Open Access

Curcumin suppresses intestinal microvascular endothelial cells invasion and angiogenesis induced by activated platelets

  • Authors:
    • Su Xu
    • Zhao‑Xiu Xu
    • Shuai Yan
    • Jin Le
    • Hao Chen
    • Lan Ming
    • Shu‑Guang Xu
    • Tao Lin
  • View Affiliations

  • Published online on: June 11, 2019     https://doi.org/10.3892/etm.2019.7662
  • Pages: 1099-1106
  • Copyright: © Xu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The present study investigated the effects and mechanism by which curcumin suppresses intestinal microvascular endothelial cells (INMECs) invasion and angiogenesis induced by activated platelets. INMECs were obtained from healthy rats, and divided into five groups: Control, platelets, platelets +2.5 µM curcumin, platelets +5.0 µM curcumin and platelets +10.0 µM curcumin. Curcumin toxicity was determined and vascular endothelial growth factor (VEGF) concentrations of the five groups were measured using ELISA. The branch point numbers were measured using a capillary tube formation experiment, invasion cell numbers were evaluated with the Transwell assay, relative protein expression levels were measured with western blot assay and immunofluorescence staining of the nucleus. The 2.5, 5 and 10 µM curcumin concentrations were found to be suitable for INMECs. Curcumin significantly downregulated VEGF concentration, suppressed vascular lumen formation and inhibited invasion cell numbers in a dose‑dependent manner. The α‑smooth muscle actin, collagen I, E‑cadherin, phosphorylated (p‑) phosphoinositide 3‑kinase (PI3K), p‑protein kinase B (AKT), p‑mammalian target of rapamycin (m‑TOR) and hypoxia inducible factor subunit alpha (HIF‑1α) protein expression levels of the curcumin‑treated groups were significantly downregulated in a dose‑dependent manner compared with the platelet group. HIF‑1α protein expression levels in the nucleus of the curcumin‑treated groups were significantly suppressed in a dose‑dependent manner compared with the platelet group. In conclusion, curcumin suppressed INMEC invasion and angiogenesis induced by activated platelets via inhibiting the activation of the PI3K/AKT/mTOR pathway.

Introduction

Crohn's Disease (CD) is a chronic recurrent inflammatory disease affecting the digestive tract. The aetiology and pathogenesis of CD remains not fully understood. However, the consensus is that intestinal mucosal immune regulation, lymphangiogenesis and intestinal mucosal barrier damage are involved in the development of CD (1,2). A crucial area of current research has been the identification of effective drug components for CD treatment. Curcumin, one of the most popular traditional medicines, is a natural phenolic substance extracted from the rhizome of Curcuma longa. It has widely been used as a traditional herb in China and Southeast Asian countries for thousands of years. Recent studies have confirmed that curcumin possesses several pharmacological properties including antioxidant, anti-inflammatory and antitumour abilities; it also promotes wound healing, is spasmodic, serves as an anticoagulant and provides liver protection (35). Several clinical and animal experiments have reported that curcumin has beneficial therapeutic effects on inflammatory bowel disease with few adverse effects. Therefore, curcumin possesses substantial potential for clinical application (69); however the effects and mechanism of curcumin in CD treatment remain unclear. The present study determined suitable curcumin concentrations for further experimentation in cells using MTT assay. Then the effects of curcumin on suppressing angiogenesis and cell invasion were determined. Finally, the mechanisms by which curcumin can be used to treat CD were investigated by measuring relative protein expression levels using western blot analysis and immunofluorescence staining.

