Epidermal growth factor treatment has protective effects on the integrity of the blood‑brain barrier against cerebral ischemia injury in bEnd3 cells

  • Authors:
    • Shu Yang
    • Hong Jin
    • Zhi‑Gang Zhao
  • View Affiliations

  • Published online on: January 21, 2019     https://doi.org/10.3892/etm.2019.7186
  • Pages: 2397-2402
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Tight junctions (TJs) serve an important role in maintaining the integrity of the blood‑brain barrier (BBB), while neurological disorders, including ischemic stroke, induce TJ disruption and increase BBB permeability; results include edema formation and hemorrhage transformation. Cerebral endothelium protection presents a promising approach in ischemic stroke therapy. In the current study, protective effects of the epidermal growth factor (EGF) on ischemia‑induced disruption of BBB integrity were examined using an oxygen‑glucose deprivation (OGD) model in bEnd3 cells. Expression levels of claudin‑5 and TJ protein‑1 (ZO‑1) were determined by reverse transcription‑quantitative polymerase chain reaction and western blot analysis. Cell viability was evaluated by cell counting kit‑8 assay and the endothelial permeability of Lucifer yellow (LY) was assessed using Transwell assays. The results revealed that post‑ischemia administration of EGF (250 ng/ml) significantly attenuated the decrease in mRNA (P<0.05) and protein (P<0.01) expression levels of claudin‑5 and ZO‑1, and the increase in endothelial permeability of LY (P<0.05) induced by 4 h OGD exposure followed by 24 h reoxygenation. In addition, EGF did not significant affect cell viability. The current study suggested a potential of EGF to improve BBB integrity against ischemic injury by upregulating the expression of TJ proteins and reducing endothelial permeability.

Introduction

The blood-brain barrier (BBB) is a specialized tissue that restricts certain substances from entering the brain and protects normal neuronal function (1). The BBB is composed of cerebral endothelial cells, astrocytes and pericytes (2). Brain endothelial cells form the structural basis of the BBB and are characterized by the presence of tight junctions (TJs). TJs are composed of multiple protein complexes, including occludin, claudin-5 and TJ protein-1 (ZO-1) (3). Under normal conditions, TJs seal the gaps between endothelial cells and prevent the paracellular movement of molecules, wherein pathological insults caused by neurological disorders may lead to TJ disruption and BBB leakage, aggravating brain damage (4).

Ischemic stroke, which accounts for ~85% of all cases of stroke, is one of the most common diseases worldwide with a high disability and mortality rates (5). The obstruction of blood vessels reduces oxygen and glucose supply to the cerebral tissue and results in an energy failure, which triggers a cascade of events, including the release of proinflammatory cytokines, elevated levels of reactive oxygen species (ROS) and the activation of matrix metalloproteinases (MMPs) (6). These lead to TJ protein degradation, which can be exacerbated by thrombolytic therapy (7). Enhanced BBB permeability induces the occurrence of deleterious complications, including brain edema and hemorrhage transformation (8). Numerous compounds, including α-lipoic acid, ruscogenin and calycosin-7-O-β-D-glucoside, were reported to protect the ischemia-induced BBB injury through decreasing the generation of ROS or inhibiting the activity of MMPs (911). Therefore, targeting BBB integrity suggests a promising therapeutic approach for ischemic stroke treatment.

The epidermal growth factor (EGF) is a potent mitogen for mediating neuronal proliferation and inducing central nerve system (CNS) progenitor cells to produce astrocytes and neurons (12). EFG bioavailability in the brain is ensured through production in the CNS by glial cells and neurons and through uptake from the peripheral circulation (13). EGF has been examined as a neurotrophic factor against stroke in animal models in vivo (14). It also reduces BBB permeability by activating the EGF receptor and the downstream mitogen-activated protein kinase (MAPK) signaling pathway (15,16). However, further investigation is required as to whether EGF may preserve BBB integrity against ischemic insult.

In the current study, an in vitro oxygen-glucose deprivation (OGD) model was established based on bEnd3 cells, to explore the benefit of EGF on BBB integrity and protection against ischemic injury. TJ protein expression levels were measured in addition to endothelial permeability and cell viability, to demonstrate the protective effect of EGF against ischemic injury by improving BBB integrity.

