Puerarin regulates neovascular glaucoma through pigment epithelium‑derived growth factor‑induced NF‑κB signaling pathway

  • Authors:
    • Hui‑Yu Wei
    • Ya‑Jie Zhang
    • Shao‑Zhen Zhao
  • View Affiliations

  • Published online on: March 28, 2018     https://doi.org/10.3892/mmr.2018.8800
  • Pages: 7866-7874
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Neovascular glaucoma is an ophthalmic disease and a potentially blinding secondary glaucoma caused by the formation of abnormal new blood vessels on the iris, which can prevent the normal drainage of water from the anterior segment of the eye. Evidence from China has suggested that puerarin benefits many diseases including myocardial infarction, stable angina, cerebral ischemia and glaucoma in a clinical setting. In the present study, the aim was to investigate the efficacies of puerarin on neovascular glaucoma in a mouse model. The molecular mechanism of puerarin‑mediated treatment for neovascular glaucoma was also investigated both in vitro and in vivo. Inflammatory responses in mice with neovascular glaucoma were analyzed by western blotting. Oxidative stress levels were investigated following treatment with puerarin in a mouse model of neovascular glaucoma. The results indicated that puerarin markedly improved growth of vascular endothelial cells. The present study reported that puerarin treatment markedly decreased interleukin (IL)‑1β, IL‑17A and tumor necrosis factor‑α expression levels in mice with neovascular glaucoma. It was found that puerarin significantly decreased oxidative stress levels by reducing reactive oxygen species, superoxide dismutase and malondialdehyde levels, as well as neuronal nitric oxide synthase (NOS) and inducible NOS expression levels. Results indicated that expression levels of pigment epithelium‑derived growth factor were significantly inhibited following treatment with puerarin. Mechanism analysis demonstrated that treatment with puerarin effectively inhibited nuclear factor (NF)‑κB activity and its target protein levels p65, inhibitor of NF‑κB kinase subunit β and inhibitor of NF‑κB kinase subunit α in vascular endothelial cells. Increasing endothelial‑derived growth factor (EDGF) expression levels could stimulate NF‑κB activity and abolish the inhibitory effects of puerarin. An animal study reported that puerarin treatment presented therapeutic effects for mice with neovascular glaucoma. Numbers of new vessels in iris were recovered to normal following puerarin treatment. In conclusion, these results indicated that puerarin treatment can inhibit inflammatory responses and oxidative stress, platelet‑derived growth factor (PDGF) expression and NF‑κB activity, suggesting puerarin may be a potential agent for the treatment of neovascular glaucoma through PDGF‑induced NF‑κB signaling pathway.

Introduction

Neovascular glaucoma is a serious ophthalmic disease that affects visual sense and leads to open-angle glaucoma and angle closure glaucoma (1). Neovascular glaucoma presents a potentially blinding secondary glaucoma caused by the formation of abnormal new blood vessels on iris, which can prevent normal drainage of aqueous liquid from the anterior segment of the eye (2,3). There are >40 different diseases that can lead to the cause of neovascular glaucoma, which are almost extensive involvement in posterior section of oxygen or localized front section of oxygen (4). Studies have suggested that central retinal vein occlusion, diabetic retinopathy and other diseases are the most common inducer of neovascular glaucoma (5,6). Mechanisms of the initiation and progression of neovascular glaucoma have been widely investigated and evidence indicates that imbalance of new vessels in iris area contributes to the initiation of neovascular glaucoma (7). Therefore, many theories have been proposed for explaining the neovascularization in the progression of neovascular glaucoma.

Currently, various therapeutic schedules for neovascular glaucoma have been put forward including physical and drug therapies (8). Physical treatment includes panrentinal photocoagulation, retina frozen and iris-cornea-angle photocoagulation (9). Drug treatments include hormonal drugs and antibody drugs (10,11). In previous years, a large numbers of traditional Chinese medicine have been investigated for the treatment of ophthalmic diseases and presented many benefits and encouraging comprehensive effects on hypertension in eyes (12). This study has stimulated scientists to investigate the mechanisms of traditional Chinese medicine-mediated treatments for human diseases. Treatments of neovascular glaucoma by using traditional Chinese medicine have been widely accepted and achieved clinical outcomes (13). In the current study, the authors investigated the efficacy of puerarin for the treatment of neovascular glaucoma in a mice model.