Materials and methods

Experimental materials

A total of 12 Male Wistar rats of specific pathogen-free grade (weight, 300–350 g; 8.0±0.5 week age) were purchased from Zhejiang Experimental Animal Center (no. 170525003; animal licence no. SCXK 2014-0001) and supplied by the Experimental Animals Center of Nanjing University of Chinese Medicine (Nanjing, China). The animals were housed in controlled conditions (temperature, 23±2°C; humidity, 45–60%; 12-h light/dark cycle; free access to water and a standard diet). Adenosine diphosphate (ADP) was purchased from Sigma-Aldrich (Merck KGaA, Darmstadt, Germany). Foetal bovine serum and RPM 11650 culture medium were purchased from Gibco (Thermo Fisher Scientific, Inc., Waltham, MA, USA). Curcumin, activated platelets and MTT kit were purchased from Sigma-Aldrich (Merck KGaA). Total protein and Nuclear Protein Extraction kits were purchased from Sigma-Aldrich. The vascular endothelial growth factor (VEGF) ELISA kit (cat. no. 20170320; Wuhan Huamei Bioengineering Co., Ltd., Wuhan, China), anti-α-smooth muscle actin (SMA; cat. no. 19245; Cell Signaling Technology, Inc., Danvers, MA, USA), anti-collagen I (cat. no. ab90395; Abcam, Cambridge, UK), anti-E-cadherin (cat. no. 3195; Cell Signaling Technology, Inc.), anti-VEGF (cat. no. 13689-1; Wuhan Sanying Biotechnology, Wuhan, China), anti-phosphoinositide 3-kinase (PI3K; cat. no. 21890-1; Wuhan Sanying Biotechnology), anti-phosphorylated (p-) PI3K (cat. no. 4228; Cell Signaling Technology, Inc.), anti-protein kinase B (AKT; cat. no. 60203-2; Wuhan Sanying Biotechnology), anti-p-AKT (cat. no. 4060; Cell Signaling Technology, Inc.), anti-mammalian target of rapamycin (mTOR; cat. no. ab32028; Abcam), anti-p-mTOR (cat. no. ab84400; Abcam), anti-hypoxia inducible factor subunit α (HIF-1α; cat. no. ab51608; Abcam), anti-laminin subunit beta 1 (LAMB1; cat. no. 109293; Abcam) and anti-GAPDH (cat. no. 10494-1; Wuhan Sanying Biotechnology) were purchased from the respective suppliers. This study was approved by the Ethics Committee of Nanjing University of Chinese Medicine (approval no. 2017011001).

INMEC isolation and culture

INMECs were isolated following the methods described in a previous review (10). INMECs, at fourth or fifth passage, were cultured in the RPM 11650 culture medium containing 10% FBS and used for the following experiments.

MTT assay

Cells in the logarithmic growth phase were inoculated in a 96-well plate and routinely cultured until cell adherence was observed. Curcumin solution at concentrations of 0, 0.625, 1.25, 2.5, 5, 10, 20 and 40 µM were added to the samples for 4 h. Then the supernatant was removed and 200 µl dimethyl sulfoxide was added to each well. The samples were placed on a rocking bed at low-speed oscillation for 1 min. The absorbance values were measured at 490 nm wavelength and the cell viability of the different groups was measured. The experiment was repeated three times.

Cell grouping

According to the MTT assay result, INMECs were randomly divided into the following groups: Control, platelets and three different concentrations of curcumin-treated groups (2.5, 5 and 10 µM). The INMECs were cultured at a density of 1×104 cells/well for 4 h. The supernantants of the different groups were collected following centrifugation to measure VEGF concentrations then cells were collected for further experiments. The experiment was repeated three times.

ELISA

INMECs were treated with 25 µM ADP for 24 h at room temperature, followed by the curcumin treatment of various concentrations. Culture medium was collected and centrifuged at 10,000 × g for 5 min at 4°C. The VEGF concentrations of the groups were measured with an ELISA kit at 490 nm according to the supplier's instructions. Each group was assigned nine wells.

Capillary tube formation experiment

Matrigel matrix glue was added to a 48-well culture plate at 37°C for 30 min. INMECs were added to wells at a density of 1×105 cells/well. Normal serum, normal serum containing activated platelets and three different curcumin concentrations (2.5, 5 and 10 µM) + normal serum containing activated platelets were added to wells and cultured for 12 h. An inverted microscope was used to observe and photograph capillary tube formation, and Image J v.6.0 software (National Institutes of Health, Bethesda, MD, USA) was used to quantitatively analyse cavity formation length. Each group included three replicates. Five fields were selected for each well, and the experiment was repeated three times.