Materials and methods

Materials

bEnd3 cells were obtained from the American Type Culture Collection (Manassas, VA, USA). Dulbecco's modified Eagle's medium (DMEM), fetal bovine serum (FBS), penicillin-streptomycin solution and Hank's balanced salt solution (HBSS) were purchased from HyClone (GE Healthcare Life Sciences, Logan, UT, USA). Lucifer yellow (LY) was purchased from Sigma-Aldrich (Merck KGaA, Darmstadt, Germany). Rabbit claudin-5 antibody (cat. no. ab15106) was obtained from Abcam (Cambridge, UK). Rabbit ZO-1 antibody (cat. no. 21773-1-AP) was purchased from ProteinTech Group, Inc. (Chicago, IL, USA). Mouse GAPDH antibody (cat. no. C1312), horseradish peroxide (HRP)-conjugated goat anti-rabbit (cat. no. C1309) and anti-mouse (cat. no. C1308) IgG antibodies, the Cell Counting kit-8 (CCK8) and the bicinchoninic acid (BCA) assay kit were obtained from Applygen Technologies, Inc. (Beijing, China). Polyvinylidene difluoride (PVDF) membranes and the chemiluminescent HRP substrate (cat. no. WBKLS0100) were purchased from EMD Millipore (Billerica, MA, USA). The 24-well polyester Transwell inserts (pore size, 0.4 µM; diameter, 6.5 mm) were obtained from Corning Inc. (Corning, NY, USA).

Cell culture and treatment

bEnd3 cells were cultured in DMEM supplemented with 10% FBS and 1% penicillin-streptomycin at 37°C with 5% CO2 at a density of 3×105/well in six-well plates. After 24 h, confluent bEnd3 cells were placed with glucose-free and serum-free DMEM (Beijing Solarbio Science & Technology Co., Ltd., Beijing, China) and incubated in an anaerobic chamber (1% O2) flushed with 95% N2 and 5% CO2 for 4, 6 and 12 h (10). For reoxygenation (RO), cells were subsequently returned to normoxic conditions and incubated in glucose-containing and serum-free standard DMEM for 24 h. For the control group, the bEnd3 cells were incubated in glucose-containing and serum-free standard DMEM medium for the same period of time under normoxic conditions for the entire duration of the experiment. EGF-administered groups were treated with mouse EGF (250 ng/ml; ProteinTech Group, Inc.) diluted with standard DMEM following a 4-h OGD period at the beginning of the RO process.

Cell viability

bEnd3 cells (5×103/well) were seeded in 96-well plates and incubated for 24 h prior to OGD treatment for 4, 6 and 12 h as detailed above. A total of 10 µl CCK8 solution was subsequently added to each well and incubated for 1 h at 37°C. Absorbance was measured at 450 nm using a plate reader. Changes in cell viability were expressed compared with respective normoxic groups.

Western blot analysis

Cells were harvested following RO and homogenized in radioimmunoprecipitation assay buffer (Applygen Technologies, Inc.) containing the protease inhibitor. The protein concentration was determined by BCA assays. Proteins (40 µg) were separated on 12 and 8% SDS-PAGE gels for claudin-5 and ZO-1 analysis, respectively, and transferred to PVDF membranes. Following blocking with 5% non-fat milk for 1 h at room temperature, membranes were incubated with primary antibodies of claudin-5 (dilution, 1:1,000), ZO-1 (dilution, 1:1,000) and GAPDH (dilution, 1:2,000) at 4°C overnight, followed by incubation with secondary antibodies (dilution, 1:5,000) for 1 h at room temperature. Protein bands were visualized by chemiluminescence detection using a FluorChem FC2 system (Cell Biosciences, Inc., Santa Clara, CA, USA) and band densities were quantified by Image J (version 1.43; National Institute of Health, Bethesda, MD, USA). Bands were normalized to GAPDH and expressed in comparison with respective normoxic controls.

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

Total RNA was extracted from bEnd3 cells following RO using TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc., Waltham, MA, USA) according to the manufacturer's protocol and quantified using a spectrophotometer. RNA (1 µg) was reverse-transcribed into cDNA using the PrimeScript™ RT reagent kit with gDNA Eraser (Takara Bio, Inc., Otsu, Japan) according to the manufacturer's protocol. qPCR was performed using the SYBR Premix Ex Taq II kit (Takara Bio, Inc.) according to the manufacturer's protocol using the following thermocycling conditions: 95°C for 30 sec, followed by 40 cycles of 95°C for 5 sec, 60°C for 20 sec and 72°C for 30 sec. Primer sequences were as follows: Claudin-5, forward, 5′-GTTAAGGCACGGGTAGCACT-3′ and reverse, 5′-TACTTCTGTGACACCGGCAC-3′; ZO-1, forward, 5′-AAGTTGGCAAGAGAGGAGCC-3′ and reverse, 5′-CAACCGCATTTGGCGTTACA-3′; GAPDH, forward, 5′-ACGTCTGCCACGATAACACC-3′ and reverse, 5′-CTGCATGATTGGGTCACGTC-3. GAPDH was used as internal control and mRNA expression levels were quantified using the 2−ΔΔCq method (17).

