Cadmium exposure enhances VE‑cadherin expression in endothelial cells via suppression of ROCK signaling

  • Authors:
    • Xiaorui Li
    • Xiao Li
    • Rong Sun
    • Mei Gao
    • Hui Wang
  • View Affiliations

  • Published online on: March 28, 2022     https://doi.org/10.3892/etm.2022.11282
  • Article Number: 355
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Vascular endothelium is a target of cadmium (Cd), which is a global pollutant of the environment. However, the detailed effects and underlying mechanisms remain to be elucidated. In the present study, human umbilical vein endothelial cells (HUVECs) were treated with 0.1, 1, 5, 10, 50 µM cadmium chloride (CdCl2) for 12 h. It was found that vascular endothelial (VE)‑cadherin mRNA and protein expression was upregulated by Cd in HUVECs in a dose‑dependent manner. Higher levels of VE‑cadherin were detected at cell‑to‑cell junctions in HUVECs treated with 10 µM CdCl2 compared with normal condition. The phosphorylation level of myosin‑binding subunit of myosin phosphatase, a downstream substrate of Rho‑associated protein kinase (ROCK), was reduced by 10 µM CdCl2, suggesting that Cd inhibited the Rho/ROCK pathway. Activation of ROCK by narciclasine reversed the Cd‑induced increase of VE‑cadherin expression. By contrast, ROCK pathway inhibitor Y27632 increased VE‑cadherin expression in HUVECs. Following inhibition of the ROCK pathway, Cd did not significantly alter the level of VE‑cadherin. Taken together, the results suggested that Cd exposure enhanced VE‑cadherin expression in endothelial cells via suppression of ROCK signaling.

Introduction

Cadmium (Cd) is one of the global pollutants of the environment (1). Tobacco smoke, Cd-contaminated food and industrial contamination are the main sources of Cd toxicity in humans (2,3). Chronic exposure to Cd has a major effect on humans, causing damage to multiple organs including kidneys, liver, lung, pancreas and testes (3-5). In addition, studies have reported that the vascular endothelial cell (VEC) is another main target of Cd (6,7). Accumulated Cd impairs endothelial function at a variety of molecular levels, including cell adhesion molecules, metal ion transporters and protein kinase signaling pathways (7,8). As blood vessels are widely distributed in organs, functional damage of VECs further causes toxicity in the parenchymal cells of human organs (8,9). Therefore, it is of great significance to investigate the underlying mechanism of Cd toxicity on VECs.

Vascular endothelial (VE) cadherin, a principal adhesion protein, is composed of a large ectodomain with five cadherin repeats, a transmembrane domain and a cytoplasmic tail (10). Cell-to-cell adhesion is established by interactions of ectodomain of VE-cadherin (11). The cytoplasmic tail of VE-cadherin is regulated by a range of phosphatases and kinases (12). Phosphorylation of the VE-cadherin cytoplasmic tail leads to VE-cadherin dissociation from the junctions (11). VE-cadherin is produced by various stimuli to regulate vascular permeability (13-16). It has been reported that VE-cadherin mRNA expression is reduced in chick embryo model treated with 50 µM Cd (17). In addition, low concentration of Cd induces membrane dissociation of VE-cadherin in human renal glomerular endothelial cells and human umbilical vein endothelial cells (HUVECs) (18,19). Further research is required on the effect of Cd on VE-cadherin.

Rho is a member of guanosine triphosphatase (GTPase) family (20). Rho-associated coiled-coil kinase (ROCK), including ROCK1 and ROCK2, is a serine/threonine kinase downstream of Rho GTPases (20). ROCK pathway regulates various cellular functions, including contraction, cytoskeleton organization, cell-to-cell adhesion and permeability (21). Lipopolysaccharide has been reported to activate the RhoA/ROCK signaling pathway to weaken cell junctions by reducing the expression of VE-cadherin and altering distribution of VE-cadherin (22). However, whether Cd regulates VE-cadherin by ROCK pathway is uncertain.