Transwell invasion assay

Transwell chambers in a 24-well plate were used for the Transwell invasion assay. For each sample, the surface of the membrane was evenly spread with Dulbecco's modified Eagle's medium (DMEM)-diluted Matrigel containing a diluted growth factor (Matrigel:DMEM, 1:4, v/v) and maintained in an incubator (37°C, 5% CO2) for 30 min prior to cell seeding. Then 100 µl of INMEC suspension (1×105/ml) in the logarithmic growth phase was inoculated into the upper chamber and 600 µl of serum free-DMEM was added. Each group was assigned three wells. Normal culture medium, activated platelets, activated platelets +2.5 µM curcumin, activated platelets +5 µM curcumin and activated platelets +10 µM curcumin were added to the wells and cultured for 24 h. Cells were stained with 0.1% crystal violet (Beyotime Institute of Biotechnology) at room temperature for 5 min, The chamber was then removed, medium discarded and samples were washed twice with PBS. Tweezers were used to place the filter membrane onto a slide plate, and five fields were randomly selected under an inverted light microscope to observe and count the number of migratory cells (magnification, ×100).

Western blot analysis

INMECs were collected from the groups and the total proteins or nuclear proteins were extracted using a total protein extraction kit or nucleus protein kit respectively. The protein concentrations were quantified by bicinchoninic acid assay. In brief, 50 µg protein samples were loaded/lane to perform electrophoresis with 12% SDS-PAGE then transferred to polyvinylidene difluoride membranes. Following blocking of the membranes with 5% skimmed milk at 37°C for 1 h, the membranes were incubated with primary antibodies (α-SMA, 1:1,000; collagen I, 1:1,000; E-cadherin, 1:2,000; VEGFR, 1:1,000; PI3K, 1:1,000; p-PI3K, 1:1,000; AKT, 1:1,000; p-AKT, 1:1,000; mTOR, 1:1,000; p-mTOR, 1:1,000; HIF-1α, 1:1,000; LamB1, 1:2,000 and GAPDH, 1:500). The samples were incubated overnight at 4°C and washed thrice with PBS. Membranes were incubated with HRP-marked second antibody (1:2,000; cat. no. ab205718; Abcam, Cambridge, UK) at room temperature for 1 h. Enhanced chemiluminescence reagent was used to visualise protein bands and band intensity were analysed using Image J v6.0 software (National Institutes of Health). GAPDH was used as a reference in this experiment.

Immunofluorescence staining

INMECs were inoculated into 48-well plates, and the corresponding treatments were added for 12 h. Then cells were washed with PBS, fixed with 2% polyoxymethylene for 30 min and washed again with PBS (30 sec; three times) at room temperature. The cells were permeated with 1% Triton X-100 then 5% bovine serum albumin was used to block samples at room temperature for 1 h. HIF-1α antibody (1:1,000; cat. no. ab51608; Abcam) was added and samples were incubated overnight at 4°C then washed with PBS. Fluorescein-conjugated secondary antibody (1:1,000; cat. no. ab205718) was added to samples at room temperature for 2 h then DAPI was used to stain the nuclei and buffered glycerol was added to mount samples. HIF-1α staining was observed under a laser confocal microscope (magnification, ×100) following 1 h.

Statistical analysis

Data were presented as mean ± standard deviation. SPSS 22.0 software (IBM Corp., Armonk, NY, USA) was used for data analysis. A one-way analysis of variance with a post hoc Dunnett's t-test was used to assess differences amongst the groups. P<0.05 was considered to indicate statistical significance.

Results

Cell viability following treatment with different curcumin concentrations

There were no significant differences in cell viability amongst the 0, 0.625, 1.25, 2.5, 5 and 10 µM curcumin concentration groups (P>0.05; Fig. 1). However, cell viability of the 20 and 40 µM curcumin concentration groups significantly decreased compared with the 0 µM curcumin concentration group (P<0.001; Fig. 1). The results demonstrated that the 0, 0.625, 1.25, 2.5, 5 and 10 µM curcumin concentrations were suitable for INMECs in this in vitro study.

VEGF concentration decreases with increasing curcumin concentration

Compared with the control group, the VEGF concentration of the platelets group was significantly upregulated (P<0.001; Fig. 2), indicating that activated platelets stimulated INMECs to secrete VEGF. With curcumin supplementation, the VEGF concentrations of the different curcumin-treated groups were significantly suppressed compared with the platelets group (P<0.05; Fig. 2). Furthermore, there was a significant difference in terms of VEGF concentration amongst the different curcumin concentration-treated groups (Fig. 2).

