appears to suppress PDGF-BB production, EPC recruitment, and angiogenesis, as well as AMD development in vivo.

4. Discussion

Neovascular AMD is a serious type of late AMD and an angiogenesis-dependent disease [42]. By triggering the mobilization of EPCs from the bone marrow and their recruitment during pathological states, VEGF supports the differentiation of EPCs into mature endothelial cells at angiogenesis sites [43]. EPCs may serve as a biomarker of neovascular AMD, based on the higher levels of EPC expression in blood from patients with AMD compared with blood from healthy controls [21]. Our analysis of gene expression profiles downloaded from the eyeIntegration v1.0 database confirmed high levels of EPC-specific markers (CD34 and CD133) in the retinal tissue of patients with AMD. Our preclinical findings also indicated that EPCs contribute to angiogenesis and AMD progression. Our results are consistent with pre-existing clinical evidence and emphasize the importance of EPCs in neovascular AMD.
VEGF is described as an endothelial cell-specific mitogen [44] that promotes the proliferation of new vessels and increases vascular permeability, and is considered to be a critical angiogenic factor of all angiogenesis processes [45]. Anti-VEGF treatment is an established therapeutic approach for neovascular AMD [46]. However, although the existing anti-VEGF agents, including bevacizumab, effectively inhibit VEGF activation [47] and EPC-derived pathological angiogenesis [48], many patients do not show the expected efficacy of anti-VEGF agents after repeated administration [11].
EPC-targeting therapies may therefore be a promising option to inhibit the angiogenesis process in neovascular AMD. In this study, EPCs were used to investigate the antiangiogenic effects of melatonin. Our findings show that melatonin inhibits VEGF-induced EPC angiogenesis in a concentration-dependent manner, without any evidence of cytotoxicity. Moreover, melatonin significantly suppressed the VEGF-induced stimulation of EPC recruitment and angiogenesis in vivo. Importantly, the corneal alkali burn model indicated that the inhibitory effects of melatonin on EPC-derived pathological angiogenesis are similar to those of bevacizumab, indicating that melatonin has potential against EPC-derived angiogenesis in neovascular AMD.
There is much evidence to indicate that the binding of VEGF to VEGFR2 regulates angiogenesis progression in physiological and pathological conditions, and promotes EPC-derived angiogenesis [49,50]. In our study, we found that melatonin inhibits VEGF-induced angiogenesis in EPCs by blocking the VEGF/VEGFR2 signaling pathway. Notably, molecular docking software results identified a good affinity between melatonin and the VEGFR2 ECD. To the best of our knowledge, this study is the first to identify that melatonin has the potential to bind with VEGFR2 and that melatonin may compete with VEGF in this binding process. Our in vitro evidence also showed that melatonin dramatically reduced the phosphorylation of VEGFR2 in EPCs. All of these results reveal that melatonin inhibits EPC-derived angiogenesis by regulating VEGF/VEGFR2 activity.
VEGF and PDGF-BB, as well as their receptors, are critical for normal and pathologic angiogenesis [51]. In neovascular AMD, VEGF promotes new vessel growth, while PDGF-BB maintains the interaction between pericytes and endothelial cells in maturing vessels [51]. While VEGF antagonists can inhibit angiogenesis in neovascular AMD, dual VEGF/PDGF inhibitors have proven to be even more beneficial in inhibiting angiogenesis in human endothelial cells and human pericytes in neovascular AMD [51], as well as in suppressing neovascularization in a mouse model of laser-induced CNV [52]. In our study, melatonin significantly reduced VEGF-induced PDGF-BB production in EPCs, as well as EPC migration and tube formation. In addition, our findings from the corneal alkali burn model suggest that melatonin can significantly inhibit corneal levels of PDGF-BB expression and angiogenic activity. Our results suggest that melatonin suppresses angiogenesis in EPCs by inhibiting PDGF-BB production.
The promotion or inhibition of angiogenesis is part of the homeostatic balance, with positive and negative effects outside the optimum range. Melatonin influences this balance, with evidence from several clinical research investigations demonstrating that this hormone has antiangiogenic effects in cancer and chronic ocular diseases. For instance, in cancer treatment, adjuvant melatonin appears to be very effective in early-stage disease and helps to reduce the side effect profiles after radiotherapy and chemotherapy [53]. In patients with central serous chorioretinopathy, treatment with oral melatonin (3 mg) three times daily for 1 month resulted in significant improvements from baseline in best-corrected visual acuity (BCVA) and decreases in central macular thickness (CMT), without any adverse effects [54]. The attractive side effects profile and relatively low cost of melatonin suggest that this hormone may be appropriate in a chronic ocular disease such as AMD, although supporting data from large prospective studies are needed before melatonin can be used in the clinic [54]. The FDA-approved the anti-VEGF agents bevacizumab, aflibercept, and ranibizumab, and have shown good therapeutic results in neovascular AMD, but anti-VEGF agents are limited by the necessity for monthly injections in the clinic and the long-term nature of treatment [55]. Moreover, the high cost of anti-VEGF therapy is a heavy burden for patients [56]. Our study evidence suggests that melatonin may overcome new existing therapeutic obstacles with anti-VEGF agents and offer a novel option for treating neovascular AMD.
In conclusion, our study indicates that melatonin inhibits VEGF-induced increases in PDGF-BB expression in EPCs by inhibiting the signaling of VEGFR2, c-Src, FAK, NF-κB and AP-1, all of which appear to effectively inhibit EPC angiogenesis (Figure 8). Thus, melatonin shows promising therapeutic potential, alone and in combination with a VEGF inhibitor, for neovascular AMD.