Puerarin, a natural flavonoid, is a major isoflavonoid compound extracted from Pueraria and exhibits potential therapeutic effects for many human diseases (14,15). Puerarin has been reported to have many benefits and medicinal properties (16). In addition, puerarin can attenuate amyloid-β-induced cognitive impairment through suppression of apoptosis in the rat hippocampus in vivo (17). A previous study reported that puerarin can prevent the rat liver against oxidative stress-mediated DNA damage and apoptosis induced by lead (18).

Furthermore, the synthesis of puerarin derivatives and their protective effect on the myocardial ischemia and reperfusion injury also have been examined in animal experiments (19). The present study design investigated the therapeutic effects of puerarin for the treatment of neovascular glaucoma in a mouse model and results indicated that puerarin significantly improved symptoms of neovascular glaucoma in a 30-day treatment period.

In the present study, the purpose was to research the efficacy of puerarin for neovascular glaucoma and potential mechanisms of puerarin-mediated signaling pathway in the process of neovascularization in the progression of neovascular glaucoma. In addition, the authors analyzed inflammatory responses and oxidative stress in mice with neovascular glaucoma. Notably, the authors also studied the NF-κB signaling pathway in vascular endothelial cells located on iris area in mice with neovascular glaucoma. The authors aimed to explain the mechanism by which puerarin contributes to prevention of neovascularization induced by hypoxia through inhibition of the pigment epithelium-derived growth factor-induced NF-κB signaling pathway.

Materials and methods

Ethical statement

The current study was approved by the ethics committee of Tianjin Medical University College of Optometry (Tianjin, China). All surgery and euthanasia were made to minimize suffering.

Cells culture and regents

Vascular endothelial cells on iris area were isolated from experimental mice and cultured in minimum essential medium (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) supplemented with 10% FBS (Invitrogen; Thermo Fisher Scientific, Inc., Waltham, MA, USA). Vascular endothelial cells were cultured in a 37°C humidified atmosphere of 5% CO2.

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) analysis

Total mRNA was obtained from vascular endothelial cells on iris area by using RNAeasy Mini kit (Qiagen, Inc., Valencia, CA, USA). Expression levels of nNOS and iNOS in vascular endothelial cells were determined by qRT-PCR using TaqPath™ 1-Step RT-qPCR Master Mix, CG (Applied Biosystems™) according to manufacturer's instructions (20). All the forward and reverse primers were synthesized by Invitrogen; Thermo Fisher Scientific, Inc. (nNOS forward, 5′-CGGCTGTGCTTTAATGGAGAT-3′ and reverse, 5′-GAGGAGACGCTGTTGAATCG-3′; iNOS forward, 5′-AAGGATGGGAAGTACTGG-3′ and reverse, 5′-CAGTGTTTTCCTCAGAAAGAG-3′; ROS forward, 5′-GAGCCTTCTGGTGAAAATCAA-3′ and reverse, 5′-ACGATTCAGCTCATCCTCCA-3′; β-actin forward, 5′-AGAGCTACGAGCTGCCTGA-3′ and reverse, 5′-AGCACTGTGTTGGCGTACAG-3′). The PCR conditions were 95°C for 15 min, and 95°C for 10 sec, followed by 40 cycles by 54°C for 20 sec and 72°C for 20 sec. Relative mRNA expression level changes were calculated by 2−ΔΔCq (21). The results are expressed as the n-fold way compared to control.

Western blotting

Vascular endothelial cells were on iris area isolated from experimental mice and homogenized in lysate buffer containing protease-inhibitor (Sigma-Aldrich; Merck KGaA) and were centrifuged at 8,000 × g at 4°C for 10 min. The supernatant of the mixture were used for analysis of the protein. 12.5% SDS-PAGE assays were performed as previously described (22). Proteins were transferred onto a polyvinylidene fluoride (PVDF) membrane (Merck Millipore). For western blotting, primary goat anti-mouse antibodies: p65 (1:1,000, ab32536), IKK-β (1:1,000, ab124957), IκBα (1:1,000, ab109300), vascular endothelial cell growth factor (VEGF; 1:1,000, ab32152), TGF-β (1:1,000, ab31013), angiogenin (1:1,000, ab139947), Neurotrophin 3 (1:1,000, ab53685) VSF (1:1,000, ab9695), IL-1β (1:1,000, ab200478), IL-17 (1:1,000, ab79056), IFN-γ (1:1,000, ab32152), SOD (1:1,000, ab137037), malondialdehyde (1:1,000, ab6463) and β-actin (1:1,000, ab8227) (all from Abcam, Cambridge, UK) were added following blocking (5% skimmed milk) for 60 min at 37°C. Following washing with PBS three times, the secondary rabbit anti-goat antibodies goat anti-rabbit IgG mAb (1:5,000, PV-6001, ZSGB-BIO; OriGene Technologies, Beijing, China) were used to detect purpose protein for 2 h at 37°C. The results were visualized by using enhanced chemiluminescence substrate ECL Select™ Ventana Benchmark automated staining system (Roche Diagnostics, Basel, Switzerland).