In the present study, the results showed that Cd increased VE-cadherin expression in HUVECs in a dose-dependent manner. It also found that 10 µM Cd inhibited ROCK pathway. Narciclasine, an activator of ROCK pathway, reversed Cd-induced VE-cadherin expression. In addition, the expression of VE-cadherin is unchanged in HUVECs treated with 10 µM narciclasine in the absence or presence of 10 µM CdCl2. ROCK pathway inhibitor Y27632 increased VE-cadherin expression in HUVECs in a dose-dependent manner. With pretreatment of 20 µM Y27632, 10 µM Cd did not alter VE-cadherin expression. The present study demonstrated a role for the ROCK pathway in regulating the VE-cadherin expression induced by Cd in HUVECs.

Materials and methods

Reagents and antibodies

CdCl2 was purchased from Millipore Sigma and dissolved in phosphate buffered saline (PBS). Narciclasine (MedChem Express) and Y27632 (Selleck Chemicals) were dissolved in dimethyl sulfoxide (DMSO). The primary antibody against VE-cadherin was purchased from Abcam. The primary antibodies against myosin phosphatase-targeting subunit (MYPT), phosphorylated (p)-MYPT (Ser507) and GAPDH were purchased from Cell Signaling Technology, Inc. The secondary antibody was goat anti-rabbit IgG (cat. no. 7074; Cell Signaling Technology, Inc.).

Cell Culture

HUVECs were purchased from American Type Culture Collection and maintained in Dulbecco's modified Eagle's medium (DMEM) supplemented with 10% fetal bovine serum (FBS; Lonza Group Ltd.), 10% FBS, 100 U/ml penicillin and 100 µg/ml streptomycin at 37˚C in 5% CO2.

Reverse transcription-quantitative (RT-q) PCR

HUVECs were seeded at a density of 5x105 cells/ml in a 65-mm dish with different treatment. RNA extraction, cDNA synthesis and qPCR were performed according to the manufacturer's protocols. Total RNA from the treated HUVECs was isolated with the E.Z.N.A. Total RNA kit II (Omega Bio-Tek, Inc.). Complementary cDNA was synthesized using the RevertAid First strand cDNA Synthesis kit (Thermo Fisher Scientific, Inc.). Diluted cDNA (4.6 µl; 50 ngcDNA) and 5.4 µl of primer and supermix mixture (SYBR premix Ex Tap™ II and dH2O; Takara Biotechnology Co., Ltd.) were used in each RT-qPCR reaction. The RT-qPCR process was performed on the CFX96 Real-Time System (Bio-Rad Laboratories, Inc.). Reaction conditions were: 95˚C for 5 min, 40 cycles of 95˚C for 10 sec and 60˚C for 32 sec. All PCR reactions were repeated three times and the mRNA levels were normalized to β-actin. Relative quantitative values were obtained from cycle threshold (Ct) and the 2-∆∆Cq method (23). The human VE-cadherin PCR primers were 5'-CAGCCCAAAGTGTGTGAGAA-3' (sense) and 5'-CGGTCAAACTGCCCATACTT-3' (antisense). The human β-actin PCR primers were 5'-TTGCCGACAGGATGCAGAA-3' (sense) and 5'-GCCGATCCACACGGAGTACT-3' (antisense).

Western blotting

Western blotting was performed as previously described (24). Cells were homogenized with in RIPA buffer at 4˚C. Protein concentrations were measured using Pierce BCA Assay kit (Thermo Fisher Scientific, Inc.). Supernatants of cell lysates were mixed with loading buffer and heated at 95˚C for 5 min. Samples containing 25 µg protein was separated by 10% SDS-PAGE and transferred to 0.45 µm polyvinylidene difluoride (PVDF) membrane. Membranes were blocked with 5% non-fat milk for 2 hat room temperature and incubated with primary antibody against VE-cadherin (1:1,000; cat. no. ab33168; Abcam), MYPT (1:1,000; cat. no. 2634; Cell Signaling Technology, Inc.), phosphorylated (p)-MYPT (1:1,000; cat. no. 3040; Cell Signaling Technology, Inc.) and GAPDH (1:3,000, cat. no. 2118; Cell Signaling Technology, Inc.) at 4˚C overnight. After washing with TBS-T (0.5% Tween), the membranes were incubated with secondary antibodies for 2 h at room temperature and then washed with TBS-T. The secondary antibody was HRP-linked goat anti-rabbit IgG antibody (1:8,000; cat. no. 7074; Cell Signaling Technology, Inc.). Following the manufacturer's instructions, specific binding was revealed by an ECL kit (Pierce; Thermo Fisher Scientific, Inc.). Densitometry analysis was performed with ImageJ software 1.48 (National Institute Health).