Branch point numbers decrease with increasing curcumin concentrations

There was a significantly higher branch point number in the platelets group compared with the control group (P<0.05; Fig. 3), suggesting that activated platelets induced angiogenesis in INMECs. However, there were significantly fewer branch points in the curcumin-treated groups than in the platelets group (P<0.05; Fig. 3) with curcumin concentration exhibiting a dose-dependent association (P<0.05; Fig. 3).

Cell invasion decreases with increasing curcumin concentration

Compared with the platelets group, the 5 µM (P<0.05; Fig. 4) and 10 µM (P<0.01; Fig. 4) curcumin-treated groups had significantly decreased cell invasion. These results suggested that 5 and 10 µM curcumin concentrations suppressed the number of migrated INMECs in vitro.

α-SMA, collagen I, E-cadherin and VEGFR protein expression levels following curcumin treatment

Compared with the control group, the platelets group exhibited significantly upregulated α-SMA, collagen I and E-cadherin protein expression levels (P<0.05; Fig. 5). However, α-SMA, collagen I and E-cadherin protein expression levels were significantly suppressed in a dose-dependent manner in the curcumin-treated groups compared with the platelets group (P<0.05; Fig. 5). There were no significant differences in VEGFR protein levels amongst the different groups (P>0.05; Fig. 5).

PI3K/AKT/mTOR pathway relative to HIF-1α protein expression levels following curcumin treatment

There were no significant differences in PI3K, AKT and mTOR protein expression levels amongst the five groups (P>0.05; Fig. 6). The p-PI3K, p-AKT, p-mTOR and HIF-1α protein expression levels were significantly suppressed in the curcumin-treated groups, in a dose-dependent manner, compared with the platelets group (P<0.05; Fig. 6). These results suggested that curcumin affects HIF-1α by regulating phosphorylation of the PI3K/AKT/mTOR pathway.

HIF-1α and LamB1 protein expression levels in the nucleus following curcumin treatment

Compared with the control group, the platelets group significantly upregulated HIF-1α protein expression levels in the nucleus (P<0.05; Fig. 7). Curcumin treatment significantly suppressed HIF-1α protein expression levels in the 5.0 and 10.0 µM curcumin-treated groups (P<0.05; Fig. 7). In addition, there was a significant difference between the 5 and 10 µM curcumin-treated groups (P<0.05; Fig. 7).

HIF-1α protein nuclear translocation

The immunofluorescence staining results determined that HIF-1α protein expression levels increased following stimulation with activated platelets (Fig. 8). By contrast, curcumin suppressed HIF-1α protein expression levels and decreased HIF-1α protein nuclear translocation in vitro (Fig. 8).

Discussion

The most commonly used drugs for the treatment of CD include aminosalicylic acid, glucocorticoids, immunosuppressive agents, inflammatory transmitter inhibitors, targeted biological immunotherapy and antibiotics (11). In the treatment of CD, traditional Chinese medicine has been proven to be multi-channel medicines and can be used for long-term treatment of patients, with fewer adverse events (12,13). During CD development, INMECs produce blood vessels induced by activated platelets (1416). Curcumin is a traditional natural drug extract that exhibits anti-inflammatory, antitumour and immunoregulation properties (1719). However, the treatment effects of curcumin on CD remain unclear. Firstly, the present study evaluated the safety of curcumin on INMECs. MTT assay results demonstrated that curcumin concentrations below 20 µM did not cause significant toxicity. The effects and mechanisms of curcumin on angiogenesis and the invasion of INMECs induced by activated platelet stimulation were then investigated. The results demonstrated that VEGF concentration was significantly reduced, branch point number was decreased and number of migrated cells were decreased with curcumin treatment in a dose-dependent manner. Finally, the relative protein expression levels were investigated to determine the mechanism of action.

Previous studies demonstrated that α-SMA, collagen I, E-cadherin and VEGFR have key roles in cell migration (2024). The present study demonstrated that curcumin suppressed INMEC angiogenesis and invasion by regulating α-SMA, collagen I and E-cadherin protein expression levels. By contrast, curcumin did not have significant effects on VEGFR protein expression levels. α-SMA, collagen I and E-cadherin genes are downstream of the HIF-1α signalling pathway, therefore the upstream signalling pathway was investigated to determine the effects of curcumin in vitro.