Animal study

A total of 20 female C57BL/6 mice (8 weeks, 25–30 g body weight) were purchased from The Jackson Laboratory (Bar Harbor, ME, USA) and housed with a 12 h light-dark artificial cycle. All mice were free to access food and water. To detect the efficacy of puerarin on neovascular glaucoma, mice model of neovascular glaucoma was established by mutation in collagen 8A2 according to a previous study (23). Mice with neovascular glaucoma were divided into two groups (n=10/group) and received treatment with puerarin (0.2 mg/kg) with PBS as negative control.

Immunohistochemical staining

Immunohistochemical staining was performed by an avidin-biotin-peroxidase technique. Paraffin-embedded tumor tissue sections were prepared and epitope retrieval was performed for further analysis. The paraffin sections were subjected with hydrogen peroxide (3%) for 10–15 min, which subsequently were blocked by a regular blocking solution (5% skimmed milk) for 10–15 min at 37°C. Finally, the sections were incubated in anti-p65 (1:1,000, ab32536), anti-IKK-β (1:1,000, ab124957), anti-IκBα (1:1,000, ab109300), anti-VEGF (1:1,000, ab32152), anti-TGF-β (1:1,000, ab31013) and anti-angiogenin (1:1,000, ab139947) (all from Abcam), respectively, at 4°C for 12 h following blocking. All sections were washed three times and incubated with secondary antibodies for 1 h at 37°C and were observed by six random fields of view under the microscope.

Activity of NF-κB

Activity of NF-κB in vascular endothelial cells from healthy mice and mice with neovascular glaucoma were analyzed following treatment with puerarin. The activity of NF-κB was conducted according to previous study (24).

Inflammatory response analysis

Inflammatory responses in serum of mice with neovascular glaucoma were analyzed after treatment with puerarin. Percentage of numbers of lymphocytes, monocytes and neutrophils were examined by flow cytometry according to a previous study (25).

Statistical analysis

All data are presented as mean ± standard error of the mean, are conducted in triplicate and performed using GraphPad Prism 5.0 (GraphPad Software, Inc., La Jolla, CA, USA). Statistical differences between experimental groups were analyzed by Student's t-test. P<0.05 was considered to indicate a statistically significant difference.

Results

Puerarin inhibits aberrant growth of vascular endothelial cells induced by vaso-stimulating factor in hypoxia

The authors first detected the effects of puerarin on vascular endothelial cells in vitro. The expression levels of vaso-stimulating factor (VSF) and vaso-formative factor (VFF) were upregulated in mice with neovascular glaucoma compared to healthy mice (Fig. 1A and B). In addition, as presented in Fig. 1C, VSF could induce vascular endothelial cells growth under hypoxic conditions, compared to normoxia. However, the authors found that puerarin significantly inhibited growth of vascular endothelial cells induced by VSF in hypoxia (Fig. 1D). Furthermore, it was observed that puerarin improved viability of vascular endothelial cells in normoxia (Fig. 1E). Moreover, results suggested that heparin-binding growth factors and VEGF mRNA expression levels were inhibited in vascular endothelial cells induced VSF (Fig. 1F). Taken together, these results suggested that puerarin can inhibit aberrant growth of vascular endothelial cells induced by VSF in hypoxia, which may be beneficial for neovascular glaucoma.

Puerarin suppresses expression levels inflammatory responses and factors in mice with neovascular glaucoma

The authors next analyzed the anti-inflammatory effects of puerarin in mice with neovascular glaucoma in vivo. Interleukin (IL)-1β, IL-17A and tumor necrosis factor (TNF)-α expression levels were increased in vascular endothelial cells induced by neovascular glaucoma, whereas puerarin treatment may significantly decreased upregulation of IL-1β, IL-17A and TNF-α in vitro (Fig. 2A-C). In addition, the authors observed that numbers of lymphocytes, monocytes and neutrophils were upregulated and puerarin could become downregulated in serum of mice with neovascular glaucoma (Fig. 2D-F). Furthermore, it was observed that interferon-γ and neurotrophic factor expression levels were increased in vascular endothelial cells in mice with neovascular glaucoma following treatment with puerarin (Fig. 2G and H). Collectively, the results suggested that puerarin can suppress expression levels inflammatory responses and factors in mice with neovascular glaucoma both in vitro and in vivo.