Immunofluorescence

HUVECs were grown into monolayer on fibronectin-coated glass chamber slides and were then treated with 10 µM CdCl2 for 12 h. Then, cells were washed with PBS and fixed with 4% paraformaldehyde for 15 min. After washing three times with PBS for 10 min, the cells were stained with a primary antibody against human VE-cadherin (cat. no. a33168; Abcam) at a dilution of 1:500 overnight at 4˚C and were incubated with the Alexa Fluor 546 donkey anti-rabbit secondary antibody (1:200; cat. no. A10040; Thermo Fisher Scientific, Inc.) for 2 h at room temperature. After washing three times with PBS for 10 min at room temperature, samples were imaged using an Olympus BX51 fluorescence microscope (Olympus Corporation) with an excitation wavelength of 546 nm (magnification, x200). The chamber slide with the monolayer was divided in 16 equal areas (4x4) and one field was randomly chosen in each area. The image mostly close to the average staining intensity was chosen as representative image.

Statistical data analysis

All the data were analyzed using GraphPad Prism 5.0 (GraphPad Software, Inc.). Data are presented as means ± SD. Statistical significance was assessed using one-way analysis of variance followed by Tukey's post hoc test or Student's t-test. A statistical difference of P<0.05 was considered significant.

Results

High dose Cd increases VE-Cadherin expression

The present study analyzed the mRNA and protein expression of VE-cadherin in HUVECs treated with different concentrations of Cd for 12 h. Fig. 1A showed that relative VE-cadherin mRNA expression was not changed at concentrations of 0.1, 1 and 5 µM of Cd. However, it was significantly upregulated by 10 and 50 µM Cd. Western blotting showed that Cd increases VE-cadherin protein expression following treatment with Cd at concentrations of 1, 5, 10 and 50 µM (Fig. 1B and C). A previous study showed that Cd disrupts VE-cadherin mediated cell-to-cell adhesion of HUVECs (25). Immunofluorescent staining with VE-cadherin antibody on HUVECs treated with Cd was performed. As shown in Fig. 1D, VE-cadherin is mainly distributed in cytoplasm membrane under normal condition. After treatment with 10 µM Cd, the fluorescence became stronger, suggesting that higher levels of VE-cadherin were detected at cell-to-cell junctions between cells.

Cd inhibits ROCK activity in HUVECs

To examine whether Cd affected ROCK activity, the phosphorylation levels of ROCK downstream substrate, myosin-binding subunit of myosin phosphatase (MYPT), were evaluated by western blotting. HUVECs treated with 10 µM Cd showed a significant decrease in p-MYPT after 12 h (Fig. 2A and B), suggesting that Cd inhibited the Rho/ROCK pathway.

ROCK activation reduces Cd-induced increase of VE-cadherin expression

Narciclasine is an activator of the Rho/ROCK pathway (26). The present study examined whether narciclasine inhibited Cd-induced VE-cadherin expression. As shown in Fig. 3A-C, 10 µM narciclasine reduced mRNA and protein level of VE-cadherin upon Cd treatment (P<0.01). With pretreatment of 10 µM narciclasine, 10 µM Cd did not increases VE-cadherin protein in HUVECs (Fig. 3D), suggesting Cd induced VE-cadherin expression through inhibition of ROCK signaling.

Inhibition of ROCK upregulates the expression of VE-cadherin

To examine the effect of ROCK pathway on VE-cadherin, HUVECs were treated with different concentrations of ROCK inhibitors Y27632 for 12 h. Y27632 increased the expression of VE-cadherin mRNA at 10 and 20 µM (P<0.05; Fig. 4A). Y27632 also increased the protein levels of VE-cadherin (P<0.05; Fig. 4B and C), suggesting that ROCK pathway negatively regulated VE-cadherin expression. With pretreatment of 10 µM Y27632, Cd did not significantly alter the level of VE-cadherin in HUVECs (Fig. 4D).