Supplementary Materials

The following supporting information can be download at: https://www.mdpi.com/article/10.3390/cells12050799/s1, Figure S1: Melatonin suppresses tube formation by inhibiting PDGF-BB expression in EPCs. Figure S2: Melatonin suppresses tube formation by inhibiting VEGFR2, c-Src, and FAK expression in EPCs. Figure S3: Melatonin suppresses tube formation by inhibiting NF-κB and AP-1 expression in EPCs. Methodology for the MTT assay, western blot analysis, and luciferase reporter assay are detailed in the Supplementary File.

Author Contributions

Conceptualization, L.-W.L. and C.-H.T.; Data curation, C.-Y.K.; Formal analysis, S.-F.Y.; Funding acquisition, C.-H.T.; Investigation, C.-Y.L. and Y.-C.F.; Methodology, S.-W.W., C.-Y.K., Y.-C.F., and J.-J.L.; Resources, S.-W.W.; Software, W.-C.H. and T.K.H.; Validation, L.-W.L., W.-C.H. and T.K.H.; Writing—original draft, L.-W.L.; Writing—review & editing, C.-H.T. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by grants from the Ministry of Science and Technology of Taiwan (MOST 111-2314-B-039-048-MY3), China Medical University (CMU111-ASIA-04; CMU111-ASIA-05), China Medical University Hospital (DMR-111-108; DMR-111-229; DMR-110-176; DMR-111-165; DMR-112-090; DMR-112-146; DMR-112-096) and MacKay Medical College (MMC-RD-110-1B-P015; MMC-RD-110-1B-P023; MMC-RD-111-1B-P010; MMC-RD-111-1B-P016).

Institutional Review Board Statement

All animal procedures were approved and performed according to guidance issued by the Institutional Animal Care and Use Committee of China Medical University (Approval no. 2021-174).

Informed Consent Statement

Not applicable.

Data Availability Statement

The data generated and analyzed will be made from the corresponding author on reasonable request.