Puerarin attenuates oxidative stress in vascular endothelial cells and in mice with neovascular glaucoma

A previous study suggested that neovascular glaucoma is associated with oxidative stress (26). Therefore, the authors analyzed the effects of puerarin on changes of oxidative stress in vascular endothelial cells and in mice with neovascular glaucoma. As illustrated in Fig. 3A-C, puerarin significantly decreased oxidative stress levels by reducing reactive oxygen species (ROS), superoxide dismutase (SOD) and malondialdehyde levels in vascular endothelial cells in vitro. In addition, neuronal nitric oxide synthase (NOS) and inducible NOS mRNA expression levels were markedly decreased by puerarin in vascular endothelial cells (Fig. 3D and E). Furthermore, NADPH oxidase activity was improved by puerarin in vascular endothelial cells in mice with neovascular glaucoma compared to the control (Fig. 3F). Collectively, the results suggested that puerarin can attenuate oxidative stress in vascular endothelial cells and in mice with neovascular glaucoma.

Puerarin regulates aberrant growth of vascular endothelial cells through platelet derived growth factor (PDGF)-induced NF-κB signaling pathway. In order to identify the mechanism of puerarin-mediated neovascular glaucoma, the authors analyzed the NF-κB signaling pathway and the target proteins in vascular endothelial cells. As presented in Fig. 4A-C, puerarin markedly inhibited expression levels of p65, inhibitor of NF-κB kinase subunit β (IKK-β) and inhibitor of NF-κB kinase subunit α (IκBα) in vascular endothelial cells. In addition, NF-κB activity was inhibited by puerarin treatment in vascular endothelial cells (Fig. 4D). Puerarin decreased PDGF expression, and restoration of PDGF expression abolished puerarin-inhibited NF-κB activity (Fig. 4E). In addition, PDGF restoration abolished the beneficial effects of puerarin on inflammatory responses and oxidative stress (Fig. 4F and G). Furthermore, PDGF restoration also promoted TGF-β and VEGF expression levels (Fig. 4H). These results suggested that puerarin can regulate aberrant growth of vascular endothelial cells through the PDGF-induced NF-κB signaling pathway.

Puerarin shows inhibition of aberrant neovascularization in mice with neovascular glaucoma

To analyze the therapeutic effects of puerarin for neovascular glaucoma, a mouse model of neovascular glaucoma was established by mutation in collagen 8A2 (23). Intraocular pressure of experimental mice was measured for examine the therapeutic efficacy of puerarin. As demonstrated in Fig. 5A, intraocular pressure was significantly improved by puerarin treatment. It was also observed aberrant vascular proliferation was inhibited in mice with neovascular glaucoma treated by puerarin (Fig. 5B). In addition, fluorescence staining showed that p65, IKK-β and IκBα expression levels were significantly downregulated by puerarin treatment in vascular endothelial cells (Fig. 5C). Furthermore, the results indicated that expression levels of VEGF, TGF-β and angiogenin were downregulated in vascular endothelial cells in puerarin-treat mice (Fig. 5D). Histological analysis indicated that neovascularization and retinal degeneration were improved in mice with neovascular glaucoma following treatment with puerarin (Fig. 5E and F). Importantly, visual function and retinal whole-mount analysis suggested that puerarin could markedly improve visual function and pathological hemorrhages in mice with neovascular glaucoma (Fig. 5G and H). Taken together, these results puerarin presented benefits for the inhibition of aberrant neovascularization and improves pathological symptoms in mice with neovascular glaucoma.

Discussion

Neovascular glaucoma is a disease that new fiber vascular membrane is overgrowth of the iris and trabecular, which leads to fibrous thickening of the filtration membrane in the operation area and the peripheral anterior synechia of the iris area (27). Neovascular glaucoma is also known as hemorrhagic glaucoma due to new blood vessels rupture and recurrent anterior chamber bleeding, which has been considered as intractable ophthalmic diseases (28). Therefore, treatments for neovascular glaucoma has become a nightmare for patients in clinical. Although many treatments have proposed for patients with neovascular glaucoma, the efficacy is limited for patients and the recurrence rate is higher following patients receiving clinical treatments (29). In the present study, the authors introduced an efficient drug of puerarin for the treatment of neovascular glaucoma in an animal model. Not only have the therapeutic effects of neovascular glaucoma been investigated, but also elaborated the molecular mechanism of puerarin-mediated improvement of neovascular glaucoma. Our results suggested that puerarin can inhibit aberrant growth of vascular endothelial cells, suppress inflammatory responses and factors and oxidative stress in vascular endothelial cells and in mice with neovascular glaucoma. Importantly, the findings indicated that puerarin regulates growth of vascular endothelial cells, neovascularization and retinal degeneration through PDGF-induced NF-κB signaling pathway in mice with neovascular glaucoma.