Discussion

Cd exposure has been reported to cause dysfunction of VECs (8,27). Depending on the dose of exposure, Cd differentially affects vascular VECs, including permeability, apoptosis and proliferation (7,28,29). The present study demonstrated that Cd upregulated expression of VE-cadherin via inhibition of ROCK activities.

The regulation of signaling pathways in response to Cd toxicity is dependent on Cd concentration (30). A previous study demonstrated that low-dose Cd (4 µM) impairs adherens junctions by inducing VE-cadherin and β-catenin redistribution, causing hyperpermeability in HUVEC monolayers (19,31). In the present study, Cd increased VE-cadherin expression in HUVECs in a concentration-dependent manner. The effect of Cd (10 µM) on VE-cadherin was the more remarked than other concentration. In a previous study, treatment of HUVECs with Cd reduces VE-cadherin localization to cell junctions in a concentration-dependent manner (32). Similarly, in the present study, lower levels of VE-cadherin were noted at cell-to-cell junctions between cells following 10 µM Cd treatment. However, Cd (10 µM) has been reported to induce VEC hyperpermeability, suggesting that the increased expression of VE-cadherin induced by 10 µM Cd fails to rescue vascular hyperpermeability (33). One reason may be that apoptosis and senescence of VECs during 10 µM Cd exposure leads to enhanced vessel wall permeability to cytokines, growth factors, lipids and immune cells (34).

The present study demonstrated that 10 µM of Cd inhibited ROCK activity. The ROCK pathway increases vascular permeability by causing junction protein remodeling and endothelial barrier dysfunction (35,36). ROCK inhibits the expression of tight junction components, including occludin and claudin-1 (37,38). The present study found that ROCK also negatively regulated the expression of VE-cadherin in HUVECs, In addition, Cd did not increase the expression of VE-cadherin in the presence of ROCK inhibitor Y27632, suggesting that ROCK mediated Cd-induced VE-cadherin expression. The results of the present study are consistent with previous studies. For example, the ROCK pathway inhibitor partially limits the increased monolayer permeability in lethal toxin-treated VECs through restoration of VE-cadherin expression and membrane localization (39). Inhibition of ROCK decreases the tension across VE-cadherin adhesion and VE-cadherin dissociation rate, resulting in accumulation of VE-cadherin in adherens junctions (40). FPND, a ROCK inhibitor, protects vascular integrity through cytoskeletal rearrangement and enhancement of cell-to-cell junctions in VECs via the ROCK1 and VE-cadherin signaling pathways (41).

In conclusion, the results of the present study suggested that ROCK inhibition contributes to Cd-induced expression of VE-cadherin in endothelial cells. It increases our understanding of Cd-induced vascular dysfunction.

Acknowledgements

The authors thank Dr Jing Liu of Shandong Provincial Qianfoshan Hospital (Jinan, China) for scientific discussion and technical support.

Funding

Funding: This study was supported by a grant from Science and Technology Development Plan of Shandong Province (grant no. 2013GSF11805).

Availability of data and materials

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

Authors' contributions

MG and HW conceived and designed the experiments. XiaoruiL, XiaoL and RS performed the experiments. XiaoruiL, XiaoL and RS analyzed the data. XiaoruiL, MG and HW wrote the paper. XiaoruiL and MG confirm the authenticity of all the raw data. All authors have read and approved the final 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 

Apykhtina OL, Dybkova SM, Sokurenko LM and Chaikovsky YB: Cytotoxic and genotoxic effects of cadmium sulfide nanoparticles. Exp Oncol. 40:194–199. 2018.PubMed/NCBI

2 

Surolia R, Karki S, Kim H, Yu Z, Kulkarni T, Mirov SB, Carter AB, Rowe SM, Matalon S, Thannickal VJ, et al: Heme oxygenase-1-mediated autophagy protects against pulmonary endothelial cell death and development of emphysema in cadmium-treated mice. Am J Physiol Lung Cell Mol Physiol. 309:L280–l292. 2015.PubMed/NCBI View Article : Google Scholar

3 

Angeli JK, Cruz Pereira CA, de Oliveira Faria T, Stefanon I, Padilha AS and Vassallo DV: Cadmium exposure induces vascular injury due to endothelial oxidative stress: The role of local angiotensin II and COX-2. Free Radic Biol Med. 65:838–848. 2013.PubMed/NCBI View Article : Google Scholar