PI3K/AKT/mTOR/HIF-1α pathway stimulation is a crucial component in cell angiogenesis and migration, and can induce α-SMA, collagen I and E-cadherin protein activation (2530). The present study identified that curcumin affected phosphorylation of the PI3K/AKT/mTOR pathway; when suppressed, HIF-1α total and nuclear protein expression decreased. HIF-1α nuclear translocation is an essential part of the association between the PI3K/AKT/mTOR pathway and downstream genes (α-SMA, collagen I and E-cadherin). HIF-1α nuclear translocation was observed by immunofluorescence staining with the results demonstrating that HIF-1α nuclear translocation was inhibited by curcumin supplementation. In conclusion, curcumin was identified to have a dose-dependent inhibitory effect on INMEC angiogenesis and invasion induced by activated platelets, likely via inhibiting PI3K/AKT/mTOR pathway activation.

Acknowledgements

Not applicable.

Funding

The present study was supported by the National Natural Science Foundation of China (grant no. 81603622) and the Natural Science Foundation of Jiangsu Province of China (grant no. BK20161319).

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

SX was responsible for drafting the manuscript, as well as the acquisition, analysis and interpretation of data. ZXX collected data. SY interpreted the data. JL was responsible for drafting the manuscript. HC contributed to the study conception. LM, XGS and LT designed and supervised the current study. All authors read and approved the final manuscript.

Ethics approval and consent to participate

This study was approved by the Ethics committee of Nanjing University of Chinese Medicine (approval no. 2017011001).

Competing interests

The authors declare that they have no competing interests.

References

1 

Baumgart DC and Sandborn WJ: Cronhn's disease. Lancet. 380:1590–1605. 2012. View Article : Google Scholar : PubMed/NCBI

2 

Liu ZJ, Yadav PK, Su JL, Wang JS and Fei K: Potential role of Th17 cells in the pathogenesis of inflammatory bowel disease. World J Gastroenterol. 15:5784–5788. 2009. View Article : Google Scholar : PubMed/NCBI

3 

Ramsewak RS, DeWitt DL and Nair MG: Cytotoxicity, antioxidant and anti-inflammatory activities of curcumins I–III from Curcuma longa. Phytomedicine. 7:303–308. 2000. View Article : Google Scholar : PubMed/NCBI

4 

Aggarwal BB and Harikumar KB: Potential therapeutic effects of curcumin, the anti-inflammatory agent, against neurodegenerative, cardiavascular, pulmonary, metabolic, autoimmune and neoplastic disease. Int J Biochem Cell Biol. 41:40–59. 2009. View Article : Google Scholar : PubMed/NCBI

5 

Yadav VR, Suresh S, Devi K and Yadav S: Effect of cyclodextrin complexation of curcumin on its solubility and antiangiogenic and anti-inflammatory activity in rat colitis model. AAPS PharmSciTech. 10:752–762. 2009. View Article : Google Scholar : PubMed/NCBI

6 

Salh B, Assi K, Templeman V, Parhar K, Owen D, Gómez-Muñoz A and Jacobson K: Curcumin attenuates DNB-induced murine colitis. Am J Physiol Gastrointest Liver Physiol. 285:G235–G243. 2003. View Article : Google Scholar : PubMed/NCBI

7 

Venkataranganna MV, Rafiq M, Gopumadhavan S, Peer G, Babu UV and Mitra SK: NCB-02 (standardized Curcumin preparation) protects dinitrochlorobenzene-induced colitis through down-regulation of NFkappa-B and iNOS. World J Gastroenterol. 13:1103–1107. 2007. View Article : Google Scholar : PubMed/NCBI

8 

Hanai H, Iida T, Takeuchi K, Watanabe F, Maruyama Y, Andoh A, Tsujikawa T, Fujiyama Y, Mitsuyama K, Sata M, et al: Curcumin maintenance therapy for ulcerative colitis: Randomized, multicenter, double-blind, placebo-controlled trial. Clin Gastroenterol Hepatol. 4:1502–1506. 2006. View Article : Google Scholar : PubMed/NCBI