Acknowledgments

We would like to thank Suh-Hang Juo of China Medical University for providing guidance on the corneal alkali burn model, and to Iona J. MacDonald of China Medical University for her editing.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Ma, Q.; Reiter, R.J.; Chen, Y. Role of melatonin in controlling angiogenesis under physiological and pathological conditions. Angiogenesis 2019, 23, 91–104. [Google Scholar] [CrossRef] [PubMed]
  2. Wang, Y.; Zhong, Y.; Zhang, L.; Wu, Q.; Tham, Y.; Rim, T.H.; Kithinji, D.M.; Wu, J.; Cheng, C.; Liang, H.; et al. Global Incidence, Progression, and Risk Factors of Age-Related Macular Degeneration and Projection of Disease Statistics in 30 Years: A Modeling Study. Gerontology 2021, 68, 721–735. [Google Scholar] [CrossRef] [PubMed]
  3. Patel, P.; Sheth, V. New and Innovative Treatments for Neovascular Age-Related Macular Degeneration (nAMD). J. Clin. Med. 2021, 10, 2436. [Google Scholar] [CrossRef] [PubMed]
  4. Folkman, J. Angiogenesis. Annu. Rev. Med. 2006, 57, 1–18. [Google Scholar] [CrossRef]
  5. Kahlon, R.; Shapero, J.; Gotlieb, A.I. Angiogenesis in atherosclerosis. Can. J. Cardiol. 1992, 8, 60–64. [Google Scholar]
  6. Tsai, C.H.; Chen, C.J.; Gong, C.L.; Liu, S.C.; Chen, P.C.; Huang, C.C.; Hu, S.L.; Wang, S.W.; Tang, C.H. CXCL13/CXCR5 axis facilitates endothelial progenitor cell homing and angiogenesis during rheumatoid arthritis progression. Cell Death Dis. 2021, 12, 846. [Google Scholar] [CrossRef]
  7. Ng, E.W.; Adamis, A.P. Targeting angiogenesis, the underlying disorder in neovascular age-related macular degeneration. Can. J. Ophthalmol. 2005, 40, 352–368. [Google Scholar] [CrossRef]
  8. Ding, K.; Eaton, L.; Bowley, D.; Rieser, M.; Chang, Q.; Harris, M.C.; Clabbers, A.; Dong, F.; Shen, J.; Hackett, S.F.; et al. Generation and characterization of ABBV642, a dual variable domain immunoglobulin molecule (DVD-Ig) that potently neutralizes VEGF and PDGF-BB and is designed for the treatment of exudative age-related macular degeneration. mAbs 2016, 9, 269–284. [Google Scholar] [CrossRef] [Green Version]
  9. Li, E.; Donati, S.; Lindsley, K.B.; Krzystolik, M.G.; Virgili, G. Treatment regimens for administration of an-ti-vascular endothelial growth factor agents for neovascular age-related macular degeneration. Cochrane Database Syst. Rev. 2020, 5, CD012208. [Google Scholar]
  10. Gu, L.; Chen, H.; Tuo, J.; Gao, X.; Chen, L. Inhibition of experimental choroidal neovascularization in mice by an-ti-VEGFA/VEGFR2 or non-specific siRNA. Exp. Eye Res. 2010, 91, 433–439. [Google Scholar] [CrossRef]
  11. Ricci, F.; Bandello, F.; Navarra, P.; Staurenghi, G.; Stumpp, M.; Zarbin, M. Neovascular Age-Related Macular De-generation: Therapeutic Management and New-Upcoming Approaches. Int. J. Mol. Sci. 2020, 21, 8242. [Google Scholar] [CrossRef]
  12. Asahara, T.; Masuda, H.; Takahashi, T.; Kalka, C.; Pastore, C.; Silver, M.; Kearne, M.; Magner, M.; Isner, J.M. Bone Marrow Origin of Endothelial Progenitor Cells Responsible for Postnatal Vasculogenesis in Physiological and Pathological Neovascularization. Circ. Res. 1999, 85, 221–228. [Google Scholar] [CrossRef] [Green Version]
  13. Lee, H.-P.; Chen, P.-C.; Wang, S.-W.; Fong, Y.-C.; Tsai, C.-H.; Tsai, F.-J.; Chung, J.-G.; Huang, C.-Y.; Yang, J.-S.; Hsu, Y.-M.; et al. Plumbagin suppresses endothelial progenitor cell-related angiogenesis in vitro and in vivo. J. Funct. Foods 2018, 52, 537–544. [Google Scholar] [CrossRef]
  14. Lee, H.P.; Wang, S.W.; Wu, Y.C.; Tsai, C.H.; Tsai, F.J.; Chung, J.G.; Huang, C.Y.; Yang, J.S.; Hsu, Y.M.; Yin, M.C.; et al. Glucocerebroside reduces endothelial progenitor cell-induced angiogenesis. Food Agric. Immunol. 2019, 30, 1033–1045. [Google Scholar] [CrossRef] [Green Version]
  15. Lin, C.Y.; Tzeng, H.E.; Li, T.M.; Chen, H.T.; Lee, Y.; Yang, Y.C.; Wang, S.W.; Yang, W.H.; Tang, C.H. WISP-3 inhibition of miR-452 promotes VEGF-A expression in chondrosar-coma cells and induces endothelial progenitor cells angiogenesis. Oncotarget 2017, 8, 39571–39581. [Google Scholar] [CrossRef]
  16. Tsai, C.H.; Liu, S.C.; Chung, W.H.; Wang, S.W.; Wu, M.H.; Tang, C.H. Visfatin Increases VEGF-dependent Angiogene-sis of Endothelial Progenitor Cells during Osteoarthritis Progression. Cells 2020, 9, 1315. [Google Scholar] [CrossRef]
  17. Li, T.M.; Liu, S.C.; Huang, Y.H.; Huang, C.C.; Hsu, C.J.; Tsai, C.H.; Wang, S.W.; Tang, C.H. YKL-40-Induced Inhibition of miR-590-3p Promotes Interleukin-18 Expres-sion and Angiogenesis of Endothelial Progenitor Cells. Int. J. Mol. Sci. 2017, 18, 920. [Google Scholar] [CrossRef] [Green Version]