4 

Chen H, Lu Y, Cao Z, Ma Q, Pi H, Fang Y, Yu Z, Hu H and Zhou Z: Cadmium induces NLRP3 inflammasome-dependent pyroptosis in vascular endothelial cells. Toxicol Lett. 246:7–16. 2016.PubMed/NCBI View Article : Google Scholar

5 

Chen X, Li J, Cheng Z, Xu Y, Wang X, Li X, Xu D, Kapron CM and Liu J: Low dose cadmium inhibits proliferation of human renal mesangial cells via activation of the JNK pathway. Int J Environ Res Public Health. 13(990)2016.PubMed/NCBI View Article : Google Scholar

6 

Liu F, Wang B, Li L, Dong F, Chen X, Li Y, Dong X, Wada Y, Kapron CM and Liu J: Low-dose cadmium upregulates VEGF expression in lung adenocarcinoma cells. Int J Environ Res Public Health. 12:10508–10521. 2015.PubMed/NCBI View Article : Google Scholar

7 

Prozialeck WC, Edwards JR and Woods JM: The vascular endothelium as a target of cadmium toxicity. Life Sci. 79:1493–1506. 2006.PubMed/NCBI View Article : Google Scholar

8 

Nagarajan S, Rajendran S, Saran U, Priya MK, Swaminathan A, Siamwala JH, Sinha S, Veeriah V, Sonar P, Jadhav V, et al: Nitric oxide protects endothelium from cadmium mediated leakiness. Cell Biol Int. 37:495–506. 2013.PubMed/NCBI View Article : Google Scholar

9 

Fujiwara Y, Yamamoto C, Yoshida E, Kumagai Y and Kaji T: Heparan sulfate chains potentiate cadmium cytotoxicity in cultured vascular endothelial cells. Arch Toxicol. 90:259–267. 2016.PubMed/NCBI View Article : Google Scholar

10 

Lampugnani MG, Resnati M, Raiteri M, Pigott R, Pisacane A, Houen G, Ruco LP and Dejana E: A novel endothelial-specific membrane protein is a marker of cell-cell contacts. J Cell Biol. 118:1511–1522. 1992.PubMed/NCBI View Article : Google Scholar

11 

Lagendijk AK and Hogan BM: VE-cadherin in vascular development: A coordinator of cell signaling and tissue morphogenesis. Curr Top Dev Biol. 112:325–352. 2015.PubMed/NCBI View Article : Google Scholar

12 

Dejana E and Vestweber D: The role of VE-cadherin in vascular morphogenesis and permeability control. Prog Mol Biol Transl Sci. 116:119–144. 2013.PubMed/NCBI View Article : Google Scholar

13 

Du L, Dong F, Guo L, Hou Y, Yi F, Liu J and Xu D: Interleukin-1β increases permeability and upregulates the expression of vascular endothelial-cadherin in human renal glomerular endothelial cells. Mol Med Rep. 11:3708–3714. 2015.PubMed/NCBI View Article : Google Scholar

14 

Li L, Chen X, Dong F, Liu Q, Zhang C, Xu D, Allen TD and Liu J: Dihydroartemisinin up-regulates VE-cadherin expression in human renal glomerular endothelial cells. J Cell Mol Med. 22:2028–2032. 2018.PubMed/NCBI View Article : Google Scholar

15 

Zhang C, Liu Q, Dong F, Li L, Du J, Xie Q, Hu H, Yan S, Zhou X, Li C, et al: Catalpol downregulates vascular endothelial-cadherin expression and induces vascular hyperpermeability. Mol Med Rep. 13:373–378. 2016.PubMed/NCBI View Article : Google Scholar

16 

Liu J, Dong F, Jeong J, Masuda T and Lobe CG: Constitutively active Notch1 signaling promotes endothelialmesenchymal transition in a conditional transgenic mouse model. Int J Mol Med. 34:669–676. 2014.PubMed/NCBI View Article : Google Scholar