9 

Holt PR, Katz S and Kirshoff R: Curcumin therapy in inflammatory bowel disease: A pilot study. Dig Dis Sci. 50:2191–2193. 2005. View Article : Google Scholar : PubMed/NCBI

10 

Kohn EA, Du Z, Sato M, Van Schyndle CM, Welsh MA, Yang YA, Stuelten CH, Tang B, Ju W, Bottinger EP and Wakefield LM: A novel approach for the generation of genetically modified mammary epithelial cell cultures yields new insights into TGFβ signaling in the mammary gland. Breast Cancer Res. 12:R832010. View Article : Google Scholar : PubMed/NCBI

11 

Gomollón F: Developments in the treatment of inflammatory bowel disease: 2014 overview. Gastroenterol Hepatol. 37 (Suppl 3):S14–S21. 2014.(In Spanish). View Article : Google Scholar

12 

Ng SC, Lam YT, Tsoi KK, Chan FK, Sung JJ and Wu JC: Systematic review: The efficacy of herbal therapy in inflammatory bowel disease. Aliment Pharmacol Ther. 38:854–863. 2013. View Article : Google Scholar : PubMed/NCBI

13 

Rahimi R, Nikfar S and Abdollahi M: Induction of clinical response and remission of inflammatory bowel disease by use of herbal medicines: A meta-analysis. World J Gastroenterol. 19:5738–5749. 2013. View Article : Google Scholar : PubMed/NCBI

14 

Leonetti D, Reimund JM, Tesse A, Viennot S, Martinez MC, Bretagne AL and Andriantsitohaina R: Circulating microparticles from Crohn's disease patients cause endothelial and vascular dysfunctions. PLoS One. 8:e730882013. View Article : Google Scholar : PubMed/NCBI

15 

Danese S, Katz JA, Saibeni S, Papa A, Gasbarrini A, Vecchi M and Fiocchi C: Activated platelets are the source of elevated levels of soluble CD40 ligand in the circulation of inflammatory bowel disease patients. Gut. 52:1435–1441. 2003. View Article : Google Scholar : PubMed/NCBI

16 

Scaldaferri F, Lancellotti S, Pizzoferrato M and De Cristofaro R: Haemostatic system in inflammatory bowel diseases: New players in gut inflammation. World J Gastroenterol. 17:594–608. 2011. View Article : Google Scholar : PubMed/NCBI

17 

Reddy PH, Manczak M, Yin X, Grady MC, Mitchell A, Tonk S, Kuruva CS, Bhatti JS, Kandimalla R, Vijayan M, et al: Protective effects of indian spice curcumin against amyloid-β in Alzheimer's disease. J Alzheimers Dis. 61:843–866. 2018. View Article : Google Scholar : PubMed/NCBI

18 

Singh N, Sachdev A and Gopinath P: Polysaccharide functionalized single walled carbon nanotubes as nanocarriers for delivery of curcumin in lung cancer cells. J Nanosci Nanotechnol. 18:1534–1541. 2018. View Article : Google Scholar : PubMed/NCBI

19 

Masuelli L, Granato M, Benvenuto M, Mattera R, Bernardini R, Mattei M, d'Amati G, D'Orazi G, Faggioni A, Bei R and Cirone M: Chloroquine supplementation increases the cytotoxic effect of curcumin against Her2/neu overexpressing breast cancer cells in vitro and in vivo in nude mice while counteracts it in immune competent mice. Oncoimmunology. 6:e13561512017. View Article : Google Scholar : PubMed/NCBI

20 

Wu KH, Ho CT, Chen ZF, Chen LC, Whang-Peng J, Lin TN and Ho YS: The apple polyphenol phloretin inhibits breast cancer cell migration and proliferation via inhibition of signals by type 2 glucose transporter. J Food Drug Anal. 26:221–23. 2018. View Article : Google Scholar : PubMed/NCBI

21 

Zhang Z, Wang Y, Zhang J, Zhong J and Yang R: COL1A1 promotes metastasis in colorectal cancer by regulating the WNT/PCP pathway. Mol Med Rep. 17:5037–5042. 2018.PubMed/NCBI