Aberrant growth of vascular endothelial cells contributes to the deterioration of vascular proliferation and visual function for patients with neovascular glaucoma (30). As is known, inhibition of aberrant growth of vascular endothelial cells is a potential treatment for patients with neovascular glaucoma (9). In previous years, anti-vascular endothelial growth factor for neovascular glaucoma therapy has been prevalent and achieves efficacy for patients with neovascular glaucoma (31). However, long-term treatment of anti-vascular endothelial growth factor obviously reduces pharmacodynamics of the antibody (32). The outcomes suggested that puerarin can inhibit the aberrant growth of vascular endothelial cells and avoid the defects of anti-vascular endothelial growth factor antibody, which presents an ideal agent in anti-glaucoma therapy.

Inflammatory responses have been identified associated with the initiation and progression of glaucoma (33). Chang et al (34) suggested increasing mast cell numbers as a possible indicator of preoperative glaucoma surgery inflammation was observed in the conjunctiva of glaucoma patients. In addition, Furtado et al (35) have showed that inflammation in patients under topical glaucoma is an indicator following treatment with indication to surgery. Vohra et al (36) have indicated that the role of inflammation in the pathogenesis of glaucoma. Furthermore, evidences have indicated that other factors such as genetics, excitotoxicity and blood flow can act as potential causal factors for progressive of glaucoma (37). The current results demonstrated that puerarin could inhibit inflammatory responses in mice with neovascular glaucoma. These inhibitory effects of IL-1β, IL-17A, TNF-α, lymphocytes, monocytes and neutrophils on experimental mice are beneficial for the recovery of neovascular glaucoma. Interestingly, the authors also observed that oxidative stress is downregulated by puerarin treatment.

Previous studies have reported that oxidative-antioxidative balance is associated with progression of neovascular glaucoma and that the resulting oxidative stress constitutes a major contribution involved in the pathophysiology of neovascular glaucoma (38). Research has indicated that retinal pigment epithelial damage induces more production of PDGF and breaks the blood-retinal barrier, and retinal inflammation in dogs with primary glaucoma (39). Oxidative stress can regulate the activity of complement in human glaucoma (40). These results simultaneously suggested that decreasing of oxidative stress in vascular endothelial cells may be contributed to the inhibition of aberrant growth of vascular endothelial cells. The results revealed that puerarin can markedly suppress ROS, SOD and malondialdehyde levels in vascular endothelial cells in experimental mice. Downregulation of oxidative stress contributes to the decreasing of intraocular pressure. Notably, this downregulation is regulated by the NF-κB signaling pathway in mice with neovascular glaucoma.

The NF-κB signaling pathway involves in various processes of neovascular glaucoma. In the present study, the authors investigated the relationships between the NF-κB pathway and neovascular glaucoma in a mouse model following treatment with puerarin. A previous study has suggested that the NF-κB pathway can regulate the activation of NLRP3 and caspase-8 inflammasomes in acute glaucoma (41). In addition, NF-κB-mediated inflammatory stress response may be predictor of the glaucoma marker SELE in trabecular meshwork cells (42). In the present study, puerarin treatment significantly inhibits NF-κB expression and activity in vascular endothelial cells in experimental mice. Stimulation of NF-κB activity canceled the inhibitory effects of puerarin for inflammatory responses and oxidative stress.

In conclusion, the study investigated the efficacy and potential mechanism of puerarin-mediated improvement of neovascular glaucoma in a mice model. Puerarin treatment not only inhibits aberrant growth of vascular endothelial cells, inflammatory responses and oxidative stress, but also improves neovascularization, retinal degeneration and visual function in mice with neovascular glaucoma. Most importantly, the NF-κB pathway is involved in puerarin-inhibited inflammatory responses and oxidative stress is observed in vitro and in vivo. These outcomes suggested that puerarin is beneficial for the treatment of neovascular glaucoma through regulation of the PDGF-induced NF-κB signaling pathway.