  18. Tsai, H.-C.; Cheng, S.-P.; Han, C.-K.; Huang, Y.-L.; Wang, S.-W.; Lee, J.-J.; Lai, C.-T.; Fong, Y.-C.; Tang, C.-H. Resistin enhances angiogenesis in osteosarcoma via the MAPK signaling pathway. Aging 2019, 11, 9767–9777. [Google Scholar] [CrossRef]
  19. Wang, C.Q.; Lin, C.Y.; Huang, Y.L.; Wang, S.W.; Wang, Y.; Huang, B.F.; Lai, Y.W.; Weng, S.L.; Fong, Y.C.; Tang, C.H.; et al. Sphingosine-1-phosphate promotes PDGF-dependent endothelial pro-genitor cell angiogenesis in human chondrosarcoma cells. Aging 2019, 11, 11040–11053. [Google Scholar] [CrossRef]
  20. Scotti, F.; Maestroni, A.; Palini, A.; Introini, U.; Setaccioli, M.; Lorenzi, M.; Zerbini, G. Endothelial progenitor cells and response to ranibizumab in age-related macular degeneration. Retina 2014, 34, 1802–1810. [Google Scholar] [CrossRef]
  21. Mucciolo, D.P.; Marcucci, R.; Sodi, A.; Cesari, F.; Murro, V.; Rogolino, A.; Rizzo, S.; Giusti, B.; Virgili, G.; Prisco, D.; et al. Circulating endothelial and progenitor cells in age-related macular degeneration. Eur. J. Ophthalmol. 2019, 30, 956–965. [Google Scholar] [CrossRef] [PubMed]
  22. Stehle, J.H.; Saade, A.; Rawashdeh, O.; Ackermann, K.; Jilg, A.; Sebestény, T.; Maronde, E. A survey of molecular details in the human pineal gland in the light of phylogeny, structure, function and chronobiological diseases. J. Pineal Res. 2011, 51, 17–43. [Google Scholar] [CrossRef]
  23. Ostrin, L.A. Ocular and systemic melatonin and the influence of light exposure. Clin. Exp. Optom. 2018, 102, 99–108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Crooke, A.; Huete-Toral, F.; Colligris, B.; Pintor, J. The role and therapeutic potential of melatonin in age-related ocular diseases. J. Pineal Res. 2017, 63, e12430. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Pescosolido, N.; Gatto, V.; Stefanucci, A.; Rusciano, D. Oral treatment with the melatonin agonist agomelatine lowers the intraocular pressure of glaucoma patients. Ophthalmic Physiol. Opt. 2015, 35, 201–205. [Google Scholar] [CrossRef]
  26. Klettner, A.; Kampers, M.; Töbelmann, D.; Roider, J.; Dittmar, M. The Influence of Melatonin and Light on VEGF Secretion in Primary RPE Cells. Biomolecules 2021, 11, 114. [Google Scholar] [CrossRef]
  27. Chien, S.Y.; Huang, C.Y.; Tsai, C.H.; Wang, S.W.; Lin, Y.M.; Tang, C.H. Interleukin-1beta induces fibroblast growth factor 2 expression and subsequently promotes endothelial progenitor cell angiogenesis in chondrocytes. Clin. Sci. 2016, 130, 667–681. [Google Scholar] [CrossRef] [Green Version]
  28. Swamy, V.; McGaughey, D. Eye in a Disk: EyeIntegration Human Pan-Eye and Body Transcriptome Database Version 1.0. Investig. Ophthalmol. Vis. Sci. 2019, 60, 3236–3246. [Google Scholar] [CrossRef] [Green Version]
  29. Lin, C.C.; Chen, K.B.; Tsai, C.H.; Tsai, F.J.; Huang, C.Y.; Tang, C.H.; Yang, J.S.; Hsu, Y.M.; Peng, S.F.; Chung, J.G. Casticin inhibits human prostate cancer DU 145 cell migration and inva-sion via Ras/Akt/NF-kappaB signaling pathways. J. Food Biochem. 2019, 43, e12902. [Google Scholar] [CrossRef]
  30. Achudhan, D.; Liu, S.; Lin, Y.; Lee, H.; Wang, S.; Huang, W.; Wu, Y.; Kuo, Y.; Tang, C. Antcin K inhibits VEGF-dependent angiogenesis in human rheumatoid arthritis synovial fibroblasts. J. Food Biochem. 2021, 46, e14022. [Google Scholar] [CrossRef]
  31. Hagman, H.; Bendahl, P.O.; Lidfeldt, J.; Belting, M.; Johnsson, A. Protein array profiling of circulating angiogene-sis-related factors during bevacizumab containing treatment in metastatic colorectal cancer. PLoS ONE 2018, 13, e0209838. [Google Scholar] [CrossRef]
  32. Wu, M.H.; Huang, C.Y.; Lin, J.A.; Wang, S.W.; Peng, C.Y.; Cheng, H.C.; Tang, C.H. Endothelin-1 promotes vascular endothelial growth factor-dependent an-giogenesis in human chondrosarcoma cells. Oncogene 2014, 33, 1725–1735. [Google Scholar] [CrossRef] [Green Version]
  33. Chow, E.K.; O’Connell, R.M.; Schilling, S.; Wang, X.F.; Fu, X.Y.; Cheng, G. TLR agonists regulate PDGF-B production and cell proliferation through TGF-beta/type I IFN crosstalk. EMBO J. 2005, 24, 4071–4081. [Google Scholar] [CrossRef] [Green Version]
  34. Chen, C.-Y.; Su, C.-M.; Hsu, C.-J.; Huang, C.-C.; Wang, S.-W.; Liu, S.-C.; Chen, W.-C.; Fuh, L.-J.; Tang, C.-H. CCN1 Promotes VEGF Production in Osteoblasts and Induces Endothelial Progenitor Cell Angiogenesis by Inhibiting miR-126 Expression in Rheumatoid Arthritis. J. Bone Miner. Res. 2016, 32, 34–45. [Google Scholar] [CrossRef] [Green Version]
  35. Storgard, C.; Mikolon, D.; Stupack, D.G. Angiogenesis Assays in the Chick CAM. Methods Mol. Biol. 2005, 294, 123–136. [Google Scholar] [CrossRef]
  36. Cheng, F.J.; Huynh, T.K.; Yang, C.S.; Hu, D.W.; Shen, Y.C.; Tu, C.Y.; Wu, Y.C.; Tang, C.H.; Huang, W.C.; Chen, Y.; et al. Hesperidin Is a Potential Inhibitor against SARS-CoV-2 Infection. Nutrients 2021, 13, 2800. [Google Scholar] [CrossRef]