17 

Gheorghescu A and Thompson J: Delayed vasculogenesis and impaired angiogenesis due to altered Ang-2 and VE-cadherin levels in the chick embryo model following exposure to cadmium. Pediatr Surg Int. 32:175–186. 2016.PubMed/NCBI View Article : Google Scholar

18 

Li L, Dong F, Xu D, Du L, Yan S, Hu H, Lobe CG, Yi F, Kapron CM and Liu J: Short-term, low-dose cadmium exposure induces hyperpermeability in human renal glomerular endothelial cells. J Appl Toxicol. 36:257–265. 2016.PubMed/NCBI View Article : Google Scholar

19 

Dong F, Guo F, Li L, Guo L, Hou Y, Hao E, Yan S, Allen TD and Liu J: Cadmium induces vascular permeability via activation of the p38 MAPK pathway. Biochem Biophys Res Commun. 450:447–452. 2014.PubMed/NCBI View Article : Google Scholar

20 

Etienne-Manneville S and Hall A: Rho GTPases in cell biology. Nature. 420:629–635. 2002.PubMed/NCBI View Article : Google Scholar

21 

Wang J, Liu X and Zhong Y: Rho/Rho-associated kinase pathway in glaucoma (Review). Int J Oncol. 43:1357–1367. 2013.PubMed/NCBI View Article : Google Scholar

22 

Huang Y, Tan Q, Chen R, Cao B and Li W: Sevoflurane prevents lipopolysaccharide-induced barrier dysfunction in human lung microvascular endothelial cells: Rho-mediated alterations of VE-cadherin. Biochem Biophys Res Commun. 468:119–124. 2015.PubMed/NCBI View Article : Google Scholar

23 

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.PubMed/NCBI View Article : Google Scholar

24 

Guo L, Dong F, Hou Y, Cai W, Zhou X, Huang AL, Yang M, Allen TD and Liu J: Dihydroartemisinin inhibits vascular endothelial growth factor-induced endothelial cell migration by a p38 mitogen-activated protein kinase-independent pathway. Exp Ther Med. 8:1707–1712. 2014.PubMed/NCBI View Article : Google Scholar

25 

Prozialeck WC: Evidence that E-cadherin may be a target for cadmium toxicity in epithelial cells. Toxicol Appl Pharmacol. 164:231–249. 2000.PubMed/NCBI View Article : Google Scholar

26 

Lefranc F, Sauvage S, Van Goietsenoven G, Mégalizzi V, Lamoral-Theys D, Debeir O, Spiegl-Kreinecker S, Berger W, Mathieu V, Decaestecker C and Kiss R: Narciclasine, a plant growth modulator, activates Rho and stress fibers in glioblastoma cells. Mol Cancer Ther. 8:1739–1750. 2009.PubMed/NCBI View Article : Google Scholar

27 

Wang X, Dong F, Wang F, Yan S, Chen X, Tozawa H, Ushijima T, Kapron CM, Wada Y and Liu J: Low dose cadmium upregulates the expression of von Willebrand factor in endothelial cells. Toxicol Lett. 290:46–54. 2018.PubMed/NCBI View Article : Google Scholar

28 

Wei T, Jia J, Wada Y, Kapron CM and Liu J: Dose dependent effects of cadmium on tumor angiogenesis. Oncotarget. 8:44944–44959. 2017.PubMed/NCBI View Article : Google Scholar

29 

Chen X, Li L, Liu F, Hoh J, Kapron CM and Liu J: Cadmium induces glomerular endothelial cell-specific expression of complement factor H via the -1635 AP-1 binding site. J Immunol. 202:1210–1218. 2019.PubMed/NCBI View Article : Google Scholar

30 

Messner B, Turkcan A, Ploner C, Laufer G and Bernhard D: Cadmium overkill: Autophagy, apoptosis and necrosis signalling in endothelial cells exposed to cadmium. Cell Mol Life Sci. 73:1699–1713. 2016.PubMed/NCBI View Article : Google Scholar

31 

Zhang H, Li L, Wang Y, Dong F, Chen X, Liu F, Xu D, Yi F, Kapron CM and Liu J: NF-ĸB signaling maintains the survival of cadmium-exposed human renal glomerular endothelial cells. Int J Mol Med. 38:417–422. 2016.PubMed/NCBI View Article : Google Scholar