References

1 

Ng E, Choong AM and Walker PJ: Neovascular glaucoma post carotid endarterectomy: A case report and review of the literature. Ann Vasc Dis. 8:103–105. 2015. View Article : Google Scholar : PubMed/NCBI

2 

Kiddee W, Tantisarasart T and Wangsupadilok B: Neovascular glaucoma: A retrospective review of 5-year experience in Songklanagarind Hospital. J Med Assoc Thai. 95 Suppl 4:S36–S42. 2012.PubMed/NCBI

3 

Martinez-Carpio PA, Bonafonte-Márquez E, Heredia-Garcia CD and Bonafonte-Royo S: Efficacy and safety of intravitreal injection of bevacizumab in the treatment of neovascular glaucoma: Systematic review. Arch Soc Esp Oftalmol. 83:579–588. 2008.(In Spanish). View Article : Google Scholar : PubMed/NCBI

4 

Takihara Y, Inatani M, Fukushima M, Iwao K, Iwao M and Tanihara H: Trabeculectomy with mitomycin C for neovascular glaucoma: Prognostic factors for surgical failure. Am J Ophthalmol. 147(912–918): e12009.

5 

Mishra KK, Daftari IK, Weinberg V, Cole T, Quivey JM, Castro JR, Phillips TL and Char DH: Risk factors for neovascular glaucoma after proton beam therapy of uveal melanoma: A detailed analysis of tumor and dose-volume parameters. Int J Radiat Oncol Biol Phys. 87:330–336. 2013. View Article : Google Scholar : PubMed/NCBI

6 

Goto A, Inatani M, Inoue T, Awai-Kasaoka N, Takihara Y, Ito Y, Fukushima M and Tanihara H: Frequency and risk factors for neovascular glaucoma after vitrectomy in eyes with proliferative diabetic retinopathy. J Glaucoma. 22:572–576. 2013. View Article : Google Scholar : PubMed/NCBI

7 

Calugaru D and Calugaru M: Neovascular glaucoma-etipathogeny and diagnosis. Oftalmologia. 56:3–14. 2012.(In Romanian). PubMed/NCBI

8 

Rachitskaya AV, Lee RK, Dubovy SR and Schiff ER: Combined central retinal vein and central retinal artery occlusions and neovascular glaucoma associated with interferon treatment. Eur J Ophthalmol. 22:284–287. 2012. View Article : Google Scholar : PubMed/NCBI

9 

Altintas AG, Arifoglu HB, Tutar E, Koklu G and Ozcan PY: Effect on anterior chamber bevacizumab injection combined with seton implantation in treatment of rubeosis iridis in neovascular glaucoma. Cutan Ocul Toxicol. 31:124–127. 2012. View Article : Google Scholar : PubMed/NCBI

10 

Caujolle JP, Maschi C, Freton A, Pages G and Gastaud P: Treatment of neovascular glaucoma after proton therapy for uveal melanomas with ranibizumab injection: Preliminary results. Ophthalmic Res. 47:57–60. 2012. View Article : Google Scholar : PubMed/NCBI

11 

Călugăru D and Călugăru M: Treatment of neovascular glaucoma. Oftalmologia. 56:20–39. 2012.(In Romanian).

12 

Zhou WY and Li YH: A survey on treatment of dry eye by traditional chinese medicine and integrative chinese and Western medicine. Chin J Integr Med. 12:154–159. 2006. View Article : Google Scholar : PubMed/NCBI

13 

Mi XS, Zhong JX, Chang RC and So KF: Research advances on the usage of traditional Chinese medicine for neuroprotection in glaucoma. J Integr Med. 11:233–240. 2013. View Article : Google Scholar : PubMed/NCBI

14 

Zhang HY, Yi XN, Liu YH, Lao ML and Zhang XF: The protective effect of puerarin on Abeta(25–35)-induced PC12 cell injury. Zhong Yao Cai. 33:763–767. 2010.(In Chinese). PubMed/NCBI

15 

Zhang W, Liu CQ, Wang PW, Sun SY, Su WJ, Zhang HJ, Li XJ and Yang SY: Puerarin improves insulin resistance and modulates adipokine expression in rats fed a high-fat diet. Eur J Pharmacol. 649:398–402. 2010. View Article : Google Scholar : PubMed/NCBI

16 

Wang L and Zhao Y: Effect of decoction method on pharmacokinetics of puerarin in dogs. Zhong Yao Cai. 33:1442–1444. 2010.(In Chinese). PubMed/NCBI