22 

Guo H, Zhang X, Chen Q, Bao Y, Dong C and Wang X: miR-132 suppresses the migration and invasion of lung cancer cells by blocking USP9X-induced epithelial-mesenchymal transition. Am J Transl Res. 10:224–234. 2018.PubMed/NCBI

23 

Ogunbolude Y, Dai C, Bagu ET, Goel RK, Miah S, MacAusland-Berg J, Ng CY, Chibbar R, Napper S, Raptis L, et al: FRK inhibits breast cancer cell migration and invasion by suppressing epithelial-mesenchymal transition. Oncotarget. 8:113034–113065. 2017. View Article : Google Scholar : PubMed/NCBI

24 

Wang L, Tong D, Guo Q, Wang X, Wu F, Li Q, Yang J, Zhao L, Qin Y, Liu Y and Huang C: HOXD3 targeted by miR-203a suppresses cell metastasis and angiogenesis through VEGFR in human hepatocellular carcinoma cells. Sci Rep. 8:24312018. View Article : Google Scholar : PubMed/NCBI

25 

Choi YH, Jin GY, Li LC and Yan GH: Inhibition of protein kinase C delta attenuates allergic airway inflammation through suppression of PI3K/Akt/mTOR/HIF-1 alpha/VEGF pathway. PLoS One. 8:e817732013. View Article : Google Scholar : PubMed/NCBI

26 

Zhu Y, Tan J, Xie H, Wang J, Meng X and Wang R: HIF-1α regulates EMT via the Snail and β-catenin pathways in paraquat poisoning-induced early pulmonary fibrosis. J Cell Mol Med. 20:688–697. 2016. View Article : Google Scholar : PubMed/NCBI

27 

Zhang J, Zhu L, Fang J, Ge Z and Li X: LRG1 modulates epithelial-mesenchymal transition and angiogenesis in colorectal cancer via HIF-1α activation. J Exp Clin Cancer Res. 35:292016. View Article : Google Scholar : PubMed/NCBI

28 

Baumann B, Hayashida T, Liang X and Schnaper HW: Hypoxia-inducible factor-1α promotes glomerulosclerosis and regulates COL1A2 expression through interactions with Smad3. Kidney Int. 90:797–808. 2016. View Article : Google Scholar : PubMed/NCBI

29 

Watanabe T, Yasue A and Tanaka E: Hypoxia-inducible factor-1α is required for transforming growth factor-β1-induced type I collagen, periostin and α-smooth muscle actin expression in human periodontal ligament cells. Arch Oral Biol. 59:595–600. 2014. View Article : Google Scholar : PubMed/NCBI

30 

Yang N, Liang Y, Yang P and Ji F: Propofol suppresses LPS-induced nuclear accumulation of HIF-1α and tumor aggressiveness in non-small cell lung cancer. Oncol Rep. 37:2611–2619. 2017. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2019
Volume 18 Issue 2

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
Xu S, Xu ZX, Yan S, Le J, Chen H, Ming L, Xu SG and Lin T: Curcumin suppresses intestinal microvascular endothelial cells invasion and angiogenesis induced by activated platelets. Exp Ther Med 18: 1099-1106, 2019
APA
Xu, S., Xu, Z., Yan, S., Le, J., Chen, H., Ming, L. ... Lin, T. (2019). Curcumin suppresses intestinal microvascular endothelial cells invasion and angiogenesis induced by activated platelets. Experimental and Therapeutic Medicine, 18, 1099-1106. https://doi.org/10.3892/etm.2019.7662
MLA
Xu, S., Xu, Z., Yan, S., Le, J., Chen, H., Ming, L., Xu, S., Lin, T."Curcumin suppresses intestinal microvascular endothelial cells invasion and angiogenesis induced by activated platelets". Experimental and Therapeutic Medicine 18.2 (2019): 1099-1106.
Chicago
Xu, S., Xu, Z., Yan, S., Le, J., Chen, H., Ming, L., Xu, S., Lin, T."Curcumin suppresses intestinal microvascular endothelial cells invasion and angiogenesis induced by activated platelets". Experimental and Therapeutic Medicine 18, no. 2 (2019): 1099-1106. https://doi.org/10.3892/etm.2019.7662