32 

Woods JM, Leone M, Klosowska K, Lamar PC, Shaknovsky TJ and Prozialeck WC: Direct antiangiogenic actions of cadmium on human vascular endothelial cells. Toxicol In Vitro. 22:643–651. 2008.PubMed/NCBI View Article : Google Scholar

33 

Wolf MB and Baynes JW: Cadmium and mercury cause an oxidative stress-induced endothelial dysfunction. Biometals. 20:73–81. 2007.PubMed/NCBI View Article : Google Scholar

34 

Dong Z, Wang L, Xu J, Li Y, Zhang Y, Zhang S and Miao J: Promotion of autophagy and inhibition of apoptosis by low concentrations of cadmium in vascular endothelial cells. Toxicol In Vitro. 23:105–110. 2009.PubMed/NCBI View Article : Google Scholar

35 

Eisa-Beygi S and Wen XY: Could pharmacological curtailment of the RhoA/Rho-kinase pathway reverse the endothelial barrier dysfunction associated with Ebola virus infection? Antiviral Res. 114:53–56. 2015.PubMed/NCBI View Article : Google Scholar

36 

Liu J, Wada Y, Katsura M, Tozawa H, Erwin N, Kapron CM, Bao G and Liu J: Rho-associated coiled-coil kinase (ROCK) in molecular regulation of angiogenesis. Theranostics. 8:6053–6069. 2018.PubMed/NCBI View Article : Google Scholar

37 

Grothaus JS, Ares G, Yuan C, Wood DR and Hunter CJ: Rho kinase inhibition maintains intestinal and vascular barrier function by upregulation of occludin in experimental necrotizing enterocolitis. Am J Physiol Gastrointest Liver Physiol. 315:G514–G528. 2018.PubMed/NCBI View Article : Google Scholar

38 

Yang M, Chen XM, Du XG, Cao FF, Vijaya Luxmi S and Shen Q: Continuous blood purification ameliorates endothelial hyperpermeability in SAP patients with MODS by regulating tight junction proteins via ROCK. Int J Artif Organs. 36:700–709. 2013.PubMed/NCBI View Article : Google Scholar

39 

Warfel JM and D'Agnillo F: Anthrax lethal toxin-mediated disruption of endothelial VE-cadherin is attenuated by inhibition of the Rho-associated kinase pathway. Toxins (Basel). 3:1278–1293. 2011.PubMed/NCBI View Article : Google Scholar

40 

Daneshjou N, Sieracki N, van Nieuw Amerongen GP, Conway DE, Schwartz MA, Komarova YA and Malik AB: Rac1 functions as a reversible tension modulator to stabilize VE-cadherin trans-interaction. J Cell Biol. 208:23–32. 2015.PubMed/NCBI View Article : Google Scholar

41 

Li S, Ai N, Shen M, Dang Y, Chong CM, Pan P, Kwan YW, Chan SW, Leung GPH, Hoi MPM, et al: Discovery of a ROCK inhibitor, FPND, which prevents cerebral hemorrhage through maintaining vascular integrity by interference with VE-cadherin. Cell Death Discov. 3(17051)2017.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

May-2022
Volume 23 Issue 5

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
Li X, Li X, Sun R, Gao M and Wang H: Cadmium exposure enhances VE‑cadherin expression in endothelial cells via suppression of ROCK signaling. Exp Ther Med 23: 355, 2022
APA
Li, X., Li, X., Sun, R., Gao, M., & Wang, H. (2022). Cadmium exposure enhances VE‑cadherin expression in endothelial cells via suppression of ROCK signaling. Experimental and Therapeutic Medicine, 23, 355. https://doi.org/10.3892/etm.2022.11282
MLA
Li, X., Li, X., Sun, R., Gao, M., Wang, H."Cadmium exposure enhances VE‑cadherin expression in endothelial cells via suppression of ROCK signaling". Experimental and Therapeutic Medicine 23.5 (2022): 355.
Chicago
Li, X., Li, X., Sun, R., Gao, M., Wang, H."Cadmium exposure enhances VE‑cadherin expression in endothelial cells via suppression of ROCK signaling". Experimental and Therapeutic Medicine 23, no. 5 (2022): 355. https://doi.org/10.3892/etm.2022.11282