Permeability assay

bEnd3 cells (3.3×104/well) were seeded in Transwell inserts and incubated for 5 days at 37°C prior to performing the OGD. Following 4 h of OGD and 24 h of RO treatment, LY (50 µM) dissolved in HBSS was added to the upper compartment and the lower compartment contained blank HBSS. At indicated time points (15, 30, 45 and 60 min), samples were taken from the lower compartment and analyzed using a plate reader (excitation, 430 nM; emission, 540 nM). The permeability coefficient (Papp) was measured by the following equation: Papp=A/(SxC0), where A represents the rate of drug accumulated at the lower compartment, S represents the membrane area and C0 is the initial drug concentration at the upper compartment.

Statistical analysis

Data are expressed as the mean ± standard deviation, representative of three replicates. Differences between groups were assessed by one-way analysis of variance followed by Tukey's post hoc test using SPSS (version 19.0; IBM Corp., Armonk, NY, USA). P<0.05 was considered to indicate a statistically significant difference.

Results

Effects of OGD on TJ protein expression and cell viability

Claudin-5 is a key TJ protein that determines BBB permeability and integrity (18). As an indicator of ischemia-induced BBB disruption, claudin-5 expression was measured in bEnd3 cells, following exposure to OGD for various periods and 24 h RO. As presented in Fig. 1A, 4, 6 and 12 h OGD exposure resulted in a time-dependent decrease in claudin-5 protein expression compared with the normoxic controls. Cell viability analysis revealed no significant difference between the control and treatment groups following 4 and 6 h OGD exposure. In contrast, 12 h OGD exposure revealed a significant reduction of 45% in cell viability compared with the normoxic control (P<0.01; Fig. 1B).

EGF attenuates OGD-induced BBB disruption

To identify protective effects of EGF on ischemia-induced BBB disruption, EGF was added to bEnd3 cells immediately following 4 h OGD exposure prior to RO and subsequently TJ protein expression levels were measured. In addition, endothelial cell permeability of LY and cell viability were determined. As presented in Fig. 2A and B, 4 h OGD exposure caused a significant decrease in claudin-5 and ZO-1 protein expression compared with the control (71.3 and 41.2%, respectively; P<0.01), which was significantly ameliorated by EGF treatment (P<0.01 vs. OGD). However, the differences in the decrease of claudin-5 levels between the same groups in Fig. 1 (54.2%) and Fig. 2 (71.3%) may be attributed to the differential cell growth status and density prior to performing OGD in separate experiments. A significant decrease in mRNA expression by 61.5 and 52.1% was observed for claudin-5 and ZO-1 in the OGD group compared with the control group (P<0.01), while EGF significantly reversed this decrease (P<0.05; Fig. 2C). In addition, EGF treatment significantly attenuated an increase in endothelial permeability of LY following 4 h OGD exposure (P<0.05 vs. OGD; Fig. 2D). No significant differences were observed in cell viability following EGF treatment compared with the OGD and normoxic control groups (P>0.05; Fig. 2E). Results indicated that the protective effect of EGF on BBB integrity may be via enhancing TJ protein expression and decreasing BBB permeability.

Discussion

Cerebral endothelial cells are a crucial component of the neurovascular unit and are implicated in many biological functions, including regulating vascular tone, providing trophic support for neurons and mediating inflammatory responses (9). They also contribute to BBB integrity by forming TJs, which block the paracellular flux (19). However, neurological disorders, including ischemic stroke, damage the cerebral endothelium leading to TJ dysfunction and BBB leakage, which are associated with complications, including brain edema and hemorrhage transformation (20). Therefore, there is an increasing interest in the protection of BBB integrity in stroke therapy (21).