  37. Latendresse, J.R.; Warbrittion, A.R.; Jonassen, H.; Creasy, D.M. Fixation of testes and eyes using a modified Da-vidson’s fluid: Comparison with Bouin’s fluid and conventional Davidson’s fluid. Toxicol. Pathol. 2002, 30, 524–533. [Google Scholar] [CrossRef]
  38. Kim, J.W.; Jeong, H.; Yang, M.-S.; Lim, C.W.; Kim, B. Therapeutic effects of zerumbone in an alkali-burned corneal wound healing model. Int. Immunopharmacol. 2017, 48, 126–134. [Google Scholar] [CrossRef]
  39. Grierson, R.; Meyer-Rüsenberg, B.; Kunst, F.; Berna, M.J.; Richard, G.; Thill, M. Endothelial Progenitor Cells and Plasma Vascular Endothelial Growth Factor and Stromal Cell-Derived Factor-1 During Ranibizumab Treatment for Neovascular Age-Related Macular Degeneration. J. Ocul. Pharmacol. Ther. 2013, 29, 530–538. [Google Scholar] [CrossRef]
  40. Alvarez-García, V.; González, A.; Alonso-González, C.; Martínez-Campa, C.; Cos, S. Antiangiogenic effects of melatonin in endothelial cell cultures. Microvasc. Res. 2013, 87, 25–33. [Google Scholar] [CrossRef]
  41. Cheng, J.; Yang, H.L.; Gu, C.J.; Liu, Y.K.; Shao, J.; Zhu, R.; He, Y.Y.; Zhu, X.Y.; Li, M.Q. Melatonin restricts the viability and angiogenesis of vascular endothelial cells by suppressing HIF-1alpha/ROS/VEGF. Int. J. Mol. Med. 2019, 43, 945–955. [Google Scholar] [PubMed] [Green Version]
  42. Fallah, A.; Sadeghinia, A.; Kahroba, H.; Samadi, A.; Heidari, H.R.; Bradaran, B.; Zeinali, S.; Molavi, O. Therapeutic targeting of angiogenesis molecular pathways in an-giogenesis-dependent diseases. Biomed Pharmacother. 2019, 110, 775–785. [Google Scholar] [CrossRef] [PubMed]
  43. Lee, H.-P.; Wang, S.-W.; Wu, Y.-C.; Lin, L.-W.; Tsai, F.-J.; Yang, J.-S.; Li, T.-M.; Tang, C.-H. Soya-cerebroside inhibits VEGF-facilitated angiogenesis in endothelial progenitor cells. Food Agric. Immunol. 2020, 31, 193–204. [Google Scholar] [CrossRef] [Green Version]
  44. Hornig, C.; Barleon, B.; Ahmad, S.; Vuorela, P.; Ahmed, A.; Weich, H.A. Release and Complex Formation of Soluble VEGFR-1 from Endothelial Cells and Biological Fluids. Lab. Investig. 2000, 80, 443–454. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Shibuya, M. Vascular Endothelial Growth Factor (VEGF) and Its Receptor (VEGFR) Signaling in Angiogene-sis: A Crucial Target for Anti- and Pro-Angiogenic Therapies. Genes Cancer 2011, 2, 1097–1105. [Google Scholar] [CrossRef]
  46. Mantel, I.; Borgo, A.; Guidotti, J.; Forestier, E.; Kirsch, O.; Derradji, Y.; Waridel, P.; Burdet, F.; Mehl, F.; Schweizer, C.; et al. Molecular Biomarkers of Neovascular Age-Related Macular Degenera-tion With Incomplete Response to Anti-Vascular Endothelial Growth Factor Treatment. Front. Pharmacol. 2020, 11, 594087. [Google Scholar] [CrossRef]
  47. Cho, Y.-K.; Park, D.-H.; Jeon, I.-C. Medication Trends for Age-Related Macular Degeneration. Int. J. Mol. Sci. 2021, 22, 11837. [Google Scholar] [CrossRef]
  48. Sudo, K.; Sato, K.; Sakamoto, S.; Hasegawa, Y.; Asano, M.; Okuda, Y.; Takeda, M.; Sano, M.; Watanabe, H.; Shioya, T.; et al. Association Between Endothelial Progenitor Cells and Treatment Re-sponse in Non-Squamous Non-small Cell Lung Cancer Treated with Bevacizumab. Anticancer Res. 2017, 37, 5565–5571. [Google Scholar]
  49. Wang, X.; Bove, A.M.; Simone, G.; Ma, B. Molecular Bases of VEGFR-2-Mediated Physiological Function and Pathological Role. Front. Cell Dev. Biol. 2020, 8, 599281. [Google Scholar] [CrossRef]
  50. Lee, H.-P.; Liu, Y.-C.; Chen, P.-C.; Tai, H.-C.; Li, T.-M.; Fong, Y.-C.; Chang, C.-S.; Wu, M.-H.; Chiu, L.-P.; Wang, C.-J.; et al. Tanshinone IIA inhibits angiogenesis in human endothelial progenitor cells in vitro and in vivo. Oncotarget 2017, 8, 109217–109227. [Google Scholar] [CrossRef] [Green Version]
  51. Siedlecki, J.; Wertheimer, C.; Wolf, A.; Liegl, R.; Priglinger, C.; Priglinger, S.; Eibl-Lindner, K. Combined VEGF and PDGF inhibition for neovascular AMD: An-ti-angiogenic properties of axitinib on human endothelial cells and pericytes in vitro. Graefes Arch. Clin. Exp. Ophthalmol. 2017, 255, 963–972. [Google Scholar] [CrossRef]
  52. Tavakoli, S.; Puranen, J.; Bahrpeyma, S.; Lautala, V.E.; Karumo, S.; Lajunen, T.; del Amo, E.M.; Ruponen, M.; Urtti, A. Liposomal sunitinib for ocular drug delivery: A potential treat-ment for choroidal neovascularization. Int. J. Pharm. 2022, 620, 121725. [Google Scholar] [CrossRef]
  53. Bhattacharya, S.; Patel, K.K.; Dehari, D.; Agrawal, A.K.; Singh, S. Melatonin and its ubiquitous anticancer effects. Mol. Cell. Biochem. 2019, 462, 133–155. [Google Scholar] [CrossRef]