17 

Li J, Wang G, Liu J, Zhou L, Dong M, Wang R, Li X, Li X, Lin C and Niu Y: Puerarin attenuates amyloid-beta-induced cognitive impairment through suppression of apoptosis in rat hippocampus in vivo. Eur J Pharmacol. 649:195–201. 2010. View Article : Google Scholar : PubMed/NCBI

18 

Liu CM, Ma JQ and Sun YZ: Puerarin protects the rat liver against oxidative stress-mediated DNA damage and apoptosis induced by lead. Exp Toxicol Pathol. 64:575–582. 2012. View Article : Google Scholar : PubMed/NCBI

19 

Feng ZQ, Wang YY, Guo ZR, Chu FM and Sun PY: The synthesis of puerarin derivatives and their protective effect on the myocardial ischemia and reperfusion injury. J Asian Nat Prod Res. 12:843–850. 2010. View Article : Google Scholar : PubMed/NCBI

20 

Xiao S, Wang J and Xiao N: MicroRNAs as noninvasive biomarkers in bladder cancer detection: A diagnostic meta-analysis based on qRT-PCR data. Int J Biol Markers. 31:e276–e285. 2016. View Article : Google Scholar : PubMed/NCBI

21 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

22 

Wai-Hoe L, Wing-Seng L, Ismail Z and Lay-Harn G: SDS-PAGE-based quantitative assay for screening of kidney stone disease. Biol Proced Online. 11:145–160. 2009. View Article : Google Scholar : PubMed/NCBI

23 

Steinhart MR, Cone FE, Nguyen C, Nguyen TD, Pease ME, Puk O, Graw J, Oglesby EN and Quigley HA: Mice with an induced mutation in collagen 8A2 develop larger eyes and are resistant to retinal ganglion cell damage in an experimental glaucoma model. Mol Vis. 18:1093–1106. 2012.PubMed/NCBI

24 

Tanaka T, Imamura T, Yoneda M, Irie A, Ogi H, Nagata M, Yoshida R, Fukuma D, Kawahara K, Shinohara M and Nakayama H: Enhancement of active MMP release and invasive activity of lymph node metastatic tongue cancer cells by elevated signaling via the TNF-α-TNFR1-NF-kB pathway and a possible involvement of angiopoietin-like 4 in lung metastasis. Int J Oncol. 49:1377–1384. 2016. View Article : Google Scholar : PubMed/NCBI

25 

Ao X, Sellati TJ and Stenken JA: Enhanced microdialysis relative recovery of inflammatory cytokines using antibody-coated microspheres analyzed by flow cytometry. Anal Chem. 76:3777–3784. 2004. View Article : Google Scholar : PubMed/NCBI

26 

Awodele O, Oreagba IA, Olayemi SO, Oladipo I, Iruegbukpe CO, Balogun BG, Balogun MM and Adedokun AO: Evaluation and comparison of the indices of systemic oxidative stress among Black-africans with Age-related cataracts or primary glaucoma. Middle East Afr J Ophthalmol. 22:489–494. 2015. View Article : Google Scholar : PubMed/NCBI

27 

Jeon S, Lee NY and Park CK: Neovascular glaucoma following stereotactic radiosurgery for an optic nerve glioma: A case report. Korean J Ophthalmol. 24:252–255. 2010. View Article : Google Scholar : PubMed/NCBI

28 

Widder RA, Lemmen KD and Dietlein TS: Neovascular glaucoma. Klin Monbl Augenheilkd. 227:R15–R27. 2010.(In German). View Article : Google Scholar : PubMed/NCBI

29 

Toyama S, Tsuji H, Mizoguchi N, Nomiya T, Kamada T, Tokumaru S, Mizota A, Ohnishi Y and Tsujii H: Working Group for Ophthalmologic Tumors: Long-term results of carbon ion radiation therapy for locally advanced or unfavorably located choroidal melanoma: Usefulness of CT-based 2-port orthogonal therapy for reducing the incidence of neovascular glaucoma. Int J Radiat Oncol Biol Phys. 86:270–276. 2013. View Article : Google Scholar : PubMed/NCBI

30 

Wittström E, Holmberg H, Hvarfner C and Andréasson S: Clinical and electrophysiologic outcome in patients with neovascular glaucoma treated with and without bevacizumab. Eur J Ophthalmol. 22:563–574. 2012. View Article : Google Scholar : PubMed/NCBI

31 

Laplace O: Surgical session: Neovascular glaucoma and anti-vascular endothelial growth factor treatment. J Fr Ophtalmol. 32:230–235. 2009.(In French). View Article : Google Scholar : PubMed/NCBI