The current study established an in vitro OGD model using the mouse brain microvascular endothelial cell line bEnd3 to mimic clinical ischemic stroke (22,23). An increasing OGD exposure duration was observed to be associated with a decrease in claudin-5 protein expression, indicating a potential ischemia-induced BBB disruption. A 4 h OGD exposure was selected for subsequent studies as it closely reflected the limited therapeutic window of 4.5 h in ischemic stroke in a clinical setting (6). However, in contrast to previous studies, this duration of OGD exposure had no significant effect on cell viability. This may be attributed to the difference in oxygen levels during the OGD period, 1% here compared with 0.3% previously reported (24), which resulted in varying cell damage in the RO period.

The BBB phenotype of cerebral endothelial cells is regulated by the release of cytokines in the microenvironment (25). Glial-derived neurotrophic factor and basic fibroblast growth factor enhance TJ function, while proinflammatory cytokines, including interleukin-6 and tumor necrosis factor-α may damage BBB integrity (26). EGF has been reported to preserve endothelial integrity and protect TJ disruption against hydrogen peroxide (27). In the present study it was demonstrated that EGF upregulated the mRNA and protein expression of claudin-5 and ZO-1 and reduced the endothelial cell permeability of LY against ischemic insults in vitro. As OGD and EGF exerted no significant influence on cell viability, according to the conditions evaluated in the present study, the protective effect of EGF on BBB integrity was mainly attributed to the elevation in TJ protein expression levels. Furthermore, MAPK and protein kinase B (Akt) signaling, downstream pathways of EGF, serve important roles in cell proliferation, survival and apoptosis (28). Their activation participates in the regulation of TJ protein expression and TJ function (2931). Therefore, further investigation on whether EGF preserves BBB integrity against ischemic stroke by modulating the MAPK and Akt signaling pathways is required. Additionally, electron microscopy is an effective approach for observing BBB disruption at the ultrastructure level (32) and this method maybe explored in future BBB studies.

In summary, EGF ameliorated TJ protein degradation and inhibited increased BBB permeability induced by OGD treatment in bEnd3 cells. The aforementioned findings indicated a beneficial effect of EGF on BBB integrity against ischemic insult.

Acknowledgements

Not applicable.

Funding

The present study was supported by the National Natural Science Foundation of China (grant no. 81703376) and the Youth Foundation of Beijing Tiantan Hospital (grant no. 2016-YQN-11).

Availability of data and materials

The datasets used and/or analyzed during the present study are available from the corresponding author on reasonable request.

Authors' contributions

SY and ZZ designed the study. SY performed all experiments. SY and HJ interpreted the data and drafted the manuscript. ZZ critically reviewed and revised the manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Yang S, Mei S, Jin H, Zhu B, Tian Y, Huo J, Cui X, Guo A and Zhao Z: Identification of two immortalized cell lines, ECV304 and bEnd3, for in vitro permeability studies of blood-brain barrier. PLoS One. 12:e01870172017. View Article : Google Scholar : PubMed/NCBI

2 

Cecchelli R, Berezowski V, Lundquist S, Culot M, Renftel M, Dehouck MP and Fenart L: Modelling of the blood-brain barrier in drug discovery and development. Nat Rev Drug Discov. 6:650–661. 2007. View Article : Google Scholar : PubMed/NCBI

3 

Cardoso FL, Brites D and Brito MA: Looking at the blood-brain barrier: Molecular anatomy and possible investigation approaches. Brain Res Rev. 64:328–363. 2010. View Article : Google Scholar : PubMed/NCBI

4 

Ronaldson PT and Davis TP: Blood-brain barrier integrity and glial support: Mechanisms that can be targeted for novel therapeutic approaches in stroke. Curr Pharm Des. 18:3624–3644. 2012. View Article : Google Scholar : PubMed/NCBI

5 

Liu X, Zhu X, Chen M, Ge Q, Shen Y and Pan S: Resveratrol protects PC12 cells against OGD/R-induced apoptosis via the mitochondrial-mediated signaling pathway. Acta Biochim Biophys Sin (Shanghai). 48:342–353. 2016. View Article : Google Scholar : PubMed/NCBI

6 

Khatri R, McKinney AM, Swenson B and Janardhan V: Blood-brain barrier, reperfusion injury, and hemorrhagic transformation in acute ischemic stroke. Neurology. 79 (13 Suppl 1):S52–S57. 2012. View Article : Google Scholar : PubMed/NCBI

7 

Warach S and Latour LL: Evidence of reperfusion injury, exacerbated by thrombolytic therapy, in human focal brain ischemia using a novel imaging marker of early blood-brain barrier disruption. Stroke. 35 (11 Suppl 1):S2659–S2661. 2004. View Article : Google Scholar