  54. Sartini, F.; Figus, M.; Nardi, M.; Casini, G.; Posarelli, C. Non-resolving, recurrent and chronic central serous chorioretinopathy: Available treatment options. Eye 2019, 33, 1035–1043. [Google Scholar] [CrossRef]
  55. Tan, C.S.; Ngo, W.K.; Chay, I.W.; Ting, D.S.; Sadda, S.R. Neovascular Age-Related Macular Degeneration (nAMD): A Review of Emerging Treatment Options. Clin. Ophthalmol. 2022, 16, 917–933. [Google Scholar] [CrossRef]
  56. Van Asten, F.; Michels, C.T.; Hoyng, C.B.; van der Wilt, G.J.; Klevering, B.J.; Rovers, M.M.; Grutters, J.P. The cost-effectiveness of bevacizumab, ranibizumab and aflibercept for the treatment of age-related macular degeneration-A cost-effectiveness analysis from a societal perspective. PLoS ONE 2018, 13, e0197670. [Google Scholar] [CrossRef]
Figure 1. (AD) High levels of EPCs and blood vessel markers in AMD retinas were graded by Minnesota Grading System (MGS) scores. Levels of EPC-specific markers (CD34 and CD133) and blood vessel markers (CD31 and VEGF) were analyzed in gene expression profiles of normal retina tissue and neovascular AMD tissue samples downloaded from the eyeIntegration v1.0 database. p < 0.05 compared with the normal group.
Figure 1. (AD) High levels of EPCs and blood vessel markers in AMD retinas were graded by Minnesota Grading System (MGS) scores. Levels of EPC-specific markers (CD34 and CD133) and blood vessel markers (CD31 and VEGF) were analyzed in gene expression profiles of normal retina tissue and neovascular AMD tissue samples downloaded from the eyeIntegration v1.0 database. p < 0.05 compared with the normal group.
Cells 12 00799 g001
Figure 2. Melatonin decreases VEGF-induced EPC proliferation, cell migration, and tube formation without cytotoxic effects. (A) EPCs were incubated with melatonin (0.1–1 mM) for 24 h or 48 h, and cell viability was examined using the MTT assay (n = 4). (B) EPCs were incubated with VEGF (100 ng/mL) and melatonin (0.1–1 mM) for 24 h. Cell proliferation (n = 4) was examined by the CCK-8 assay. (C) EPCs were incubated with VEGF (100 ng/mL), and different concentrations of melatonin (0.1–1 mM) for 18 h. Cell migration (n = 3) was examined by the Transwell assay. (D) EPCs were incubated with VEGF (100 ng/mL) and different concentrations of melatonin (0.1–1 mM) for 6 h. The capillary-like structure formation was determined by the tube formation assay (n = 3). * p < 0.05 compared with the control group; # p < 0.05 compared with the VEGF-treated group.
Figure 2. Melatonin decreases VEGF-induced EPC proliferation, cell migration, and tube formation without cytotoxic effects. (A) EPCs were incubated with melatonin (0.1–1 mM) for 24 h or 48 h, and cell viability was examined using the MTT assay (n = 4). (B) EPCs were incubated with VEGF (100 ng/mL) and melatonin (0.1–1 mM) for 24 h. Cell proliferation (n = 4) was examined by the CCK-8 assay. (C) EPCs were incubated with VEGF (100 ng/mL), and different concentrations of melatonin (0.1–1 mM) for 18 h. Cell migration (n = 3) was examined by the Transwell assay. (D) EPCs were incubated with VEGF (100 ng/mL) and different concentrations of melatonin (0.1–1 mM) for 6 h. The capillary-like structure formation was determined by the tube formation assay (n = 3). * p < 0.05 compared with the control group; # p < 0.05 compared with the VEGF-treated group.
Cells 12 00799 g002
Figure 3. Effects of melatonin on VEGF-induced EPC angiogenesis in vivo. (A) Five-day-old fertilized chick embryos (n = 5) were treated with VEGF (100 ng/mL) and different concentrations of melatonin (0.1–1 mM) for 14 days. After treatment, the CAMs were examined by microscopy and photographed. (BD) Matrigel plugs were treated with PBS (control group) or VEGF (100 ng/mL) with different concentrations of melatonin (0.1–1 mM) and subcutaneously injected into the flanks of nude mice (n = 5). After 7 days, the plugs were photographed, and then hemoglobin levels were quantified and visualized by co-immunofluorescence staining at 20× magnification for CD31, CD34, and CD133 antibodies. * p < 0.05 compared with the control group; # p < 0.05 compared with the VEGF-treated group.
Figure 3. Effects of melatonin on VEGF-induced EPC angiogenesis in vivo. (A) Five-day-old fertilized chick embryos (n = 5) were treated with VEGF (100 ng/mL) and different concentrations of melatonin (0.1–1 mM) for 14 days. After treatment, the CAMs were examined by microscopy and photographed. (BD) Matrigel plugs were treated with PBS (control group) or VEGF (100 ng/mL) with different concentrations of melatonin (0.1–1 mM) and subcutaneously injected into the flanks of nude mice (n = 5). After 7 days, the plugs were photographed, and then hemoglobin levels were quantified and visualized by co-immunofluorescence staining at 20× magnification for CD31, CD34, and CD133 antibodies. * p < 0.05 compared with the control group; # p < 0.05 compared with the VEGF-treated group.