32 

Simha A, Braganza A, Abraham L, Samuel P and Lindsley K: Anti-vascular endothelial growth factor for neovascular glaucoma. Cochrane Database Syst Rev: CD007920. 2013. View Article : Google Scholar

33 

Reilly CM, Morris R and Dubielzig RR: Canine goniodysgenesis-related glaucoma: A morphologic review of 100 cases looking at inflammation and pigment dispersion. Vet Ophthalmol. 8:253–258. 2005. View Article : Google Scholar : PubMed/NCBI

34 

Chang L, Wong T, Ohbayashi M, Bunce C, Barton K, Ono SJ and Khaw PT: Increased mast cell numbers in the conjunctiva of glaucoma patients: A possible indicator of preoperative glaucoma surgery inflammation. Eye (Lond). 23:1859–1865. 2009. View Article : Google Scholar : PubMed/NCBI

35 

Furtado JM, Paula JS, Soares EG, Dhegaide NH, Rocha EM, Donadi E and Mde Rodrigues L: Conjunctival inflammation in patients under topical glaucoma treatment with indication to surgery. Acta Cir Bras. 27:732–735. 2012. View Article : Google Scholar : PubMed/NCBI

36 

Vohra R, Tsai JC and Kolko M: The role of inflammation in the pathogenesis of glaucoma. Surv Ophthalmol. 58:311–320. 2013. View Article : Google Scholar : PubMed/NCBI

37 

Ha Y, Liu H, Xu Z, Yokota H, Narayanan SP, Lemtalsi T, Smith SB, Caldwell RW, Caldwell RB and Zhang W: Endoplasmic reticulum stress-regulated CXCR3 pathway mediates inflammation and neuronal injury in acute glaucoma. Cell Death Dis. 6:e19002015. View Article : Google Scholar : PubMed/NCBI

38 

Schlötzer-Schrehardt U: Oxidative stress and pseudoexfoliation glaucoma. Klin Monbl Augenheilkd. 227:108–113. 2010.(In German). View Article : Google Scholar : PubMed/NCBI

39 

Mangan BG, Al-Yahya K, Chen CT, Gionfriddo JR, Powell CC, Dubielzig RR, Ehrhart EJ and Madl JE: Retinal pigment epithelial damage, breakdown of the blood-retinal barrier, and retinal inflammation in dogs with primary glaucoma. Vet Ophthalmol. 10 Suppl 1:S117–S124. 2007. View Article : Google Scholar

40 

Tezel G, Yang X, Luo C, Kain AD, Powell DW, Kuehn MH and Kaplan HJ: Oxidative stress and the regulation of complement activation in human glaucoma. Invest Ophthalmol Vis Sci. 51:5071–5082. 2010. View Article : Google Scholar : PubMed/NCBI

41 

Chi W, Chen H, Li F, Zhu Y, Yin W and Zhuo Y: HMGB1 promotes the activation of NLRP3 and caspase-8 inflammasomes via NF-kB pathway in acute glaucoma. J Neuroinflammation. 12:1372015. View Article : Google Scholar : PubMed/NCBI

42 

Itakura T, Peters DM and Fini ME: Glaucomatous MYOC mutations activate the IL-1/NF-kB inflammatory stress response and the glaucoma marker SELE in trabecular meshwork cells. Mol Vis. 21:1071–1084. 2015.PubMed/NCBI

Related Articles

Journal Cover

June-2018
Volume 17 Issue 6

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wei HY, Zhang YJ and Zhao SZ: Puerarin regulates neovascular glaucoma through pigment epithelium‑derived growth factor‑induced NF‑κB signaling pathway. Mol Med Rep 17: 7866-7874, 2018
APA
Wei, H., Zhang, Y., & Zhao, S. (2018). Puerarin regulates neovascular glaucoma through pigment epithelium‑derived growth factor‑induced NF‑κB signaling pathway. Molecular Medicine Reports, 17, 7866-7874. https://doi.org/10.3892/mmr.2018.8800
MLA
Wei, H., Zhang, Y., Zhao, S."Puerarin regulates neovascular glaucoma through pigment epithelium‑derived growth factor‑induced NF‑κB signaling pathway". Molecular Medicine Reports 17.6 (2018): 7866-7874.
Chicago
Wei, H., Zhang, Y., Zhao, S."Puerarin regulates neovascular glaucoma through pigment epithelium‑derived growth factor‑induced NF‑κB signaling pathway". Molecular Medicine Reports 17, no. 6 (2018): 7866-7874. https://doi.org/10.3892/mmr.2018.8800