8 

Zhu H, Wang Z, Xing Y, Gao Y, Ma T, Lou L, Lou J, Gao Y, Wang S and Wang Y: Baicalin reduces the permeability of the blood-brain barrier during hypoxia in vitro by increasing the expression of tight junction proteins in brain microvascular endothelial cells. J Ethnopharmacol. 141:714–720. 2012. View Article : Google Scholar : PubMed/NCBI

9 

Xie R, Li X, Ling Y, Shen C, Wu X, Xu W and Gao X: Alpha-lipoic acid pre- and post-treatments provide protection against in vitro ischemia-reperfusion injury in cerebral endothelial cells via Akt/mTOR signaling. Brain Res. 1482:81–90. 2012. View Article : Google Scholar : PubMed/NCBI

10 

Cao G, Jiang N, Hu Y, Zhang Y, Wang G, Yin M, Ma X, Zhou K, Qi J, Yu B and Kou J: Ruscogenin attenuates cerebral ischemia-induced blood-brain barrier dysfunction by suppressing TXNIP/NLRP3 inflammasome activation and the MAPK pathway. Int J Mol Sci. 17(pii): E14182016. View Article : Google Scholar : PubMed/NCBI

11 

Fu S, Gu Y, Jiang JQ, Chen X, Xu M, Chen X and Shen J: Calycosin-7-O-β-D-glucoside regulates nitric oxide/caveolin-1/matrix metalloproteinases pathway and protects blood-brain barrier integrity in experimental cerebral ischemia-reperfusion injury. J Ethnopharmacol. 155:692–701. 2014. View Article : Google Scholar : PubMed/NCBI

12 

Pillai DR, Shanbhag NC, Dittmar MS, Boqdahn U and Schlachetzki F: Neurovascular protection by targeting early blood-brain barrier disruption with neurotrophic factors after ischemia-reperfusion in rats*. J Cereb Blood Flow Metab. 33:557–566. 2013. View Article : Google Scholar : PubMed/NCBI

13 

Plata-Salamán CR: Epidermal growth factor and the nervous system. Peptides. 12:653–663. 1991. View Article : Google Scholar : PubMed/NCBI

14 

Wang YF, Cooke MJ, Sachewsky N, Morshead CM and Shoichet MS: Bioengineered sequential growth factor delivery stimulates brain tissue regeneration after stroke. J Control Release. 172:1–11. 2013. View Article : Google Scholar : PubMed/NCBI

15 

Liu W, Wang P, Shang C, Chen L, Cai H, Ma J, Yao Y, Shang X and Xue Y: Endophilin-1 regulates blood-brain barrier permeability by controlling ZO-1 and occludin expression via the EGFR-ERK1/2 pathway. Brain Res. 1573:17–26. 2014. View Article : Google Scholar : PubMed/NCBI

16 

Chen L, Liu W, Wang P, Xue Y, Su Q, Zeng C and Shang X: Endophilin-1 regulates blood-brain barrier permeability via EGFR-JNK signaling pathway. Brain Res. 1606:44–53. 2015. View Article : Google Scholar : PubMed/NCBI

17 

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

18 

Ronaldson PT and Davis TP: Targeting blood-brain barrier changes during inflammatory pain: An opportunity for optimizing CNS drug delivery. Ther Deliv. 2:1015–1041. 2011. View Article : Google Scholar : PubMed/NCBI

19 

Wilhelm I and Krizbai IA: In vitro models of the blood-brain barrier for the study of drug delivery to the brain. Mol Pharm. 11:1949–1963. 2014. View Article : Google Scholar : PubMed/NCBI

20 

Choi KH, Kim HS, Park MS, Kim JT, Kim JH, Cho KA, Lee MC, Lee HJ and Cho KH: Regulation of caveolin-1 expression determines early brain edema after experimental focal cerebral ischemia. Stroke. 47:1336–1343. 2016. View Article : Google Scholar : PubMed/NCBI

21 

Guo J, Krause DN, Horne J, Weiss JH, Li X and Duckles SP: Estrogen-receptor-mediated protection of cerebral endothelial cell viability and mitochondrial function after ischemic insult in vitro. J Cereb Blood Flow Metab. 30:545–554. 2010. View Article : Google Scholar : PubMed/NCBI

22 

Liu J, Jin X, Liu KJ and Liu W: Matrix metalloproteinase-2-mediated occludin degradation and caveolin-1-mediated claudin-5 redistribution contribute to blood-brain barrier damage in early ischemic stroke stage. J Neurosci. 32:3044–3057. 2012. View Article : Google Scholar : PubMed/NCBI