Cells 12 00799 g003
Figure 4. Melatonin reduces VEGF-induced EPC migration and tube formation by inhibiting PDGF-BB production. (A) EPCs were incubated with VEGF (100 ng/mL) alone and in combination with melatonin (1 mM) for 24 h. Cell lysates were collected from each treatment condition and from untreated EPCs, and then profiled for proteomes using the Human Angiogenesis Protein Array. (B) Levels of PDGF-BB expression were quantified in each protein array sample. (C) EPCs were incubated with VEGF (100 ng/mL) and melatonin (0.1-1 mM) for 24 h, and PDGF-BB expression was examined by western blot analysis (n = 3). (D,E) EPCs were transfected with PDGF-BB cDNA overnight, then left untreated or were treated with VEGF (100 ng/mL) and melatonin (1 mM) for 24 h, before being examined by the Transwell (n = 3) and tube formation assays (n = 3). (F) Levels of PDGF-BB expression were analyzed in gene expression profile records downloaded from the eyeIntegration v1.0 database for normal retina tissue samples and neovascular AMD tissue samples. * p < 0.05 compared with the control group; # p < 0.05 compared with the VEGF-treated group.
Figure 4. Melatonin reduces VEGF-induced EPC migration and tube formation by inhibiting PDGF-BB production. (A) EPCs were incubated with VEGF (100 ng/mL) alone and in combination with melatonin (1 mM) for 24 h. Cell lysates were collected from each treatment condition and from untreated EPCs, and then profiled for proteomes using the Human Angiogenesis Protein Array. (B) Levels of PDGF-BB expression were quantified in each protein array sample. (C) EPCs were incubated with VEGF (100 ng/mL) and melatonin (0.1-1 mM) for 24 h, and PDGF-BB expression was examined by western blot analysis (n = 3). (D,E) EPCs were transfected with PDGF-BB cDNA overnight, then left untreated or were treated with VEGF (100 ng/mL) and melatonin (1 mM) for 24 h, before being examined by the Transwell (n = 3) and tube formation assays (n = 3). (F) Levels of PDGF-BB expression were analyzed in gene expression profile records downloaded from the eyeIntegration v1.0 database for normal retina tissue samples and neovascular AMD tissue samples. * p < 0.05 compared with the control group; # p < 0.05 compared with the VEGF-treated group.
Cells 12 00799 g004
Figure 5. Melatonin reduces the VEGF-induced stimulation of EPC migration and tube formation by inhibiting the VEGFR2/c-Src/FAK signaling pathway. (A) Molecular docking software revealed a binding affinity between melatonin and VEGFR2. (BD) EPCs were incubated with VEGF (100 ng/mL) and different concentrations of melatonin (0.1–1 mM) for 2 h. VEGFR-2, c-Src and FAK phosphorylation was examined by western blot (n = 3). (E,F) EPCs were transfected with VEGFR2 cDNA or treated with FAK or c-Src activators overnight, then left untreated or were treated with VEGF (100 ng/mL) alone and in combination with melatonin (1 mM) for 24 h, before being examined by the Transwell (n = 4) and tube formation assays (n = 4). * p < 0.05 compared with the control group; # p < 0.05 compared with the VEGF-treated group.
Figure 5. Melatonin reduces the VEGF-induced stimulation of EPC migration and tube formation by inhibiting the VEGFR2/c-Src/FAK signaling pathway. (A) Molecular docking software revealed a binding affinity between melatonin and VEGFR2. (BD) EPCs were incubated with VEGF (100 ng/mL) and different concentrations of melatonin (0.1–1 mM) for 2 h. VEGFR-2, c-Src and FAK phosphorylation was examined by western blot (n = 3). (E,F) EPCs were transfected with VEGFR2 cDNA or treated with FAK or c-Src activators overnight, then left untreated or were treated with VEGF (100 ng/mL) alone and in combination with melatonin (1 mM) for 24 h, before being examined by the Transwell (n = 4) and tube formation assays (n = 4). * p < 0.05 compared with the control group; # p < 0.05 compared with the VEGF-treated group.
Cells 12 00799 g005
Figure 6. Melatonin inhibits the VEGF-induced stimulation of EPC angiogenesis by inhibiting NF-κB and AP-1 activation. (A,B) EPCs were incubated with VEGF (100 ng/mL) and different concentrations of melatonin (0.1–1 mM) for 2 h. p65 and c-Jun phosphorylation was examined by western blot (n = 3). (C,D) EPCs were treated with NF-κB or AP-1 activators overnight, then left untreated or were treated with VEGF (100 ng/mL) alone and in combination with melatonin (1 mM) for 24 h, before being examined by the Transwell (n = 4) and tube formation assays (n = 4). (E) EPCs were transfected with the NF-κB or AP-1 luciferase plasmids and then treated with VEGF (100 ng/mL) and different concentrations of melatonin (0.1–1 mM) before determining luciferase activity (n = 4). EPCs were then treated with VEGF (100 ng/mL) and melatonin (1 mM), before undergoing (F) immunofluorescence staining with NF-κB and AP-1 antibodies, or (G,H) a ChIP assay (n = 3). * p < 0.05 compared with the control group; # p < 0.05 compared with the VEGF-treated group.