23 

Rahman NA, Rasil ANHM, Meyding-Lamade U, Craemer EM, Diah S, Tuah AA and Muharram SH: Immortalized endothelial cell lines for in vitro blood-brain barrier models: A systematic review. Brain Res. 1642:532–545. 2016. View Article : Google Scholar : PubMed/NCBI

24 

Ku JM, Taher M, Chin KY, Grace M, McIntyre P and Miller AA: Characterization of a mouse cerebral microvascular endothelial cell line (bEnd.3) after oxygen glucose deprivation and reoxygenation. Clin Exp Pharmacol Physiol. 43:777–786. 2016. View Article : Google Scholar : PubMed/NCBI

25 

Igarashi Y, Utsumi H, Chiba H, Yamada-Sasamori Y, Tobioka H, Kamimura Y, Furuuchi K, Kokai Y, Nakagawa T, Mori M and Sawada N: Glial cell line-derived neurotrophic factor induces barrier function of endothelial cells forming the blood-brain barrier. Biochem Biophys Res Commun. 261:108–112. 1999. View Article : Google Scholar : PubMed/NCBI

26 

Abbott NJ, Rönnbäck L and Hansson E: Astrocyte-endothelial interactions at the blood-brain barrier. Nat Rev Neurosci. 7:41–53. 2006. View Article : Google Scholar : PubMed/NCBI

27 

Basuroy S, Seth A, Elias B, Naren AP and Rao R: MAPK interacts with occludin and mediates EGF-induced prevention of tight junction disruption by hydrogen peroxide. Biochem J. 393:69–77. 2006. View Article : Google Scholar : PubMed/NCBI

28 

Park JH and Han HJ: Caveolin-1 plays important role in EGF-induced migration and proliferation of mouse embryonic stem cells: Involvement of PI3K/Akt and ERK. Am J Physiol Cell Physiol. 297:C935–C944. 2009. View Article : Google Scholar : PubMed/NCBI

29 

Campbell M, Collery R, McEvoy A, Gardiner TA, Stitt AW and Brankin B: Involvement of MAPKs in endostatin-mediated regulation of blood-retinal barrier function. Curr Eye Res. 31:1033–1045. 2006. View Article : Google Scholar : PubMed/NCBI

30 

Sumanasekera WK, Sumanasekera GU, Mattingly KA, Dougherty SM, Keynton RS and Klinge CM: Estradiol and dihydrotestosterone regulate endothelial cell barrier function after hypergravity-induced alterations in MAPK activity. Am J Physiol Cell Physiol. 293:C566–C573. 2007. View Article : Google Scholar : PubMed/NCBI

31 

Luo D, Zhao J and Rong J: Plant-derived triterpene celastrol ameliorates oxygen glucose deprivation-induced disruption of endothelial barrier assembly via inducing tight junction proteins. Phytomedicine. 23:1621–1628. 2016. View Article : Google Scholar : PubMed/NCBI

32 

Lu D, Mai HC, Liang YB, Xu BD, Xu AD and Zhang YS: Beneficial role of rosuvastatin in blood-brain barrier damage following experimental ischemic stroke. Front Pharmacol. 9:9262018. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2019
Volume 17 Issue 3

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yang S, Jin H and Zhao ZG: Epidermal growth factor treatment has protective effects on the integrity of the blood‑brain barrier against cerebral ischemia injury in bEnd3 cells. Exp Ther Med 17: 2397-2402, 2019
APA
Yang, S., Jin, H., & Zhao, Z. (2019). Epidermal growth factor treatment has protective effects on the integrity of the blood‑brain barrier against cerebral ischemia injury in bEnd3 cells. Experimental and Therapeutic Medicine, 17, 2397-2402. https://doi.org/10.3892/etm.2019.7186
MLA
Yang, S., Jin, H., Zhao, Z."Epidermal growth factor treatment has protective effects on the integrity of the blood‑brain barrier against cerebral ischemia injury in bEnd3 cells". Experimental and Therapeutic Medicine 17.3 (2019): 2397-2402.
Chicago
Yang, S., Jin, H., Zhao, Z."Epidermal growth factor treatment has protective effects on the integrity of the blood‑brain barrier against cerebral ischemia injury in bEnd3 cells". Experimental and Therapeutic Medicine 17, no. 3 (2019): 2397-2402. https://doi.org/10.3892/etm.2019.7186