Figure 6. Melatonin inhibits the VEGF-induced stimulation of EPC angiogenesis by inhibiting NF-κB and AP-1 activation. (A,B) EPCs were incubated with VEGF (100 ng/mL) and different concentrations of melatonin (0.1–1 mM) for 2 h. p65 and c-Jun phosphorylation was examined by western blot (n = 3). (C,D) EPCs were treated with NF-κB or AP-1 activators overnight, then left untreated or were treated with VEGF (100 ng/mL) alone and in combination with melatonin (1 mM) for 24 h, before being examined by the Transwell (n = 4) and tube formation assays (n = 4). (E) EPCs were transfected with the NF-κB or AP-1 luciferase plasmids and then treated with VEGF (100 ng/mL) and different concentrations of melatonin (0.1–1 mM) before determining luciferase activity (n = 4). EPCs were then treated with VEGF (100 ng/mL) and melatonin (1 mM), before undergoing (F) immunofluorescence staining with NF-κB and AP-1 antibodies, or (G,H) a ChIP assay (n = 3). * p < 0.05 compared with the control group; # p < 0.05 compared with the VEGF-treated group.
Cells 12 00799 g006
Figure 7. Antiangiogenic effects of melatonin in the corneal alkali burn model. (A) Photos of a normal cornea and an alkali-burned cornea. Stereomicroscopic findings for eyes from mice (n = 6 per group) on days 1, 3, 5, and 7 after treatment with PBS, melatonin (20 mg/kg or 60 mg/kg), or bevacizumab (5 mg/μL). (B,C) At 7 days after the alkali burn injury, corneal stromal thickness was detected by H&E staining and quantified in mice with uninjured corneas, mice with untreated injured corneas, melatonin-treated mice, or bevacizumab-treated mice. (D,E) Levels of CD31, CD34, CD133 and PDGF-BB expression in corneas were subjected to co-immunofluorescence staining and quantified. * p < 0.05 compared with uninjured corneas; # p < 0.05 compared with damaged corneas.
Figure 7. Antiangiogenic effects of melatonin in the corneal alkali burn model. (A) Photos of a normal cornea and an alkali-burned cornea. Stereomicroscopic findings for eyes from mice (n = 6 per group) on days 1, 3, 5, and 7 after treatment with PBS, melatonin (20 mg/kg or 60 mg/kg), or bevacizumab (5 mg/μL). (B,C) At 7 days after the alkali burn injury, corneal stromal thickness was detected by H&E staining and quantified in mice with uninjured corneas, mice with untreated injured corneas, melatonin-treated mice, or bevacizumab-treated mice. (D,E) Levels of CD31, CD34, CD133 and PDGF-BB expression in corneas were subjected to co-immunofluorescence staining and quantified. * p < 0.05 compared with uninjured corneas; # p < 0.05 compared with damaged corneas.
Cells 12 00799 g007
Figure 8. The schematic diagram summarizes the proposed mechanism whereby melatonin suppresses VEGF-induced EPC angiogenesis in neovascular AMD. Melatonin suppresses VEGF-induced increases in the production of PDGF-BB, the recruitment of EPCs, and EPC angiogenesis by inhibiting VEGFR2, c-Src, FAK, NF-κB and AP-1 signaling.
Figure 8. The schematic diagram summarizes the proposed mechanism whereby melatonin suppresses VEGF-induced EPC angiogenesis in neovascular AMD. Melatonin suppresses VEGF-induced increases in the production of PDGF-BB, the recruitment of EPCs, and EPC angiogenesis by inhibiting VEGFR2, c-Src, FAK, NF-κB and AP-1 signaling.
Cells 12 00799 g008
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Lin, L.-W.; Wang, S.-W.; Huang, W.-C.; Huynh, T.K.; Lai, C.-Y.; Ko, C.-Y.; Fong, Y.-C.; Lee, J.-J.; Yang, S.-F.; Tang, C.-H. Melatonin Inhibits VEGF-Induced Endothelial Progenitor Cell Angiogenesis in Neovascular Age-Related Macular Degeneration. Cells 2023, 12, 799. https://doi.org/10.3390/cells12050799

AMA Style

Lin L-W, Wang S-W, Huang W-C, Huynh TK, Lai C-Y, Ko C-Y, Fong Y-C, Lee J-J, Yang S-F, Tang C-H. Melatonin Inhibits VEGF-Induced Endothelial Progenitor Cell Angiogenesis in Neovascular Age-Related Macular Degeneration. Cells. 2023; 12(5):799. https://doi.org/10.3390/cells12050799

Chicago/Turabian Style

Lin, Liang-Wei, Shih-Wei Wang, Wei-Chien Huang, Thanh Kieu Huynh, Chao-Yang Lai, Chih-Yuan Ko, Yi-Chin Fong, Jie-Jen Lee, Shun-Fa Yang, and Chih-Hsin Tang. 2023. "Melatonin Inhibits VEGF-Induced Endothelial Progenitor Cell Angiogenesis in Neovascular Age-Related Macular Degeneration" Cells 12, no. 5: 799. https://doi.org/10.3390/cells12050799

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop